351
|
Pang L, Ernst M, Huynh J. Development of a Multiplex Immunohistochemistry Workflow to Investigate the Immune Microenvironment in Mouse Models of Inflammatory Bowel Disease and Colon Cancer. Int J Mol Sci 2021; 22:ijms222011001. [PMID: 34681666 PMCID: PMC8539370 DOI: 10.3390/ijms222011001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022] Open
Abstract
Multiplex immunohistochemistry (mIHC) enables simultaneous staining of multiple immune markers on a single tissue section. Mounting studies have demonstrated the versatility of mIHC in evaluating immune infiltrates in different diseases and the tumour microenvironment (TME). However, the majority of published studies are limited to the analysis of human patient samples. Performing mIHC on formalin-fixed paraffin-embedded (FFPE) mouse tissues, particularly with sensitive antigens, remain challenging. The aim of our study was to develop a robust and reproducible protocol to uncover the immune landscape in mouse FFPE tissues. Effective antibody stripping while maintaining sensitivity to antigens and tissue adhesion to the glass slide is critical in developing an mIHC panel to allow successive rounds of staining. Thus, we identified a highly efficient stripping method that preserves signal intensity and antigenicity to allow multiple rounds of staining. We subsequently optimised an mIHC workflow with antibodies specific against CD4, CD8α, FOXP3 and B220 to identify distinct T and B cell populations on mouse FFPE tissues. Lastly, the application of this mIHC panel was validated in a mouse model of inflammatory bowel cancer, two allograft mouse models of spontaneous colon adenocarcinoma and a sporadic mouse model of colon cancer. Together, these demonstrate the utility of the aforementioned protocol in establishing the quantity and spatial localisation of immune cells in different pathological tissues.
Collapse
|
352
|
Pang B, Jin H, Liao N, Li J, Jiang C, Shi J. Vitamin A supplementation ameliorates ulcerative colitis in gut microbiota-dependent manner. Food Res Int 2021; 148:110568. [PMID: 34507723 DOI: 10.1016/j.foodres.2021.110568] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 12/27/2022]
Abstract
Ulcerative colitis (UC), is a chronic relapsing inflammatory condition of the gastrointestinal track. The purpose of this study is to explore whether Vitamin A (VA) can treat UC and its mechanisms. A mouse model of UC was established using 3.0% (w/v) dextran sodium sulfate (DSS). VA was used to treat UC by intragastric administration of 5000 international unit (IU) retinyl acetate. Fecal microbiota transplantation (FMT) was also used to treat the UC model mice to verify the effect of influenced gut microbiota. The content of short-chain fatty acids (SCFAs) in cecal contents was quantitatively detected by gas chromatography and mass spectrometry. VA supplementation significantly ameliorated UC. 16S rRNA sequencing indicated that VA-treated mice exhibited much more abundant gut microbial diversity and flora composition. Targeted metabolomics analysis manifested the increased production of SCFAs in VA-treated mice. Gut microbiota depletion and FMT results confirmed the gut microbiota-dependent mechanism as that VA relieved UC via regulating gut microbiota: increase in SCFA-producing genera and decrease in UC-related genera. The restore of intestinal barrier and the inhibition of inflammation were also found to contribute to the amelioration of UC by VA. It was concluded that a VA supplement was enough to cause a significant change in gut microbiota and amelioration of UC.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Ning Liao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Junjun Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
353
|
Kangwan N, Pintha K, Khanaree C, Kongkarnka S, Chewonarin T, Suttajit M. Anti-inflammatory effect of Perilla frutescens seed oil rich in omega-3 fatty acid on dextran sodium sulfate-induced colitis in mice. Res Pharm Sci 2021; 16:464-473. [PMID: 34522194 PMCID: PMC8407152 DOI: 10.4103/1735-5362.323913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/17/2021] [Accepted: 07/13/2021] [Indexed: 01/25/2023] Open
Abstract
Background and purpose Ulcerative colitis is a chronic inflammatory bowel disease that involves diffused inflammation of the large intestine. Omega-3 fatty acid (FA) has been known to regulate the inflammatory response associated with ulcerative colitis pathogenesis. Perilla frutescens is a valuable source of omega-3 FA and α-linolenic acid (ALA) contained in its seed oil. Therefore, the aim of this study was to evaluate the anti-inflammatory effect of Perilla seed oil (PSO) on colitis induced by dextran sulfate sodium (DSS) in a mouse model. Experimental approach PSO was extracted using a cold-pressed extractor and FA composition of PSO was analyzed by GC-MS. Acute colitis in mice was induced with 3% DSS in drinking water for 7 days. Some mice were treated with PSO (20, 100, 200 mg/kg BW) for 3 weeks before the DSS administration. Sulfasalazine was used as a positive control. The clinical features, histopathologic, serum, and gene expression of proinflammatory cytokines in the colon were assessed. Finding/Results PSO contained the highest proportion of ALA (61.51%). Furthermore, PSO pretreatment evidently reduced body weight loss, diminished diarrhea, gross bleeding, and DSS-induced colon shortening. PSO pretreatment attenuated histopathological changes in response to DSS-induced colitis. PSO pretreatment also markedly decreased inflammatory response in serum and the colon tissue of DSS-induced mice. Conclusion and implication ALA in PSO is suggested to be mainly responsible for the reduction of DSS-induced colitis through suppressing inflammatory markers. PSO could be further developed as a functional health supplement, which would be beneficial for anti-inflammation in the colonic mucosa.
Collapse
Affiliation(s)
- Napapan Kangwan
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Komsak Pintha
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Chakkrit Khanaree
- School of Traditional and Alternative Medicine, Chiang Rai Rajabhat University, Chiang Rai, Thailand
| | - Sarawut Kongkarnka
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Teera Chewonarin
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Maitree Suttajit
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| |
Collapse
|
354
|
Youssef ME, Abd El-Fattah EE, Abdelhamid AM, Eissa H, El-Ahwany E, Amin NA, Hetta HF, Mahmoud MH, Batiha GES, Gobba N, Ahmed Gaafar AG, Saber S. Interference With the AMPKα/mTOR/NLRP3 Signaling and the IL-23/IL-17 Axis Effectively Protects Against the Dextran Sulfate Sodium Intoxication in Rats: A New Paradigm in Empagliflozin and Metformin Reprofiling for the Management of Ulcerative Colitis. Front Pharmacol 2021; 12:719984. [PMID: 34489707 PMCID: PMC8417441 DOI: 10.3389/fphar.2021.719984] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022] Open
Abstract
Empagliflozin and metformin are widely used for the treatment of type 2 diabetes. These drugs showed marked anti-inflammatory effects in different animal models via enhancing AMPK activity. Yet, the protective anti-inflammatory effects of their combination against ulcerative colitis have not been previously investigated. The current study aimed to explore the potential of empagliflozin/metformin combination to mitigate the DSS-induced rat colitis model. The modulating effects of empagliflozin and metformin on the AMPK/mTOR/NLRP3 axis and T cell polarization were delineated. In this study, distal colons were examined for macroscopic and microscopic pathological alterations. ELISA, qRT-PCR, and immunohistochemistry techniques were applied to detect proteins and cytokines involved in AMPK/mTOR/NLRP3 axis and T Cell polarization. Oral administration of empagliflozin (10 mg/kg/day) and metformin (200 mg/kg/day) combination alleviated colitis as revealed by the reduced disease activity index, macroscopic damage index, colon weight/length ratio, and histopathologic scoring values. Interestingly, empagliflozin/metformin combination significantly enhanced AMPK phosphorylation and depressed mTOR and NLRP3 expression leading to a subsequent reduction in caspase-1 cleavage and inhibition of several inflammatory cytokines, including IL-1β, and IL-18. Reduced mTOR expression and reduced IL-6 levels led to a reduction in Th17 cell polarization and maintenance. Together, the current study reveals that the protective effects of empagliflozin and metformin against DSS-induced colitis are fundamentally mediated via enhancing AMPK phosphorylation. Since adult humans with diabetes mellitus are at greater risk for developing inflammatory bowel diseases, clinical application of empagliflozin/metformin combination represents a novel therapeutic approach for treating diabetic patients with ulcerative colitis.
Collapse
Affiliation(s)
- Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Amir M Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Hanan Eissa
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Noha A Amin
- Department of Hematology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.,Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Naglaa Gobba
- Department of Pharmacology and Toxicology, College of Pharmacy, Misr University for Science and Technology, 6th of October City, Egypt
| | - Ahmed Gaafar Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port-Said University, Port-Said, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
355
|
Yip JL, Balasuriya GK, Spencer SJ, Hill-Yardin EL. The Role of Intestinal Macrophages in Gastrointestinal Homeostasis: Heterogeneity and Implications in Disease. Cell Mol Gastroenterol Hepatol 2021; 12:1701-1718. [PMID: 34506953 PMCID: PMC8551786 DOI: 10.1016/j.jcmgh.2021.08.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
Intestinal macrophages play a key role in the gut immune system and the regulation of gastrointestinal physiology, including gut motility and secretion. Their ability to keep the gut from chronic inflammation despite constantly facing foreign antigens has been an important focus in gastrointestinal research. However, the heterogeneity of intestinal macrophages has impeded our understanding of their specific roles. It is now becoming clear that subsets of intestinal macrophages play diverse roles in various gastrointestinal diseases. This occurs through a complex interplay between cytokine production and enteric nervous system activation that differs for each pathologic condition. Key diseases and disorders in which intestinal macrophages play a role include postoperative ileus, inflammatory bowel disease, necrotizing enterocolitis, as well as gastrointestinal disorders associated with human immunodeficiency virus and Parkinson's disease. Here, we review the identification of intestinal macrophage subsets based on their origins and functions, how specific subsets regulate gut physiology, and the potential for these heterogeneous subpopulations to contribute to disease states. Furthermore, we outline the potential for these subpopulations to provide unique targets for the development of novel therapies for these disorders.
Collapse
Affiliation(s)
| | | | - Sarah J. Spencer
- School of Health and Biomedical Sciences,Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Royal Melbourne Instutite of Technology, Melbourne, Victoria, Australia
| | - Elisa L. Hill-Yardin
- School of Health and Biomedical Sciences,Correspondence Address correspondence to: Elisa L. Hill-Yardin, PhD, School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
356
|
Li C, Dong N, Wu B, Mo Z, Xie J, Lu Q. Dihydroberberine, an isoquinoline alkaloid, exhibits protective effect against dextran sulfate sodium-induced ulcerative colitis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153631. [PMID: 34253428 DOI: 10.1016/j.phymed.2021.153631] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND As a chronic inflammatory disease, ulcerative colitis (UC) is relevant to a rising risk of colorectal cancer. Dihydroberberine (DHBB), a natural occurring isoquinoline alkaloid with various bioactivities, was found in many plants including Coptis chinensis Franch. (Ranunculaceae), Phellodendron chinense Schneid. (Rutaceae), and Chelidonium majus L. (Papaveraceae). However, its protective effect on UC is sparsely dissected out. PURPOSE To explore the protective role and underlying mechanism of DHBB on a model of colitis. METHODS Acute colitis model was established by gavage with 3% dextran sulfate sodium (DSS) for 8 days. Influence of DHBB on DSS-induced clinical symptoms and disease activity index (DAI) was monitored and analyzed. Pathological injury of colon tissues was examined by hematoxylin-eosin and Alcian blue staining. The expression of intestinal mucosal barrier function proteins, immune-inflammation related biomarkers and signal pathway key targets were determined by ELISA kit, Western blot, immunohistochemistry and qRT-PCR. RESULTS DHBB treatment effectively alleviated DSS-induced UC by relieving clinical manifestations, DAI scores and pathological damage, which exerted similar beneficial effect to azathioprine (AZA), and better than berberine (BBR). In addition, DHBB significantly improved the gut barrier function through up-regulating the levels of tight junction proteins and mucins. Furthermore, DHBB dramatically ameliorated colonic immune-inflammation state, which was related to the decrease of colonic pro-inflammatory cytokines and immunoglobulin through blocking TLR4/MyD88/NF-κB signal pathway. CONCLUSION These results demonstrated that DHBB exerted a significant protective effect on DSS-induced experimental UC, at least partly through suppressing immune-inflammatory response and maintaining gut barrier function.
Collapse
Affiliation(s)
- Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China
| | - Na Dong
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China
| | - Bowen Wu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China
| | - Zhimi Mo
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China
| | - Jianhui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China.
| |
Collapse
|
357
|
Chen G, Yang Z, Wen D, Guo J, Xiong Q, Li P, Zhao L, Wang J, Wu C, Dong L. Polydatin has anti-inflammatory and antioxidant effects in LPS-induced macrophages and improves DSS-induced mice colitis. Immun Inflamm Dis 2021; 9:959-970. [PMID: 34010516 PMCID: PMC8342204 DOI: 10.1002/iid3.455] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Polydatin (PD), a monocrystalline compound isolated from the root and rhizome of Polygonum cuspidatum, is widely used in inhibiting the inflammatory response and oxidative stress. PD has an anti-inflammatory effect on colitis mice; however, information regulating the mechanism by which maintains the intestinal epithelium barrier is currently scarce. Here, we assessed the anti-inflammatory and antioxidant of PD in lipopolysaccharide (LPS)-induced macrophages in vitro, and explored its effects on inhibiting intestinal inflammation and maintaining the intestinal epithelium barrier in dextran sodium sulfate (DSS)-induced colitis mice. Results showed that PD reduced the level of proinflammatory cytokines and enzymes, including tumor necrosis factor-α, interleukin-4 (IL-4), IL-6, cyclooxygenase-2, and inducible nitric oxide synthase, in LPS-induced macrophages, and improved the expression level of IL-10. PD maintained the expression of tight junction proteins in medium (LPS-induced macrophages medium)-induced MCEC cells. Additionally, PD inhibited the phosphorylation of nuclear factor-κB (NF-κB), p65, extracellular signal-regulated kinase-1/2, c-Jun N-terminal kinase, and p38 signaling pathways in LPS-induced macrophages and facilitated the phosphorylation of AKT and the nuclear translocation of Nrf2, improving the expression of HO-1 and NQO1. Furthermore, PD ameliorated the intestinal inflammatory response and improved the dysfunction of the colon epithelium barrier in DSS-induced colitis mice. Taken together, our results indicated that PD inhibited inflammation and oxidative stress, maintained the intestinal epithelium barrier, and the protective role of PD was associated with the NF-κB p65, itogen-activated protein kinases, and AKT/Nrf2/HO-1/NQO1 signaling pathway.
Collapse
Affiliation(s)
- Guangxin Chen
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Ziyue Yang
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Da Wen
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Jian Guo
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
- Department of General Surgery, Shanxi Provincial People's HospitalAffiliate of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Qiuhong Xiong
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Ping Li
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's HospitalAffiliate of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Junping Wang
- Department of Gastroenterology, Shanxi Provincial People's HospitalAffiliate of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Changxin Wu
- Institute of Biomedical SciencesShanxi UniversityTaiyuanChina
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of EducationShanxi UniversityTaiyuanShanxiChina
| | - Lina Dong
- Central Laboratory, Shanxi Provincial People's HospitalAffiliate of Shanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
358
|
Morales Fénero C, Amaral MA, Xavier IK, Padovani BN, Paredes LC, Takiishi T, Lopes-Ferreira M, Lima C, Colombo A, Saraiva Câmara NO. Short chain fatty acids (SCFAs) improves TNBS-induced colitis in zebrafish. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:142-154. [PMID: 35492385 PMCID: PMC9040093 DOI: 10.1016/j.crimmu.2021.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022] Open
Abstract
The short-chain fatty acids (SCFAs) are metabolites originated from the fermentation of dietary fibers and amino acids produced by the bacteria of the intestinal microbiota. The most abundant SCFAs, acetate, propionate, and butyrate, have been proposed as a treatment for inflammatory bowel diseases (IBDs) due to their anti-inflammatory properties. This work aimed to analyze the effects of the treatment of three combined SCFAs in TNBS-induced intestinal inflammation in zebrafish larvae. Here, we demonstrated that SCFAs significantly increased the survival of TNBS-exposed larvae, preserved the intestinal endocytic function, reduced the expression of inflammatory cytokines and the intestinal recruitment of neutrophils caused by TNBS. However, SCFAs treatment did not appear to avoid TNBS-induced tissue damage in the intestinal wall and did not restore the number of mucus-producing goblet cells. Finally, exposure to TNBS induced dysbiosis of the microbiota with an increase in Betaproteobacteria and Actinobacteria, while the treatment with SCFAs maintained these population levels similar to control. Thus, we demonstrate that the treatment of three combined SCFAs presented anti-inflammatory properties previously seen in mammals, opening an opportunity to use zebrafish to explore the potential benefit of these and other metabolites to treat inflammation.
Collapse
Affiliation(s)
- Camila Morales Fénero
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Izabella Karina Xavier
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Barbara Nunes Padovani
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lais Cavalieri Paredes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tatiana Takiishi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Mônica Lopes-Ferreira
- Center of Toxins, Immune Response and Cellular Signalling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Carla Lima
- Center of Toxins, Immune Response and Cellular Signalling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Alicia Colombo
- Department of Pathologic Anatomy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Nephrology Division, Federal University of São Paulo, Brazil
| |
Collapse
|
359
|
Zhou Y, Zhong B, Min X, Hou Y, Lin L, Wu Q, Shi J, Chen X. Therapeutic potential of isobavachalcone, a natural flavonoid, in murine experimental colitis by inhibiting NF-κB p65. Phytother Res 2021; 35:5861-5870. [PMID: 34435401 DOI: 10.1002/ptr.7246] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
The incidence of ulcerative colitis (UC), one of the two types of inflammatory bowel disease, is increasing in many countries. Various natural products have been demonstrated with therapeutic potentials for UC. Herein, the therapeutic effects and mechanisms of isobavachalcone (IBC), a natural chalcone, were evaluated in dextran sulfate sodium (DSS)-induced colitis mice and lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. The results demonstrated that IBC treatment significantly improved the clinical symptoms, assessed by the disease activity index (DAI) scores and the histological changes of the colon. The levels of myeloperoxidase (MPO), TNF-α, IL-6, IL-1β, and prostaglandin E2 (PGE2) in colon tissues were suppressed by IBC. The upregulation of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and NF-κB p65 in colon tissues were reversed by IBC as well. Furthermore, IBC significantly inhibited LPS-triggered secretion of TNF-α, IL-6, and nitrite, and nuclear translocation of NF-κB p65, in RAW264.7 cells. The luciferase reporter assay indicated that IBC significantly inhibited LPS-triggered transcription of toll-like receptor 4 (TLR4). Molecular docking results showed that the binding pocket of IBC was adjacent to Ser276 of p65-p50 heterodimer and IBC could form H-bond with Thr191. Collectively, these results demonstrated that IBC ameliorated colitis in mice possibly through inhibition of NF-κB p65.
Collapse
Affiliation(s)
- Yishan Zhou
- Key Lab for Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiangjing Min
- Key Lab for Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Qin Wu
- Key Lab for Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Lab for Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
360
|
Role of MicroRNA in Inflammatory Bowel Disease: Clinical Evidence and the Development of Preclinical Animal Models. Cells 2021; 10:cells10092204. [PMID: 34571853 PMCID: PMC8468560 DOI: 10.3390/cells10092204] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
The dysregulation of microRNA (miRNA) is implicated in cancer, inflammation, cardiovascular disorders, drug resistance, and aging. While most researchers study miRNA's role as a biomarker, for example, to distinguish between various sub-forms or stages of a given disease of interest, research is also ongoing to utilize these small nucleic acids as therapeutics. An example of a common pleiotropic disease that could benefit from miRNA-based therapeutics is inflammatory bowel disease (IBD), which is characterized by chronic inflammation of the small and large intestines. Due to complex interactions between multiple factors in the etiology of IBD, development of therapies that effectively maintain remission for this disease is a significant challenge. In this review, we discuss the role of dysregulated miRNA expression in the context of clinical ulcerative colitis (UC) and Crohn's disease (CD)-the two main forms of IBD-and the various preclinical mouse models of IBD utilized to validate the therapeutic potential of targeting these miRNA. Additionally, we highlight advances in the development of genetically engineered animal models that recapitulate clinical miRNA expression and provide powerful preclinical models to assess the diagnostic and therapeutic promise of miRNA in IBD.
Collapse
|
361
|
Houser MC, Caudle WM, Chang J, Kannarkat GT, Yang Y, Kelly SD, Oliver D, Joers V, Shannon KM, Keshavarzian A, Tansey MG. Experimental colitis promotes sustained, sex-dependent, T-cell-associated neuroinflammation and parkinsonian neuropathology. Acta Neuropathol Commun 2021; 9:139. [PMID: 34412704 PMCID: PMC8375080 DOI: 10.1186/s40478-021-01240-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background The etiology of sporadic Parkinson’s disease (PD) remains uncertain, but genetic, epidemiological, and physiological overlap between PD and inflammatory bowel disease suggests that gut inflammation could promote dysfunction of dopamine-producing neurons in the brain. Mechanisms behind this pathological gut-brain effect and their interactions with sex and with environmental factors are not well understood but may represent targets for therapeutic intervention. Methods We sought to identify active inflammatory mechanisms which could potentially contribute to neuroinflammation and neurological disease in colon biopsies and peripheral blood immune cells from PD patients. Then, in mouse models, we assessed whether dextran sodium sulfate-mediated colitis could exert lingering effects on dopaminergic pathways in the brain and whether colitis increased vulnerability to a subsequent exposure to the dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We assessed the involvement of inflammatory mechanisms identified in the PD patients in colitis-related neurological dysfunction in male and female mice, utilizing mice lacking the Regulator of G-Protein Signaling 10 (RGS10)—an inhibitor of nuclear factor kappa B (NFκB)—to model enhanced NFκB activity, and mice in which CD8+ T-cells were depleted. Results High levels of inflammatory markers including CD8B and NFκB p65 were found in colon biopsies from PD patients, and reduced levels of RGS10 were found in immune cells in the blood. Male mice that experienced colitis exhibited sustained reductions in tyrosine hydroxylase but not in dopamine as well as sustained CD8+ T-cell infiltration and elevated Ifng expression in the brain. CD8+ T-cell depletion prevented colitis-associated reductions in dopaminergic markers in males. In both sexes, colitis potentiated the effects of MPTP. RGS10 deficiency increased baseline intestinal inflammation, colitis severity, and neuropathology. Conclusions This study identifies peripheral inflammatory mechanisms in PD patients and explores their potential to impact central dopaminergic pathways in mice. Our findings implicate a sex-specific interaction between gastrointestinal inflammation and neurologic vulnerability that could contribute to PD pathogenesis, and they establish the importance of CD8+ T-cells in this process in male mice. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01240-4.
Collapse
|
362
|
Estrogen receptor actions in colitis. Essays Biochem 2021; 65:1003-1013. [PMID: 34342357 DOI: 10.1042/ebc20210010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023]
Abstract
In recent years, researchers have demonstrated that estrogen and its receptors, aside from their role in regulating several biological functions, contribute to the development and progression/severity of inflammatory bowel diseases (IBDs). IBDs include both ulcerative colitis (UC) and Crohn's disease (CD). Epidemiological data indicate a clear difference in the incidence, severity, and complications of IBDs between sexes. Men present a higher risk of developing colitis than women and a higher risk of developing colorectal cancer, a common complication of this condition. However, fluctuations of estrogen levels have yielded inconsistent data, where oral contraceptives and hormone replacement therapy have been associated with an increased risk of IBDs in premenopausal women but significantly reduce disease activity after menopause. Likewise, improvement of symptoms related to CD has been reported during pregnancy, but not in UC, who often experience worsening symptoms. In the colonic epithelium, estrogen receptor β (ERβ) is the predominant form of the protein expressed, and it helps maintain normal epithelial function and organization. Preclinical data suggest that ER expression and activation via estrogen confers different responses on disease severity depending on the model used to induce colitis, which may reflect what is observed in patients with IBDs. Hence, this review aims to provide an overview of estrogen and its receptors, particularly ERβ, in the pathophysiology of IBDs.
Collapse
|
363
|
He N, Yang Y, Wang H, Liu N, Yang Z, Li S. Unsaturated alginate oligosaccharides (UAOS) protects against dextran sulfate sodium-induced colitis associated with regulation of gut microbiota. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
364
|
Arnesen H, Müller MHB, Aleksandersen M, Østby GC, Carlsen H, Paulsen JE, Boysen P. Induction of colorectal carcinogenesis in the C57BL/6J and A/J mouse strains with a reduced DSS dose in the AOM/DSS model. Lab Anim Res 2021; 37:19. [PMID: 34315530 PMCID: PMC8317392 DOI: 10.1186/s42826-021-00096-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/01/2021] [Indexed: 12/21/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most frequently diagnosed cancers worldwide and thus mouse models of CRC are of significant value to study the pathogenesis. The Azoxymethane/Dextran sulfate sodium (AOM/DSS) model is a widely used, robust initiation-promotion model for chemical induction of colitis-associated CRC in rodents. However, the dosage of chemicals, treatment regimens and outcome measures vary greatly among studies employing this model. Thus, the aim of this study was to examine an AOM/DSS model involving a reduced (1%) dose of DSS for induction of carcinogenesis in A/J and C57BL/6J (B6) mice. Results We show that colonic preneoplastic lesions can be reliably detected in A/J and B6 mice by use of a AOM/DSS model involving a single injection of 10 mg/kg AOM followed by three 7-day cycles of a low-dose (1%) DSS administration. Supporting existing evidence of A/J mice exhibiting higher susceptibility to AOM than B6 mice, our AOM/DSS-treated A/J mice developed the highest number of large colonic lesions. Clinical symptoms in both strains subjected to the AOM/DSS treatment did not persist in-between treatment cycles, demonstrating that the animals tolerated the treatment well. Conclusions Our findings suggest that a reduced dose of DSS in the AOM/DSS model can be considered in future studies of early phase colorectal carcinogenesis in the A/J and B6 mouse strains using preneoplastic lesions as an outcome measure, and that such regimen may reduce the risk of early trial terminations to accommodate human endpoints. Overall, our data emphasize the importance of devoting attention towards choice of protocol, outcome measures and mouse strain in studies of CRC in mice according to the study purpose. Supplementary Information The online version contains supplementary material available at 10.1186/s42826-021-00096-y.
Collapse
Affiliation(s)
- Henriette Arnesen
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway. .,Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway.
| | - Mette Helen Bjørge Müller
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Mona Aleksandersen
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Gunn Charlotte Østby
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Jan Erik Paulsen
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Preben Boysen
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| |
Collapse
|
365
|
Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J Immunol Res 2021; 2021:8030297. [PMID: 34337079 PMCID: PMC8324359 DOI: 10.1155/2021/8030297] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), increases gradually worldwide in the past decades. IBD is generally associated with the change of the immune system and gut microbiota, and the conventional treatments usually result in some side effects. Bifidobacterium longum, as colonizing bacteria in the intestine, has been demonstrated to be capable of relieving colitis in mice and can be employed as an alternative or auxiliary way for treating IBD. Here, the mechanisms of the Bifidobacterium longum in the treatment of IBD were summarized based on previous cell and animal studies and clinical trials testing bacterial therapies. This review will be served as a basis for future research on IBD treatment.
Collapse
|
366
|
Qu Y, Li X, Xu F, Zhao S, Wu X, Wang Y, Xie J. Kaempferol Alleviates Murine Experimental Colitis by Restoring Gut Microbiota and Inhibiting the LPS-TLR4-NF-κB Axis. Front Immunol 2021; 12:679897. [PMID: 34367139 PMCID: PMC8339999 DOI: 10.3389/fimmu.2021.679897] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022] Open
Abstract
Intestinal microbiota dysbiosis is an established characteristic of ulcerative colitis (UC). Regulating the gut microbiota is an attractive alternative UC treatment strategy, considering the potential adverse effects of synthetic drugs used to treat UC. Kaempferol (Kae) is an anti-inflammatory and antioxidant flavonoid derived from a variety of medicinal plants. In this study, we determined the efficacy and mechanism of action of Kae as an anti-UC agent in dextran sulfate sodium (DSS)-induced colitis mice. DSS challenge in a mouse model of UC led to weight loss, diarrhea accompanied by mucous and blood, histological abnormalities, and shortening of the colon, all of which were significantly alleviated by pretreatment with Kae. In addition, intestinal permeability was shown to improve using fluorescein isothiocyanate (FITC)-dextran administration. DSS-induced destruction of the intestinal barrier was also significantly prevented by Kae administration via increases in the levels of ZO-1, occludin, and claudin-1. Furthermore, Kae pretreatment decreased the levels of IL-1β, IL-6, and TNF-α and downregulated transcription of an array of inflammatory signaling molecules, while it increased IL-10 mRNA expression. Notably, Kae reshaped the intestinal microbiome by elevating the Firmicutes to Bacteroidetes ratio; increasing the linear discriminant analysis scores of beneficial bacteria, such as Prevotellaceae and Ruminococcaceae; and reducing the richness of Proteobacteria in DSS-challenged mice. There was also an evident shift in the profile of fecal metabolites in the Kae treatment group. Serum LPS levels and downstream TLR4-NF-κB signaling were downregulated by Kae supplementation. Moreover, fecal microbiota transplantation from Kae-treated mice to the DSS-induced mice confirmed the effects of Kae on modulating the gut microbiota to alleviate UC. Therefore, Kae may exert protective effects against colitis mice through regulating the gut microbiota and TLR4-related signaling pathways. This study demonstrates the anti-UC effects of Kae and its potential therapeutic mechanisms, and offers novel insights into the prevention of inflammatory diseases using natural products.
Collapse
Affiliation(s)
- Yifan Qu
- Inner Mongolia Clinical College, Inner Mongolia Medical University, Hohhot, China
- Clinical Laboratory, Inner Mongolia People’s Hospital, Hohhot, China
| | - Xinyi Li
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Fengying Xu
- Inner Mongolia Clinical College, Inner Mongolia Medical University, Hohhot, China
| | - Shimin Zhao
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Xuemei Wu
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuzhen Wang
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Jiming Xie
- Clinical Laboratory, Inner Mongolia People’s Hospital, Hohhot, China
| |
Collapse
|
367
|
Ahmad A, Ansari MM, Kumar A, Bishnoi M, Raza SS, Khan R. Aminocellulose - grafted polycaprolactone-coated core-shell nanoparticles alleviate the severity of ulcerative colitis: a novel adjuvant therapeutic approach. Biomater Sci 2021; 9:5868-5883. [PMID: 34286723 DOI: 10.1039/d1bm00877c] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory condition of colorectal regions. Existing therapies for UC face grave lacunae including off-target and other harmful side effects, extensive first-pass metabolism, rapid clearance, limited or poor drug absorption and various other limitations, resulting in lower bioavailability. These conditions demand advanced delivery strategies to inflammatory colonic conditions so that drugs can counter stomach acid, avail protective strategies at this pH and selectively deliver drugs to the colon. Therefore, this approach was undertaken to develop and characterize nanoparticles for the delivery of drugs glycyrrhizic acid as well as budesonide in UC. Biocompatible and biodegradable aminocellulose-conjugated polycaprolactone containing budesonide was covered onto gelatinous nanoparticles (NPs) loaded with GA. Nanoparticles were prepared by the solvent evaporation technique, which showed particle size of ∼230 nm, spherical shape, almost smooth morphological characters under transmission, scanning and atomic force microscopy. These NPs also improved disease activities like occult blood in the stool, length of the colon and fecal properties. The nanoparticle therapy appreciably decreased colonic mast cellular infiltration, significantly maintained mucin protection, ameliorated histological features of the colon. Furthermore, markers of inflammation such as iNOS, COX-2, IL1-β, TNF-α, NO, and MPO were also appreciably ameliorated with the therapy of dual drug-loaded nanoparticles. Overall, these results establish that dual drug-loaded core-shell NPs exhibit superior therapeutic properties over the free or naïve forms of GA and budesonide in acute colon inflammation and present advantages that may be assigned to their ability to significantly inhibit colon inflammatory conditions.
Collapse
Affiliation(s)
- Anas Ahmad
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Sahibzada Ajit Singh Nagar, Punjab Pin - 140306, India.
| | | | | | | | | | | |
Collapse
|
368
|
Neu3 neuraminidase induction triggers intestinal inflammation and colitis in a model of recurrent human food-poisoning. Proc Natl Acad Sci U S A 2021; 118:2100937118. [PMID: 34266954 DOI: 10.1073/pnas.2100937118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intestinal inflammation is the underlying basis of colitis and the inflammatory bowel diseases. These syndromes originate from genetic and environmental factors that remain to be fully identified. Infections are possible disease triggers, including recurrent human food-poisoning by the common foodborne pathogen Salmonella enterica Typhimurium (ST), which in laboratory mice causes progressive intestinal inflammation leading to an enduring colitis. In this colitis model, disease onset has been linked to Toll-like receptor-4-dependent induction of intestinal neuraminidase activity, leading to the desialylation, reduced half-life, and acquired deficiency of anti-inflammatory intestinal alkaline phosphatase (IAP). Neuraminidase (Neu) inhibition protected against disease onset; however, the source and identity of the Neu enzyme(s) responsible remained unknown. Herein, we report that the mammalian Neu3 neuraminidase is responsible for intestinal IAP desialylation and deficiency. Absence of Neu3 thereby prevented the accumulation of lipopolysaccharide-phosphate and inflammatory cytokine expression in providing protection against the development of severe colitis.
Collapse
|
369
|
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease characterized by inflammation of the large intestine, rectal bleeding, and abdominal pain. It can be alleviated by certain bioactive compounds, including α-linolenic acid (ALA), which is a bioactive component in fermented black radish (Raphanus sativus L. var. niger). The aim of this study was to evaluate the anti-inflammatory effects of ALA in dextran sulfate sodium (DSS)-induced UC in mice. UC was induced in C57BL/6 mice by allowing them to freely drink water containing 2.5% DSS for 7 days, followed by oral administration of ALA (30 and 60 mg/kg/day) or vehicle control for 7 days. DSS-induced colitis was evaluated using the Disease Activity Index (DAI) and by measuring colon length and performing a histopathological examination. Compared to the control group, the vehicle-treated group showed a higher DAI score, shorter colon, goblet cell loss, and crypt shortening. The ALA treatment mitigated clinical signs of UC and histopathological changes. Furthermore, it mitigated intestinal inflammation by reducing the expression of ionized calcium binding adaptor molecule 1-positive macrophages in the colon. These results show that ALA alleviates DSS-induced UC by suppressing colon damage, which includes goblet cell loss, crypt shortening, and a reduction of macrophages in the colon.
Collapse
|
370
|
Lajczak-McGinley NK, Porru E, Fallon CM, Smyth J, Curley C, McCarron PA, Tambuwala MM, Roda A, Keely SJ. The secondary bile acids, ursodeoxycholic acid and lithocholic acid, protect against intestinal inflammation by inhibition of epithelial apoptosis. Physiol Rep 2021; 8:e14456. [PMID: 32562381 PMCID: PMC7305237 DOI: 10.14814/phy2.14456] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022] Open
Abstract
Increased epithelial permeability is a key feature of IBD pathogenesis and it has been proposed that agents which promote barrier function may be of therapeutic benefit. We have previously reported the secondary bile acid, ursodeoxycholic acid (UDCA), to be protective in a mouse model of colonic inflammation and that its bacterial metabolism is required for its beneficial effects. The current study aimed to compare the effects of UDCA, LCA, and a non‐metabolizable analog of UDCA, 6‐methyl‐UDCA (6‐MUDCA), on colonic barrier function and mucosal inflammation in a mouse model of colonic inflammation. Bile acids were administered daily to C57Bl6 mice by intraperitoneal injection. Colonic inflammation, induced by addition of DSS (2.5%) to the drinking water, was measured as disease activity index (DAI) and histological score. Epithelial permeability and apoptosis were assessed by measuring FITC‐dextran uptake and caspase‐3 cleavage, respectively. Cecal bile acids were measured by HPLC‐MS/MS. UDCA and LCA, but not 6‐MUDCA, were protective against DSS‐induced increases in epithelial permeability and colonic inflammation. Furthermore, UDCA and LCA inhibited colonic epithelial caspase‐3 cleavage both in DSS‐treated mice and in an in vitro model of cytokine‐induced epithelial injury. HPLC‐MS/MS analysis revealed UDCA administration to increase colonic LCA levels, whereas LCA administration did not alter UDCA levels. UDCA, and its primary metabolite, LCA, protect against intestinal inflammation in vivo, at least in part, by inhibition of epithelial apoptosis and promotion of barrier function. These data suggest that clinical trials of UDCA in IBD patients are warranted.
Collapse
Affiliation(s)
- Natalia K Lajczak-McGinley
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Emanule Porru
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Ciara M Fallon
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Jessica Smyth
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Caitriona Curley
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Paul A McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Aldo Roda
- Department of Chemistry, University of Bologna, Bologna, Italy.,INBB, National Institute of Bio structures and Biosystems, Rome, Italy
| | - Stephen J Keely
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
371
|
Yang X, Peng H, Luo Z, Luo A, Cai M, Xu L, Wang H. The dietary carcinogen PhIP activates p53-dependent DNA damage response in the colon of CYP1A-humanized mice. Biofactors 2021; 47:612-626. [PMID: 33811687 DOI: 10.1002/biof.1730] [Citation(s) in RCA: 5] [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: 01/29/2021] [Accepted: 03/24/2021] [Indexed: 11/09/2022]
Abstract
Species differences in the metabolism of xenobiotics by cytochrome P450 are critical in evaluating the use of experimental animals in studying toxic compounds relevant to human diseases. 2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), which is produced by high-temperature cooking of fish and meat, is activated to become a carcinogen by cytochrome P4501A2 (CYP1A2) through N2 -hydroxylation in humans, but is detoxified by Cyp1a2 through 4'-hydroxylation in mice. CYP1A-humanized (hCYP1A) mice, in which mouse Cyp1a is replaced with human CYP1A, show constitutive human xenobiotic metabolism by hCYP1A, thereby serving as a suitable model for studying PhIP. Previous studies have demonstrated that oral administration of PhIP induces colon tumors in hCYP1A mice; however, these studies used a super-high dose, raising concerns regarding the relevance of the mechanism to human cancer. Herein, we systematically investigated PhIP-induced colon carcinogenesis in hCYP1A mice treated with lower doses. We found that a dose 2000 times lower than that used previously, which is comparable to human daily intake levels, could induce colon tumors, albeit at a lower incidence rate. We further investigated the transcriptome changes in the colon of hCYP1A mice treated with PhIP and identified that PhIP treatment increased the expression of Bax, Btg2, Ccng1, Cdkn1a, and Trp53inp1 and decreased the expression of Igf1 and Ccnd1. Since these genes are key components of the p53-dependent DNA damage response, the altered expression patterns indicated PhIP-induced DNA damage in hCYP1A mice. Together, these results prove that hCYP1A mice are suitable for studying PhIP-induced carcinogenesis and show that PhIP is an important colorectal cancer carcinogen in human diet.
Collapse
Affiliation(s)
- Xu Yang
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongxia Peng
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Luo
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ailing Luo
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mansi Cai
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Xu
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong Wang
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
372
|
Koppens MAJ, Davis H, Valbuena GN, Mulholland EJ, Nasreddin N, Colombe M, Antanaviciute A, Biswas S, Friedrich M, Lee L, Wang LM, Koelzer VH, East JE, Simmons A, Winton DJ, Leedham SJ. Bone Morphogenetic Protein Pathway Antagonism by Grem1 Regulates Epithelial Cell Fate in Intestinal Regeneration. Gastroenterology 2021; 161:239-254.e9. [PMID: 33819486 PMCID: PMC7613733 DOI: 10.1053/j.gastro.2021.03.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS In homeostasis, intestinal cell fate is controlled by balanced gradients of morphogen signaling. The bone morphogenetic protein (BMP) pathway has a physiological, prodifferentiation role, predominantly inferred through previous experimental pathway inactivation. Intestinal regeneration is underpinned by dedifferentiation and cell plasticity, but the signaling pathways that regulate this adaptive reprogramming are not well understood. We assessed the BMP signaling landscape and investigated the impact and therapeutic potential of pathway manipulation in homeostasis and regeneration. METHODS A novel mouse model was generated to assess the effect of the autocrine Bmp4 ligand on individual secretory cell fate. We spatiotemporally mapped BMP signaling in mouse and human regenerating intestine. Transgenic models were used to explore the functional impact of pathway manipulation on stem cell fate and intestinal regeneration. RESULTS In homeostasis, ligand exposure reduced proliferation, expedited terminal differentiation, abrogated secretory cell survival, and prevented dedifferentiation. After ulceration, physiological attenuation of BMP signaling arose through upregulation of the secreted antagonist Grem1 from topographically distinct populations of fibroblasts. Concomitant expression supported functional compensation after Grem1 deletion from tissue-resident cells. BMP pathway manipulation showed that antagonist-mediated BMP attenuation was obligatory but functionally submaximal, because regeneration was impaired or enhanced by epithelial overexpression of Bmp4 or Grem1, respectively. Mechanistically, Bmp4 abrogated regenerative stem cell reprogramming despite a convergent impact of YAP/TAZ on cell fate in remodeled wounds. CONCLUSIONS BMP signaling prevents epithelial dedifferentiation, and pathway attenuation through stromal Grem1 upregulation was required for adaptive reprogramming in intestinal regeneration. This intercompartmental antagonism was functionally submaximal, raising the possibility of therapeutic pathway manipulation in inflammatory bowel disease.
Collapse
Affiliation(s)
- Martijn A J Koppens
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Hayley Davis
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Gabriel N Valbuena
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Eoghan J Mulholland
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Nadia Nasreddin
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Mathilde Colombe
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Agne Antanaviciute
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Medical Research Council Weatherall Institute of Molecular Medicine Centre for Computational Biology, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sujata Biswas
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Matthias Friedrich
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, United Kingdom
| | - Lennard Lee
- Cancer Genetics and Evolution Laboratory, Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lai Mun Wang
- Department of Laboratory Medicine, Changi General Hospital, SingHealth, Singapore, Singapore
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland; Department of Oncology and Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - James E East
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, and Oxford National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Alison Simmons
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, and Oxford National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Douglas J Winton
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Leedham
- Intestinal Stem Cell Biology Lab, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom; Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, and Oxford National Institute for Health Research Biomedical Research Centre, Oxford, United Kingdom.
| |
Collapse
|
373
|
Flores SRL, Nelson S, Woloshun RR, Wang X, Ha JH, Lee JK, Yu Y, Merlin D, Collins JF. Intestinal iron absorption is appropriately modulated to match physiological demand for iron in wild-type and iron-loaded Hamp (hepcidin) knockout rats during acute colitis. PLoS One 2021; 16:e0252998. [PMID: 34143808 PMCID: PMC8213193 DOI: 10.1371/journal.pone.0252998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/26/2021] [Indexed: 11/18/2022] Open
Abstract
Mucosal damage, barrier breach, inflammation, and iron-deficiency anemia (IDA) typify ulcerative colitis (UC) in humans. The anemia in UC appears to mainly relate to systemic inflammation. The pathogenesis of this ‘anemia of inflammation’ (AI) involves cytokine-mediated transactivation of hepatic Hamp (encoding the iron-regulatory hormone, hepcidin). In AI, high hepcidin represses iron absorption (and iron release from stores), thus lowering serum iron, and restricting iron for erythropoiesis (causing anemia). In less-severe disease states, inflammation may be limited to the intestine, but whether this perturbs iron homeostasis is uncertain. We hypothesized that localized gut inflammation will increase overall iron demand (to support the immune response and tissue repair), and that hepatic Hamp expression will decrease in response, thus derepressing (i.e., enhancing) iron absorption. Accordingly, we developed a rat model of mild, acute colitis, and studied iron absorption and homeostasis. Rats exposed (orally) to DSS (4%) for 7 days had intestinal (but not systemic) inflammation, and biomarker analyses demonstrated that iron utilization was elevated. Iron absorption was enhanced (by 2-3-fold) in DSS-treated, WT rats of both sexes, but unexpectedly, hepatic Hamp expression was not suppressed. Therefore, to gain a better understanding of regulation of iron absorption during acute colitis, Hamp KO rats were used for further experimentation. The severity of DSS-colitis was similar in Hamp KOs as in WT controls. In the KOs, increased iron requirements associated with the physiological response to colitis were satisfied by mobilizing hepatic storage iron, rather than by increasing absorption of enteral iron (as occurred in WT rats). In conclusion then, in both sexes and genotypes of rats, iron absorption was appropriately modulated to match physiological demand for dietary iron during acute intestinal inflammation, but regulatory mechanisms may not involve hepcidin.
Collapse
Affiliation(s)
- Shireen R. L. Flores
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Savannah Nelson
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Regina R. Woloshun
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Xiaoyu Wang
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Jung-Heun Ha
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Jennifer K. Lee
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Yang Yu
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
| | - Didier Merlin
- Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, GA, United States of America
| | - James F. Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
374
|
Shan D, Zheng J, Klimowicz A, Panzenbeck M, Liu Z, Feng D. Deep learning for discovering pathological continuum of crypts and evaluating therapeutic effects: An implication for in vivo preclinical study. PLoS One 2021; 16:e0252429. [PMID: 34125849 PMCID: PMC8202954 DOI: 10.1371/journal.pone.0252429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 05/16/2021] [Indexed: 11/21/2022] Open
Abstract
Applying deep learning to the field of preclinical in vivo studies is a new and exciting prospect with the potential to unlock decades worth of underutilized data. As a proof of concept, we performed a feasibility study on a colitis model treated with Sulfasalazine, a drug used in therapeutic care of inflammatory bowel disease. We aimed to evaluate the colonic mucosa improvement associated with the recovery response of the crypts, a complex histologic structure reflecting tissue homeostasis and repair in response to inflammation. Our approach requires robust image segmentation of objects of interest from whole slide images, a composite low dimensional representation of the typical or novel morphological variants of the segmented objects, and exploration of image features of significance towards biology and treatment efficacy. Both interpretable features (eg. counts, area, distance and angle) as well as statistical texture features calculated using Gray Level Co-Occurance Matrices (GLCMs), are shown to have significance in analysis. Ultimately, this analytic framework of supervised image segmentation, unsupervised learning, and feature analysis can be generally applied to preclinical data. We hope our report will inspire more efforts to utilize deep learning in preclinical in vivo studies and ultimately make the field more innovative and efficient.
Collapse
Affiliation(s)
- Dechao Shan
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Jie Zheng
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Alexander Klimowicz
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Mark Panzenbeck
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Zheng Liu
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Di Feng
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| |
Collapse
|
375
|
Pang B, Jin H, Liao N, Li J, Jiang C, Shao D, Shi J. Lactobacillus rhamnosus from human breast milk ameliorates ulcerative colitis in mice via gut microbiota modulation. Food Funct 2021; 12:5171-5186. [PMID: 33977948 DOI: 10.1039/d0fo03479g] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gut microbiota imbalance is one of the major causes of ulcerative colitis (UC). L. rhamnosus SHA113 (LRS), a strain isolated from healthy human milk, influences the regulation of gut flora. This study aims to determine whether this strain can ameliorate UC by modulating gut microbiota. Mouse models of UC were established using C57BL/6Cnc mice with intragastric administration of 3.0% (w/v) dextran sodium sulfate (DSS). LRS was used to treat the mouse models of UC with 109 cfu mL-1 cell suspension via intragastric administration. To verify the effect of gut microbiota on UC, fecal microbiota collected from the mice after the treatment with LRS were also used to treat the UC mouse models (FMT). The severity of UC was evaluated based on body weight, colon length, disease activity index (DAI), and hematoxylin-eosin staining. The microbial composition was analyzed by 16S rRNA sequencing. The mRNA expression levels of cytokines, mucins, tight junction proteins, and antimicrobial peptides in the gastrointestinal tract were detected by quantitative real-time polymerase chain reaction. The short-chain fatty acid (SCFAs) in the cecal contents of all mice were quantitatively detected by gas chromatography and mass spectrometry. Both LRS and FMT exerted excellent therapeutic effects on UC, as evidenced by the reduction in body weight loss, colon length, and colon structural integrity, as well as the increase in the DAI (disease activity index). LRS and FMT treatments showed similar effects: (1) an increase of total SCFA production in the cecal contents and the abundance of gut microbial diversity and flora composition; (2) decreases in two genera (Parabacteroides and Escherichia/Shigella) related to the DAI and the enhancement of SCFAs and IL-10 positively related genera in the gut microbiota (Bilophila, Roseburia, Akkermansia, and Bifidobacterium); (3) downregulation of the expression of tumor necrosis factor-α, interleukin IL-6, and IL-1β, and upregulation of the expression of the anti-inflammatory cytokine IL-10; and (4) upregulation of the expression of mucins (Muc1-4) and tight junction protein ZO-1. Overall, L. rhamnosus SHA113 relieves UC via the regulation of gut microbiota: increases in SCFA-producing genera and decreases in UC-related genera. In addition, a single strain is sufficient to induce a significant change in the gut microbiota and exert therapeutic effects on UC.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Ning Liao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Junjun Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| |
Collapse
|
376
|
Alshehri D, Saadah O, Mosli M, Edris S, Alhindi R, Bahieldin A. Dysbiosis of gut microbiota in inflammatory bowel disease: Current therapies and potential for microbiota-modulating therapeutic approaches. Bosn J Basic Med Sci 2021; 21:270-283. [PMID: 33052081 PMCID: PMC8112554 DOI: 10.17305/bjbms.2020.5016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
There is a growing body of evidence reinforcing the unique connections between the host microbiome, health, and diseases. Due to the extreme importance of the symbiotic relationship between the intestinal microbiome and the host, it is not surprising that any alteration in the gut microbiota would result in various diseases, including inflammatory bowel disease (IBD), Crohn's disease, (CD) and ulcerative colitis (UC). IBD is a chronic, relapsing-remitting condition that is associated with significant morbidity, mortality, compromised quality of life, and costly medical care. Dysbiosis is believed to exacerbate the progression of IBD. One of the currently used treatments for IBD are anti-tumor necrosis factor (TNF) drugs, representing a biologic therapy that is reported to have an impact on the gut microbiota composition. The efficacy of anti-TNF agents is hindered by the possibility of non-response, which occurs in 10-20% of treated patients, and secondary loss of response, which occurs in up to 30% of treated patients. This underscores the need for novel therapies and studies that evaluate the role of the gut microbiota in these conditions. The success of any therapeutic strategy for IBD depends on our understanding of the interactions that occur between the gut microbiota and the host. In this review, the health and disease IBD-associated microbiota patterns will be discussed, in addition to the effect of currently used therapies for IBD on the gut microbiota composition, as well as new therapeutic approaches that can be used to overcome the current treatment constraints.
Collapse
Affiliation(s)
- Dikhnah Alshehri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Biology, Faculty of Science, Tabuk University, Tabuk, Saudi Arabia
| | - Omar Saadah
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Mosli
- Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif Edris
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetics, Faculty of Agriculture, Ain Shams University, Cairo, Egypt; Princess Al Jawhara Albrahim Center of Excellence in Research of Hereditary Disorders (PACER-HD), Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rashad Alhindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetics, Faculty of Agriculture, Ain Shams University, Cairo, Egypt
| |
Collapse
|
377
|
Arias SL, Brito IL. Biophysical determinants of biofilm formation in the gut. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 18:100275. [PMID: 34504987 PMCID: PMC8423371 DOI: 10.1016/j.cobme.2021.100275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gastrointestinal (GI) tract harbors the most complex microbial ecosystem in the human body. The mucosal layer that covers the GI tract serves as a polymer-based defensive barrier that prevents the microbiome from reaching the epithelium and disseminating inside the body. Colonization of the mucus may result in the formation of structured polymicrobial communities or biofilms, a hallmark in pathologies such as colorectal cancer, inflammatory bowel disease, and chronic gut wounds. However, the mechanisms by which multispecies biofilms establish on the gut mucosa is unknown. Whether mucus-associated biofilms exist as part of a healthy mucosal barrier is still debated. Here, we discuss the impact that diet and microbial-derived polymers has on mucus structure and microcolony formation and highlight relevant biophysical forces that further shape nascent biofilms.
Collapse
Affiliation(s)
- Sandra L Arias
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850
| |
Collapse
|
378
|
Cuellar-Núñez ML, Gonzalez de Mejia E, Loarca-Piña G. Moringa oleifera leaves alleviated inflammation through downregulation of IL-2, IL-6, and TNF-α in a colitis-associated colorectal cancer model. Food Res Int 2021; 144:110318. [PMID: 34053523 DOI: 10.1016/j.foodres.2021.110318] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
New chemopreventive alternatives are needed due to the rising worldwide incidence of colorectal cancer. The objective was to evaluate the chemopreventive activity of Moringa oleifera leaves (MO) in a colitis-associated colon carcinogenesis model. We hypothesized that MO contain bioactive compounds capable of modulating the expression of genes involved in the inflammatory response and carcinogenesis. Forty-eight male mice (CD-1) were divided into six groups; 1: Healthy control; 2: Positive control induced with azoxymethane (AOM, 10 mg/Kg body weight, intraperitoneal injection) and three cycles of dextran sodium sulfate (DSS, 1.5% in drinking water); groups 3, 4, and 5 were induced with AOM/DSS and supplemented with 5%, 10%, and 20% of MO, respectively; group 6: had no disease induction and supplemented with 20% of MO. Mice were treated for 12 weeks and euthanized. Significant differences (p < 0.05) were found for the moringa-administered groups in morphological and histopathological parameters compared to the AOM/DSS control. A decrease in myeloperoxidase activity (~50%) and lipid peroxidation (1.9-3.1 times) were found in groups with 10% and 20% of MO compared to the AOM/DSS control (p < 0.05). The group supplemented with 10% MO showed a significant increase (~3 times) in butyrate and propionate in fecal and cecal content. Groups supplemented with 10%, and 20% MO showed a reduction in proinflammatory cytokines in serum (MCP-1, IL-6, TNF-α) compared to the AOM/DSS control. Treatment with 10% MO induced differential expression of 65 genes in colon tissue such as IL-2, IL-6, TNF, IL-1ß, and INF-γ. MO downregulated proinflammatory mediators showing chemopreventive properties against inflammatory response and colon carcinogenesis.
Collapse
Affiliation(s)
- M L Cuellar-Núñez
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Qro 76010, Mexico; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, IL 61801, United States
| | - E Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, IL 61801, United States
| | - G Loarca-Piña
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Qro 76010, Mexico.
| |
Collapse
|
379
|
Daskalaki MG, Axarlis K, Aspevik T, Orfanakis M, Kolliniati O, Lapi I, Tzardi M, Dermitzaki E, Venihaki M, Kousoulaki K, Tsatsanis C. Fish Sidestream-Derived Protein Hydrolysates Suppress DSS-Induced Colitis by Modulating Intestinal Inflammation in Mice. Mar Drugs 2021; 19:312. [PMID: 34071180 PMCID: PMC8228426 DOI: 10.3390/md19060312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease is characterized by extensive intestinal inflammation, and therapies against the disease target suppression of the inflammatory cascade. Nutrition has been closely linked to the development and suppression of inflammatory bowel disease, which to a large extent is attributed to the complex immunomodulatory properties of nutrients. Diets containing fish have been suggested to promote health and suppress inflammatory diseases. Even though most of the health-promoting properties of fish-derived nutrients are attributed to fish oil, the potential health-promoting properties of fish protein have not been investigated. Fish sidestreams contain large amounts of proteins, currently unexploited, with potential anti-inflammatory properties, and may possess additional benefits through bioactive peptides and free amino acids. In this project, we utilized fish protein hydrolysates, based on mackerel and salmon heads and backbones, as well as flounder skin collagen. Mice fed with a diet supplemented with different fish sidestream-derived protein hydrolysates (5% w/w) were exposed to the model of DSS-induced colitis. The results show that dietary supplements containing protein hydrolysates from salmon heads suppressed chemically-induced colitis development as determined by colon length and pro-inflammatory cytokine production. To evaluate colitis severity, we measured the expression of different pro-inflammatory cytokines and chemokines and found that the same supplement suppressed the pro-inflammatory cytokines IL-6 and TNFα and the chemokines Cxcl1 and Ccl3. We also assessed the levels of the anti-inflammatory cytokines IL-10 and Tgfb and found that selected protein hydrolysates induced their expression. Our findings demonstrate that protein hydrolysates derived from fish sidestreams possess anti-inflammatory properties in the model of DSS-induced colitis, providing a novel underexplored source of health-promoting dietary supplements.
Collapse
Affiliation(s)
- Maria G. Daskalaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Konstantinos Axarlis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Tone Aspevik
- Department of Nutrition and Feed Technology, Nofima AS, 5141 Bergen, Norway; (T.A.); (K.K.)
| | - Michail Orfanakis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Ourania Kolliniati
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Ioanna Lapi
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Maria Tzardi
- Laboratory of Pathology, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - Eirini Dermitzaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Maria Venihaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Katerina Kousoulaki
- Department of Nutrition and Feed Technology, Nofima AS, 5141 Bergen, Norway; (T.A.); (K.K.)
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| |
Collapse
|
380
|
Shang L, Liu H, Yu H, Chen M, Yang T, Zeng X, Qiao S. Core Altered Microorganisms in Colitis Mouse Model: A Comprehensive Time-Point and Fecal Microbiota Transplantation Analysis. Antibiotics (Basel) 2021; 10:antibiotics10060643. [PMID: 34071229 PMCID: PMC8230101 DOI: 10.3390/antibiotics10060643] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn’s disease (CD) and ulcerative colitis (UC), is characterized by chronic and relapsing inflammation within the gastrointestinal tract. Antibiotics have been used to treat IBD, primarily utilizing metronidazole. Although there does seem to be a treatment effect, the broad-spectrum antibiotics that have been used to date are crude tools and have many adverse effects. Available evidence suggests that the host microbiome is implicated in the pathogenesis of IBD, though the key bacteria remain unknown. If the bacterial population can be modified appropriately, the use of antibiotics will have a better therapeutic effect. In this study, mice were fed dextran sodium sulfate (DSS) solution for 5 days, followed by 5 days of normal drinking water, to investigate the gut microbiota response to colitis and the initial alteration of microbiota in recovery phase. Day 0 was considered the normal control, while day 5 and day 10 were considered the colitis mouse model progressive phase and recovery phase, respectively. Results showed that inflammation could induce proportional changes in the gut microbiota. Furthermore, transplanting the microbiota in progressive phase to antibiotic-induced microbiota-depleted mice could induce inflammation similar to colitis, which proves the importance of initial alteration of the microbiota for IBD recovery and the potential of the microbiota as a target for the treatment of IBD. Meanwhile, we have also identified three possible target microorganisms in the development of colitis, namely genera Muribaculaceae (negative correlation), Turicibacter (positive correlation) and Lachnospiraceae (negative correlation) in inflammation status through comprehensive analysis.
Collapse
Affiliation(s)
- Lijun Shang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Hongbin Liu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Tianren Yang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, China; (L.S.); (H.L.); (H.Y.); (M.C.); (T.Y.); (X.Z.)
- Beijing Bio-Feed Additives Key Laboratory, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62733588; Fax: +86-10-62733688
| |
Collapse
|
381
|
华 慧, 董 昕, 张 雨, 方 凡, 张 蓓, 李 向, 于 倩, 郑 葵, 颜 超. [rCsHscB derived from Clonorchis sinensis has therapeutic effect on dextran sodium sulfate-induced chronic ulcerative colitis in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:664-670. [PMID: 34134952 PMCID: PMC8214966 DOI: 10.12122/j.issn.1673-4254.2021.05.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the therapeutic effect of rCsHscB derived from Clonorchis sinensis on dextran sodium sulfate (DSS)-induced chronic ulcerative colitis in mice. OBJECTIVE C57BL/6 mice were randomized into negative control (NC) group (n= 10), rCsHscB group (n=10), DSS group (n=15), and DSS+rCsHscB group (n=15), and in the latter two groups, chronic ulcerative colitis was induced in the mice using 2% DSS. In rCsHscB and DSS+ rCsHscB groups, the mice received intraperitoneal injections of 125 μg/mL rCsHscB on the 4th and 7th day following DSS administration, and PBS was injected in the other two groups. The mice were euthanized on the 84th day, and pathological changes of the colon were evaluated by HE and Masson staining. The levels of CD4+ and CD8+ T cells in the peripheral blood and lamina propria gastric lymphocytes (LPL) were analyzed by flow cytometer; the levels of IL-6, MCP-1 and IL-10 in colon homogenate were determined using ELISA, and the phosphorylation of ERK1/2, JNK and P38 was detected with Western blotting. OBJECTIVE Compared with those in NC group, the mice in rCsHscB group exhibited no adverse responses to the treatment. The mice in DSS group had severe pathologies in the colon with significantly increased ratios of CD4+ and CD4+/CD8+ T cells in peripheral blood and LPL, increased levels of IL-6 and MCP-1 but no obvious changes in IL-10 in colon homogenate, and significantly augmented phosphorylation levels of ERK1/2, JNK and P38. Compared with those in DSS group, the mice in DSS+ rCsHscB group showed ameliorated colon pathologies with decreased CD4+T/CD8+T cell ratio in the peripheral blood and LPL, significantly decreased IL-6 and MCP-1 levels and increased IL-10 in colon homogenate, and lowered phosphorylation levels of ERK1/2, JNK and P38. OBJECTIVE rCsHscB can produce therapeutic effect on DSS-induced chronic ulcerative colitis in mice possibly by inhibiting the production of pro-inflammatory factors and regulating the balance of CD4+/CD8+T cells through the MAPK pathway.
Collapse
Affiliation(s)
- 慧 华
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 昕 董
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 雨钊 张
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 凡 方
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 蓓蓓 张
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 向阳 李
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 倩 于
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 葵阳 郑
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| | - 超 颜
- />徐州医科大学病原生物学与免疫学教研室,江苏省免疫与代谢重点实验室,徐州市感染与免疫重点实验室,江苏 徐州 221004Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogen and Immunology, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
382
|
Pabari RM, Tambuwala MM, Lajczak-McGinley N, Aljabali A, Kirby BP, Keely S, Ramtoola Z. Novel polyurethane based particulate formulations of infliximab reduce inflammation in DSS induced murine model of colitis - A preliminary study. Int J Pharm 2021; 604:120717. [PMID: 34015378 DOI: 10.1016/j.ijpharm.2021.120717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 11/19/2022]
Abstract
Our recent study showed that novel infliximab (INF) loaded polyesterurethane (INF-PU) and INF-PU-PEG particulate formulations reduced inflammation in an in-vitro epithelial inflammation model. In this study we investigated therapeutic potential of novel INF-PU and INF-PU-PEG particulate formulations to reduce inflammation in a dextran sodium sulfate (DSS) induced murine model of colitis. Severity of colitis was assessed by measurement of disease activity index (DAI) score, inflammatory markers (neutrophil infiltration, TNFα) and histological score. Treatment groups orally administered with INF-PU and INF-PU-PEG particulate formulations showed improvement in the clinical signs of colitis, similar to that observed with intraperitoneally administered INF, in both, moderate and severe DSS induced colitis model. This was related to a significant reduction in inflammatory cytokines, resulting in a significant reduction in histological score (ANOVA; p < 0.05), indicative of mucosal healing, a key goal of IBD therapy. This could be attributed to its targeted delivery to the inflamed colon and higher permeation of these particulate formulations across the inflamed colonic mucosa, as observed by the confocal images, resulting in local inhibition of TNFα at its site of production. These promising preliminary results warrant further investigation of orally administered INF and its novel particulate formulations in a wider preclinical study.
Collapse
Affiliation(s)
- Ritesh M Pabari
- RCSI, University of Medicine and Health Sciences, Dublin, Ireland.
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County, Londonderry BT52 1SA, Northern Ireland, United Kingdom
| | | | - Alaa Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Brian P Kirby
- RCSI, University of Medicine and Health Sciences, Dublin, Ireland
| | - Stephen Keely
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | |
Collapse
|
383
|
Ye J, Erland LAE, Gill SK, Bishop SL, Verdugo-Meza A, Murch SJ, Gibson DL. Metabolomics-Guided Hypothesis Generation for Mechanisms of Intestinal Protection by Live Biotherapeutic Products. Biomolecules 2021; 11:738. [PMID: 34063522 PMCID: PMC8156236 DOI: 10.3390/biom11050738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
The use of live biotherapeutic products (LBPs), including single strains of beneficial probiotic bacteria or consortiums, is gaining traction as a viable option to treat inflammatory-mediated diseases like inflammatory bowel disease (IBD). However, LBPs' persistence in the intestine is heterogeneous since many beneficial bacteria lack mechanisms to tolerate the inflammation and the oxidative stress associated with IBD. We rationalized that optimizing LBPs with enhanced colonization and persistence in the inflamed intestine would help beneficial bacteria increase their bioavailability and sustain their beneficial responses. Our lab developed two bioengineered LBPs (SBT001/BioPersist and SBT002/BioColoniz) modified to enhance colonization or persistence in the inflamed intestine. In this study, we examined colon-derived metabolites via ultra-high performance liquid chromatography-mass spectrometry in colitic mice treated with either BioPersist or BioColoniz as compared to their unmodified parent strains (Escherichia coli Nissle 1917 [EcN] and Lactobacillus reuteri, respectively) or to each other. BioPersist administration resulted in lowered concentrations of inflammatory prostaglandins, decreased stress hormones such as adrenaline and corticosterone, increased serotonin, and decreased bile acid in comparison to EcN. In comparison to BioColoniz, BioPersist increased serotonin and antioxidant production, limited bile acid accumulation, and enhanced tissue restoration via activated purine and pyrimidine metabolism. These data generated several novel hypotheses for the beneficial roles that LBPs may play during colitis.
Collapse
Affiliation(s)
- Jiayu Ye
- Department of Biology, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Lauren A E Erland
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Sandeep K Gill
- Department of Biology, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Stephanie L Bishop
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Andrea Verdugo-Meza
- Department of Biology, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Susan J Murch
- Department of Chemistry, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| | - Deanna L Gibson
- Department of Biology, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
- Department of Medicine, University of British Columbia, Syilx Okanagan Nation Territory, Kelowna, BC V1V1V7, Canada
| |
Collapse
|
384
|
Schiller M, Azulay-Debby H, Boshnak N, Elyahu Y, Korin B, Ben-Shaanan TL, Koren T, Krot M, Hakim F, Rolls A. Optogenetic activation of local colonic sympathetic innervations attenuates colitis by limiting immune cell extravasation. Immunity 2021; 54:1022-1036.e8. [PMID: 33932356 PMCID: PMC8116309 DOI: 10.1016/j.immuni.2021.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 01/16/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
The sympathetic nervous system is composed of an endocrine arm, regulating blood adrenaline and noradrenaline, and a local arm, a network of fibers innervating immune organs. Here, we investigated the impact of the local arm of the SNS in an inflammatory response in the colon. Intra-rectal insertion of an optogenetic probe in mice engineered to express channelrhodopsin-2 in tyrosine hydroxylase cells activated colonic sympathetic fibers. In contrast to systemic application of noradrenaline, local activation of sympathetic fibers attenuated experimental colitis and reduced immune cell abundance. Gene expression profiling showed decreased endothelial expression of the adhesion molecule MAdCAM-1 upon optogenetic stimulation; this decrease was sensitive to adrenergic blockers and 6-hydroxydopamine. Antibody blockade of MAdCAM-1 abrogated the optogenetic effect on immune cell extravasation into the colon and the pathology. Thus, sympathetic fibers control colonic inflammation by regulating immune cell extravasation from circulation, a mechanism likely relevant in multiple organs.
Collapse
Affiliation(s)
- Maya Schiller
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Hilla Azulay-Debby
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Nadia Boshnak
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Yehezqel Elyahu
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Ben Korin
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Tamar L Ben-Shaanan
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Tamar Koren
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Maria Krot
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Fahed Hakim
- Cancer Research Center, EMMS Nazareth, 16100, Nazareth, Israel; Azrieli faculty of medicine, Bar-Ilan university, 1311502, Safad, Israel
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel.
| |
Collapse
|
385
|
Qiu F, Zhang Z, Yang L, Li R, Ma Y. Combined effect of vitamin C and vitamin D 3 on intestinal epithelial barrier by regulating Notch signaling pathway. Nutr Metab (Lond) 2021; 18:49. [PMID: 33964955 PMCID: PMC8105975 DOI: 10.1186/s12986-021-00576-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background Tight junction proteins play crucial roles in maintaining the intestinal mucosal barrier. Although previous studies have shown that Notch signaling is closely related to tight junction proteins, the mechanism remains unclear. This study was performed to investigate whether vitamin C combined with vitamin D3 affects intestinal mucosal barrier stability via the Notch signaling pathway. Methods Intestinal epithelial barrier and notch signaling pathway were studied using guinea pig and SW480 cells. The guinea pigs were randomized into four groups (n = 6 in each group): control group (C, 200 IU/kg d VD3 + 100 mg/kg d VC), low VC group (LVC, 200 IU/kg d VD3 + 10 mg/kg d VC), medium VC group (MVC, 200 IU/kg d VD3 + 100 mg/kg d VC), and high VC group (HVC, 200 IU/kg d VD3 + 200 mg/kg d VC). Except for the control group, the other three groups were freely drinked with 2% dextran sodium sulfate solution for 4 days. And the control group was free to drink distilled water. The following cell groups were used: control group (SW480 cells without intervention); LPS group (100 ng/mL LPS); VD3 group (0.1 μmol/L VD3); VC + VD3 group (0.1, 1, 5, 10 μmol/mL VC + 0.1 μmol/L VD3). Results Electron microscopy analysis revealed that both low and high doses of vitamin C combined with vitamin D3 maintained dextran sodium sulfate-induced ulcerative colitis in the guinea pig intestinal epithelium tight junction. Compared with the control group, the expression level of ZO-1 mRNA in the colon tissue of the high-dose vitamin C group was significantly increased. In SW480 cell experiments, compared with the control group, cell migration and repair following treatment with different concentrations of vitamin C combined with vitamin D3 were significantly improved and the protein expression of Notch-1 was increased, whereas the protein expression of claudin-2 was significantly decreased. Thus, our results demonstrate that an appropriate amount of vitamin C combined with vitamin D3 can regulate the expression of claudin-2 by regulating Notch-1, relieve destruction of the intestinal mucosal barrier, and promote the repair of damage to the cell mucosal barrier. Conclusions We found that vitamin C combined with vitamin D3 protected against dextran sodium sulfate-induced ulcerative colitis in the guinea pig intestinal mucosa.
Collapse
Affiliation(s)
- Fubin Qiu
- Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, China.
| | - Zehui Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, China
| | - Linxue Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, China
| | - Rui Li
- Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, China
| | - Ying Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, China
| |
Collapse
|
386
|
Li J, Mu Y, Liu Y, Kishimura A, Mori T, Katayama Y. Effect of Size and Loading of Retinoic Acid in Polyvinyl Butyrate Nanoparticles on Amelioration of Colitis. Polymers (Basel) 2021; 13:1472. [PMID: 34063206 PMCID: PMC8124360 DOI: 10.3390/polym13091472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
Butyrate has been used in the treatment of inflammatory bowel diseases (IBD). However, the controlled release of butyrate has been indicated to be necessary in order to avoid the side effects verified at high concentrations. We previously developed nanoparticles (NPs) of polyvinyl butyrate (PVBu) as an oral butyrate donor for the controlled release of butyrate for the treatment of colitis. To examine the effect of the size of NPs on the therapeutic effect of colitis, here we prepared PVBu NPs with different sizes (100 nm and 200 nm). Both sizes of PVBu NPs significantly suppressed the inflammatory response in macrophages in vitro. PVBu NPs with 200 nm showed better effects on the amelioration of colitis compared with the 100 nm-NPs. We found unexpectedly that 200 nm-NP incorporated with all-trans retinoic acid (ATRA) showed a much better therapeutic effect than those with unloaded 200 nm-NPs, although ATRA alone was reported to worsen the inflammation. The synergistic effect of ATRA with butyrate shows evidence of being a promising approach for IBD treatment.
Collapse
Affiliation(s)
- Jinting Li
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (J.L.); (Y.M.); (A.K.)
| | - Yunmei Mu
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (J.L.); (Y.M.); (A.K.)
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (J.L.); (Y.M.); (A.K.)
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- International Research Center for Molecular System, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (J.L.); (Y.M.); (A.K.)
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (J.L.); (Y.M.); (A.K.)
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- International Research Center for Molecular System, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Rd., Chung Li 32023, Taiwan
| |
Collapse
|
387
|
Chen T, Zeineldin M, Johnson BA, Dong Y, Narkar A, Li T, Zhu J, Li R, Larman TC. Colonic epithelial adaptation to EGFR-independent growth induces chromosomal instability and is accelerated by prior injury. Neoplasia 2021; 23:488-501. [PMID: 33906087 PMCID: PMC8099723 DOI: 10.1016/j.neo.2021.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 11/17/2022]
Abstract
Although much is known about the gene mutations required to drive colorectal cancer (CRC) initiation, the tissue-specific selective microenvironments in which neoplasia arises remains less characterized. Here, we determined whether modulation of intestinal stem cell niche morphogens alone can exert a neoplasia-relevant selective pressure on normal colonic epithelium. Using adult stem cell-derived murine colonic epithelial organoids (colonoids), we employed a strategy of sustained withdrawal of epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) inhibition to select for and expand survivors. EGFR-signaling-independent (iEGFR) colonoids emerged over rounds of selection and expansion. Colonoids derived from a mouse model of chronic mucosal injury showed an enhanced ability to adapt to EGFR inhibition. Whole-exome and transcriptomic analyses of iEGFR colonoids demonstrated acquisition of deleterious mutations and altered expression of genes implicated in EGF signaling, pyroptosis, and CRC. iEGFR colonoids acquired dysplasia-associated cytomorphologic changes, an increased proliferative rate, and the ability to survive independently of other required niche factors. These changes were accompanied by emergence of aneuploidy and chromosomal instability; further, the observed mitotic segregation errors were significantly associated with loss of interkinetic nuclear migration, a fundamental and dynamic process underlying intestinal epithelial homeostasis. This study provides key evidence that chromosomal instability and other phenotypes associated with neoplasia can be induced ex vivo via adaptation to EGF withdrawal in normal and stably euploid colonic epithelium, without introducing cancer-associated driver mutations. In addition, prior mucosal injury accelerates this evolutionary process.
Collapse
Affiliation(s)
- Tiane Chen
- Department of Pathology, Division of GI/Liver Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA; Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Maged Zeineldin
- Department of Pathology, Division of GI/Liver Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA; Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Blake A Johnson
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD USA; Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Yi Dong
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Akshay Narkar
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Taibo Li
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Jin Zhu
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Rong Li
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD USA; Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, Singapore
| | - Tatianna C Larman
- Department of Pathology, Division of GI/Liver Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA; Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD USA.
| |
Collapse
|
388
|
Oxyresveratrol Ameliorates Dextran Sulfate Sodium-Induced Colitis in Rats by Suppressing Inflammation. Molecules 2021; 26:molecules26092630. [PMID: 33946346 PMCID: PMC8124641 DOI: 10.3390/molecules26092630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Colitis causes destruction of the intestinal mucus layer and increases intestinal inflammation. The use of antioxidants and anti-inflammatory agents derived from natural sources has been recently highlighted as a new approach for the treatment of colitis. Oxyresveratrol (OXY) is an antioxidant known to have various beneficial effects on human health, such as anti-inflammatory, antibacterial activity, and antiviral activity. The aim of this study was to investigate the therapeutic effect of OXY in rats with dextran sulfate sodium (DSS)-induced acute colitis. OXY ameliorated DSS-induced colitis and repaired damaged intestinal mucosa. OXY downregulated the expression of pro-inflammatory cytokine genes (TNF-α, IL-6, and IL-1β) and chemokine gene MCP-1, while promoting the production of anti-inflammatory cytokine IL-10. OXY treatment also suppressed inflammation via inhibiting cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expression in the colon, as well as the activity of myeloperoxidase (MPO). OXY exhibited anti-apoptotic effects, shifting the Bax/Bcl-2 balance. In conclusion, OXY might improve DSS-induced colitis by restoring the intestinal mucus layer and reducing inflammation within the intestine.
Collapse
|
389
|
Fabisik M, Tureckova J, Pavliuchenko N, Kralova J, Balounova J, Vicikova K, Skopcova T, Spoutil F, Pokorna J, Angelisova P, Malissen B, Prochazka J, Sedlacek R, Brdicka T. Regulation of Inflammatory Response by Transmembrane Adaptor Protein LST1. Front Immunol 2021; 12:618332. [PMID: 33986741 PMCID: PMC8111073 DOI: 10.3389/fimmu.2021.618332] [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: 10/16/2020] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
LST1 is a small adaptor protein expressed in leukocytes of myeloid lineage. Due to the binding to protein tyrosine phosphatases SHP1 and SHP2 it was thought to have negative regulatory function in leukocyte signaling. It was also shown to be involved in cytoskeleton regulation and generation of tunneling nanotubes. LST1 gene is located in MHCIII locus close to many immunologically relevant genes. In addition, its expression increases under inflammatory conditions such as viral infection, rheumatoid arthritis and inflammatory bowel disease and its deficiency was shown to result in slightly increased sensitivity to influenza infection in mice. However, little else is known about its role in the immune system homeostasis and immune response. Here we show that similar to humans, LST1 is expressed in mice in the cells of the myeloid lineage. In vivo, its deficiency results in alterations in multiple leukocyte subset abundance in steady state and under inflammatory conditions. Moreover, LST1-deficient mice show significant level of resistance to dextran sodium sulphate (DSS) induced acute colitis, a model of inflammatory bowel disease. These data demonstrate that LST1 regulates leukocyte abundance in lymphoid organs and inflammatory response in the gut.
Collapse
Affiliation(s)
- Matej Fabisik
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia.,Faculty of Science, Charles University, Prague, Czechia
| | - Jolana Tureckova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Nataliia Pavliuchenko
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia.,Faculty of Science, Charles University, Prague, Czechia
| | - Jarmila Kralova
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Balounova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Kristina Vicikova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Tereza Skopcova
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Jana Pokorna
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Pavla Angelisova
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Bernard Malissen
- Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Jan Prochazka
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czechia
| | - Tomas Brdicka
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
390
|
Pathak S, Gokhroo A, Kumar Dubey A, Majumdar S, Gupta S, Almeida A, Mahajan GB, Kate A, Mishra P, Sharma R, Kumar S, Vishwakarma R, Balakrishnan A, Atreya H, Nandi D. 7-Hydroxy Frullanolide, a sesquiterpene lactone, increases intracellular calcium amounts, lowers CD4 + T cell and macrophage responses, and ameliorates DSS-induced colitis. Int Immunopharmacol 2021; 97:107655. [PMID: 33901737 DOI: 10.1016/j.intimp.2021.107655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/19/2021] [Accepted: 04/03/2021] [Indexed: 12/16/2022]
Abstract
Sesquiterpene lactones are a class of anti-inflammatory molecules obtained from plants belonging to the Asteraceae family. In this study, the effects of 7-hydroxy frullanolide (7HF), a sesquiterpene lactone, in inhibiting CD4+ T cell and peritoneal macrophage responses were investigated. 7HF, in a dose dependent manner, lowers CD69 upregulation, IL2 production and CD4+ T cell cycling upon activation with the combination of anti-CD3 and anti-CD28. Further mechanistic studies demonstrated that 7HF, at early time points, increases intracellular Ca2+ amounts, over and above the levels induced upon activation. The functional relevance of 7HF-induced Ca2+ increase was confirmed using sub-optimal amounts of BAPTA, an intracellular Ca2+ chelator, which lowers lactate and rescues CD4+ T cell cycling. In addition, 7HF lowers T cell cycling with the combination of PMA and Ionomycin. However, 7HF increases CD4+ T cell cycling with sub-optimal activating signals: only PMA or anti-CD3. Furthermore, LPS-induced nitrite and IL6 production by peritoneal macrophages is inhibited by 7HF in a Ca2+-dependent manner. Studies with Ca2+ channel inhibitors, Ruthenium Red and 2-Aminoethoxydiphenyl borate, lowers the inhibitory effects of 7HF on CD4+ T cell and macrophage responses. In silico studies demonstrated that 7HF binds to Ca2+ channels, TRPV1, IP3R and SERCA, which is mechanistically important. Finally, intraperitoneal administration of 7HF lowers serum inflammatory cytokines, IFNγ and IL6, and reduces the effects of DSS-induced colitis with respect to colon length and colon damage. Overall, this study sheds mechanistic light on the anti-inflammatory potential of 7HF, a natural plant compound, in lowering immune responses.
Collapse
Affiliation(s)
- Sanmoy Pathak
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Abhijeet Gokhroo
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Ashim Kumar Dubey
- Undergraduate Program, Indian Institute of Science, Bangalore 560012, India
| | - Shamik Majumdar
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Souradeep Gupta
- NMR Research Facility, Indian Institute of Science, Bangalore 560012, India
| | - Asha Almeida
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Girish B Mahajan
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Abhijeet Kate
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Prabhu Mishra
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Rajiv Sharma
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Sanjay Kumar
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Ram Vishwakarma
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Arun Balakrishnan
- High Throughput Department, Piramal Research Center,1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai 400063, India
| | - Hanudatta Atreya
- NMR Research Facility, Indian Institute of Science, Bangalore 560012, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
391
|
Xu F, Cheng Y, Ruan G, Fan L, Tian Y, Xiao Z, Chen D, Wei Y. New pathway ameliorating ulcerative colitis: focus on Roseburia intestinalis and the gut-brain axis. Therap Adv Gastroenterol 2021; 14:17562848211004469. [PMID: 33948112 PMCID: PMC8053823 DOI: 10.1177/17562848211004469] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The community of gut microbes is a key factor controlling the intestinal barrier that communicates with the nervous system through the gut-brain axis. Based on our clinical data showing that populations of Roseburia intestinalis are dramatically decreased in the gut of patients with ulcerative colitis, we studied the efficacy of a strain belonging to this species in the context of colitis and stress using animal models. METHODS Dextran sulfate sodium was used to induce colitis in rats, which then underwent an enema with R. intestinalis as a treatment. The disease activity index, fecal changes and body weight of rats were recorded to evaluate colitis, while histological and immunohistochemical analyses were carried out to examine colon function, and 16S rRNA sequencing was performed to evaluate the gut microbiota change. Behavioral assays and immunohistochemical staining of brain were performed to assess the effect of R. intestinalis on the gut-brain axis. RESULTS Colitis-related symptoms in rats were significantly relieved after R. intestinalis enema, and the stool traits and colon length of rats were significantly recovered after treatment. The gut epithelial integrity and intestinal barrier were restored in treated rats, as evidenced by the higher expression of Zo-1 in colon tissues, accompanied by the restoration of gut microbiota. Meanwhile, depressive-like behaviors of rats were reduced after treatment, and laboratory experiments on neuronal cells also showed that IL-6, IL-7 and 5-HT were downregulated by R. intestinalis treatment in both serum and brain tissue, while Iba-1 expression was reduced in treated rats. CONCLUSIONS The administration of R. intestinalis contributes to restoration of the gut microbiota, promoting colon repair and the recovery of gastrointestinal function. These alterations are accompanied by the relief of depressive-like behaviors through a process modulated by the neuronal network and the regulation of inflammation by the gut-brain axis.
Collapse
Affiliation(s)
- Fenghua Xu
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Yi Cheng
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Guangcong Ruan
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Liqin Fan
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Yuting Tian
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Zhifeng Xiao
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Army Medical Center of PLA Affiliated with Army Medical University, Chongqing, China
| | | |
Collapse
|
392
|
Jin J, Jung IH, Moon SH, Jeon S, Jeong SJ, Sonn SK, Seo S, Lee MN, Song EJ, Kweon HY, Kim S, Kim TK, Kim J, Cho HR, Choi JH, Kwon B, Oh GT. CD137 Signaling Regulates Acute Colitis via RALDH2-Expressing CD11b -CD103 + DCs. Cell Rep 2021; 30:4124-4136.e5. [PMID: 32209473 DOI: 10.1016/j.celrep.2020.02.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/21/2019] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
CD137, a potent costimulatory receptor for CD8+ T cells, is expressed in various non-T cells, but little is known about its regulatory functions in these cells. In this study, we show that CD137 signaling, specifically in intestinal CD11b-CD103+ dendritic cells (DCs), restricts acute colitis progression. Mechanistically, CD137 engagement activates TAK1 and subsequently stimulates the AMPK-PGC-1α axis to enhance expression of the Aldh1a2 gene encoding the retinoic acid (RA) metabolizing enzyme RALDH2. RA can act on CD11b+CD103- DCs and induce SOCS3 expression, which, in turn, suppresses p38MAPK activation and interleukin-23 (IL-23) production. Administration of RA in DC-specific CD137-/- mice represses IL-23-producing CD11b+CD103- DCs and TH17 cells, indicating that RA is a major inhibitory effector molecule against intestinal CD11b+CD103- DCs. Additionally, the therapeutic effect of the anti-CD137 antibody is abrogated in DC-specific CD137-/- mice. Taken together, our results define a mechanism of paracrine immunoregulation operating between adjacent DC subsets in the intestine.
Collapse
Affiliation(s)
- Jing Jin
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - In-Hyuk Jung
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shin Hye Moon
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Sejin Jeon
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Seong-Keun Sonn
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Seungwoon Seo
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Mi-Ni Lee
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Eun Ju Song
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Hyae Yon Kweon
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Sinai Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Tae Kyeong Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Juyang Kim
- School of Biological Sciences and Biomedical Research Center, University of Ulsan, Ulsan 44610, South Korea
| | - Hong Rae Cho
- Department of Surgery and Biomedical Research Center, Ulsan University Hospital, College of Medicine, University of Ulsan, Ulsan 44610, South Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, South Korea
| | - Byungsuk Kwon
- School of Biological Sciences and Biomedical Research Center, University of Ulsan, Ulsan 44610, South Korea.
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 120-750, South Korea.
| |
Collapse
|
393
|
Ohya S, Matsui M, Kajikuri J, Endo K, Kito H. Increased Interleukin-10 Expression by the Inhibition of Ca 2+-Activated K + Channel K Ca3.1 in CD4 +CD25 + Regulatory T Cells in the Recovery Phase in an Inflammatory Bowel Disease Mouse Model. J Pharmacol Exp Ther 2021; 377:75-85. [PMID: 33504590 DOI: 10.1124/jpet.120.000395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract arising from abnormal responses of the innate and adaptative immune systems. Interleukin (IL)-10-producing CD4+CD25+ regulatory T (Treg) cells play a protective role in the recovery phase of IBD. In the present study, the effects of the administration of the selective Ca2+-activated K+ channel KCa3.1 inhibitor TRAM-34 on disease activities were examined in chemically induced IBD model mice. IBD disease severity, as assessed by diarrhea, visible fecal blood, inflammation, and crypt damage in the colon, was significantly lower in mice administered 1 mg/kg TRAM-34 than in vehicle-administered mice. Quantitative real-time polymerase chain reaction examinations showed that IL-10 expression levels in the recovery phase were markedly increased by the inhibition of KCa3.1 in mesenteric lymph node (mLN) Treg cells of IBD model mice compared with vehicle-administered mice. Among several positive and negative transcriptional regulators (TRs) for IL-10, three positive TRs-E4BP4, KLF4, and Blimp1-were upregulated by the inhibition of KCa3.1 in the mLN Treg cells of IBD model mice. In mouse peripheral CD4+CD25+ Treg cells induced by lectin stimulation, IL-10 expression and secretion were enhanced by the treatment with TRAM-34, together with the upregulation of E4BP4, KLF4, and Blimp1. Collectively, the present results demonstrated that the pharmacological inhibition of KCa3.1 decreased IBD symptoms in the IBD model by increasing IL-10 production in peripheral Treg cells and that IL-10high Treg cells produced by the treatment with KCa3.1 inhibitor may contribute to efficient Treg therapy for chronic inflammatory disorders, including IBD. SIGNIFICANCE STATEMENT: Pharmacological inhibition of Ca2+-activated K+ channel KCa3.1 increased IL-10 expression in peripheral Treg cells, together with the upregulation of the transcriptional regulators of IL-10: Krüppel-like factor 4, E4 promoter-binding protein 4, and/or B lymphocyte-induced maturation protein 1. The manipulation of IL-10high-producing Treg cells by the pharmacological inhibition of KCa3.1 may be beneficial in the treatment of chronic inflammatory diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Miki Matsui
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kyoko Endo
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
394
|
Zhang C, Yue H, Sun P, Hua L, Liang S, Ou Y, Wu D, Wu X, Chen H, Hao Y, Hu W, Yang Z. Discovery of chalcone analogues as novel NLRP3 inflammasome inhibitors with potent anti-inflammation activities. Eur J Med Chem 2021; 219:113417. [PMID: 33845232 DOI: 10.1016/j.ejmech.2021.113417] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
NLRP3 inflammasome activation plays a critical role in inflammation and its related disorders. Herein we report a hit-to-lead effort resulting in the discovery of a novel and potent class of NLRP3 inflammasome inhibitors. Among these, the most potent lead 40 exhibited improved inhibitory potency and almost no toxicity. Further mechanistic study indicated that compound 40 inhibited the NLRP3 inflammasome activation via suppressing ROS production. More importantly, treatment with 40 showed remarkable therapeutic effects on LPS-induced sepsis and DSS-induced colitis. This study encourages further development of more potent inhibitors based on this chemical scaffold and provides a chemical tool to identify its cellular binding target.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Hu Yue
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Lei Hua
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Shuli Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Yitao Ou
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Dan Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Xinyi Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Hao Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Ying Hao
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Wenhui Hu
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Zhongjin Yang
- Key Laboratory of Molecular Target & Clinical Pharmacology and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| |
Collapse
|
395
|
Zhuang H, Lv Q, Zhong C, Cui Y, He L, Zhang C, Yu J. Tiliroside Ameliorates Ulcerative Colitis by Restoring the M1/M2 Macrophage Balance via the HIF-1α/glycolysis Pathway. Front Immunol 2021; 12:649463. [PMID: 33868286 PMCID: PMC8044352 DOI: 10.3389/fimmu.2021.649463] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
Macrophages polarized to different phenotypes critically contribute to colitis development by coordinating inflammatory and anti-inflammatory processes. Herein, targeting the balance between the pro-inflammatory M1 and the anti-inflammatory M2 macrophage phenotypes can be a novel therapeutic approach for colitis. In the present study, we firstly demonstrated that tiliroside possessed the ability to alleviate the clinical symptoms of colitis as evidenced by decreased disease activity index (DAI) scores, longer colon length, reduced myeloperoxidase (MPO) activity, and improvement of colonic pathological damage in vivo. Furthermore, we showed that tiliroside modulated the balance between M1 and M2 macrophages toward a more anti-inflammatory status in colonic lamina propria but has little effect on the T cell population and epithelial barrier function in colitis mice. The macrophage depletion study further showed the protective effect of tiliroside was macrophage dependent in vivo. Mechanistically, our study demonstrated that tiliroside regulated cellular metabolism by inhibiting aerobic glycolysis in LPS and IFNγ stimulated macrophages. At the molecular level, tiliroside facilitated the proteasomal degradation of HIF-1α and downregulated mRNA expressions of HIF-1α dependent glycolytic enzymes in macrophages. Collectively, our data highlight the aberrant M1/M2 macrophage polarization in the initiation and development of ulcerative colitis and put forth the stage for considering tiliroside as a metabolic regulator in reprogramming macrophage polarization, which may serve as a promising therapeutic approach for treatment of inflammation-associated and metabolic disorders.
Collapse
Affiliation(s)
- Hongda Zhuang
- Translational Medicine Centre, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qi Lv
- Translational Medicine Centre, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Stake Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Zhong
- Translational Medicine Centre, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yaru Cui
- Translational Medicine Centre, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Luling He
- Translational Medicine Centre, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Cheng Zhang
- Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Jun Yu
- Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
396
|
Biological functions of NLRP3 inflammasome: A therapeutic target in inflammatory bowel disease. Cytokine Growth Factor Rev 2021; 60:61-75. [PMID: 33773897 DOI: 10.1016/j.cytogfr.2021.03.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022]
Abstract
Cases of inflammatory bowel disease (IBD), a debilitating intestinal disorder with complex pathological mechanisms, have been increasing in recent years, straining the capacity of healthcare systems. Thus, novel therapeutic targets and innovative agents must be developed. Notably, the NLRP3 inflammasome is upregulated in patients with IBD and/or in animal experimental models. As an innate immune supramolecular assembly, the NLRP3 inflammasome is persistently activated during the pathogenesis of IBD by multiple stimuli. Moreover, this protein complex regulates pro-inflammatory cytokines. Thus, targeting this multiprotein oligomer may offer a feasible way to relieve IBD symptoms and improve clinical outcomes. The mechanisms by which the NLRP3 inflammasome is activated, its role in IBD pathogenesis, and the drugs administered to target this protein complex are reviewed herein. This review establishes that the use of inflammasome-targeting drugs are effective for IBD treatment. Moreover, this review suggests that the value and potential of naturally sourced or derived medicines for IBD treatment must be recognized and appreciated.
Collapse
|
397
|
Zhou W, Zhang H, Pan Y, Xu Y, Cao Y. circRNA expression profiling of colon tissue from mesalazine-treated mouse of inflammatory bowel disease reveals an important circRNA-miRNA-mRNA pathway. Aging (Albany NY) 2021; 13:10187-10207. [PMID: 33819198 PMCID: PMC8064189 DOI: 10.18632/aging.202780] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Mesalazine (5-aminosalicylic acid, 5-ASA) has been widely used to treat inflammatory bowel disease (IBD). However, it remains unclear about the underlying biological mechanisms of IBD pathogenesis and mesalazine treatment, which could be partially clarified by exploring the profiling of circular RNAs (circRNAs) using RNA-seq. A total of 15 mice (C57BL/6) were randomly assigned to three equally sized groups: control, dextran sulfate sodium (DSS, using DSS to induce IBD), and DSS+5-ASA (using mesalazine to treat IBD). We randomly selected three mice of each group to collect colon tissues for RNA-seq and then performed bioinformatic analysis for two comparisons: DSS vs. control and DSS+5-ASA vs. DSS. Comparisons of a series of indicators (e.g., body weight) verified the establishment of DSS-induced IBD mouse model and the effectiveness of mesalazine in treating IBD. We identified 182 differentially expressed circRNAs, including 55 up-regulated and 47 down-regulated circRNAs when comparing the DSS+5-ASA with the DSS group. These 102 circRNA-associated genes were significantly involved in the N-Glycan biosynthesis and lysine degradation. The network analysis of circRNA-miRNA-mRNAs identified an important pathway, i.e., chr10:115386962-115390436+/mmu-miR-6914-5p/Atg7, which is related to autophagy. The findings provide new insights into the biological mechanisms of IBD pathogenesis and mesalazine treatment, particularly highlighting the circRNA-miRNA-mRNA pathway.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Anal-Rectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510, United States of America
| | - Haiyin Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yibin Pan
- Department of Anal-Rectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanwu Xu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongqing Cao
- Department of Anal-Rectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
398
|
Memon A, Kim BY, Kim SE, Pyao Y, Lee YG, Kang SC, Lee WK. Anti-Inflammatory Effect of Phytoncide in an Animal Model of Gastrointestinal Inflammation. Molecules 2021; 26:molecules26071895. [PMID: 33810618 PMCID: PMC8037037 DOI: 10.3390/molecules26071895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Phytoncide is known to have antimicrobial and anti-inflammatory properties. Purpose: This study was carried out to confirm the anti-inflammatory activity of two types of phytoncide extracts from pinecone waste. Methods: We made two types of animal models to evaluate the efficacy, an indomethacin-induced gastroenteritis rat model and a dextran sulfate sodium-induced colitis mouse model. Result: In the gastroenteritis experiment, the expression of induced-nitric oxide synthase (iNOS), a marker for inflammation, decreased in the phytoncide-supplemented groups, and gastric ulcer development was significantly inhibited (p < 0.05). In the colitis experiment, the shortening of the colon length and the iNOS expression were significantly suppressed in the phytoncide-supplemented group (p < 0.05). Conclusions: Through this study, we confirmed that phytoncide can directly inhibit inflammation in digestive organs. Although further research is needed, we conclude that phytoncide has potential anti-inflammatory properties in the digestive tract and can be developed as a functional agent.
Collapse
Affiliation(s)
- Azra Memon
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea; (A.M.); (Y.P.)
| | - Bae Yong Kim
- Research Institute, Phylus Co., LTD., Danyang-gun 27000, Korea;
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Korea; (S.-e.K.); (Y.-G.L.); (S.C.K.)
| | - Se-eun Kim
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Korea; (S.-e.K.); (Y.-G.L.); (S.C.K.)
| | - Yuliya Pyao
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea; (A.M.); (Y.P.)
| | - Yeong-Geun Lee
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Korea; (S.-e.K.); (Y.-G.L.); (S.C.K.)
| | - Se Chan Kang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Korea; (S.-e.K.); (Y.-G.L.); (S.C.K.)
| | - Woon Kyu Lee
- Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea; (A.M.); (Y.P.)
- Correspondence: ; Tel.: +82-10-4607-3871
| |
Collapse
|
399
|
Labarta-Bajo L, Nilsen SP, Humphrey G, Schwartz T, Sanders K, Swafford A, Knight R, Turner JR, Zúñiga EI. Type I IFNs and CD8 T cells increase intestinal barrier permeability after chronic viral infection. J Exp Med 2021; 217:152069. [PMID: 32880630 PMCID: PMC7953738 DOI: 10.1084/jem.20192276] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/29/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Intestinal barrier leakage constitutes a potential therapeutic target for many inflammatory diseases and represents a disease progression marker during chronic viral infections. However, the causes of altered gut barrier remain mostly unknown. Using murine infection with lymphocytic choriomeningitis virus, we demonstrate that, in contrast to an acute viral strain, a persistent viral isolate leads to long-term viral replication in hematopoietic and mesenchymal cells, but not epithelial cells (IECs), in the intestine. Viral persistence drove sustained intestinal epithelial barrier leakage, which was characterized by increased paracellular flux of small molecules and was associated with enhanced colitis susceptibility. Type I IFN signaling caused tight junction dysregulation in IECs, promoted gut microbiome shifts and enhanced intestinal CD8 T cell responses. Notably, both type I IFN receptor blockade and CD8 T cell depletion prevented infection-induced barrier leakage. Our study demonstrates that infection with a virus that persistently replicates in the intestinal mucosa increases epithelial barrier permeability and reveals type I IFNs and CD8 T cells as causative factors of intestinal leakage during chronic infections.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| | - Steven P Nilsen
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Gregory Humphrey
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Tara Schwartz
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Karenina Sanders
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Austin Swafford
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, La Jolla, CA.,Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA.,Department of Bioengineering, University of California, San Diego, La Jolla, CA.,Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Elina I Zúñiga
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| |
Collapse
|
400
|
Raffner Basson A, Gomez-Nguyen A, LaSalla A, Buttó L, Kulpins D, Warner A, Di Martino L, Ponzani G, Osme A, Rodriguez-Palacios A, Cominelli F. Replacing Animal Protein with Soy-Pea Protein in an "American Diet" Controls Murine Crohn Disease-Like Ileitis Regardless of Firmicutes: Bacteroidetes Ratio. J Nutr 2021; 151:579-590. [PMID: 33484150 PMCID: PMC7948210 DOI: 10.1093/jn/nxaa386] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The current nutritional composition of the "American diet" (AD; also known as Western diet) has been linked to the increasing incidence of chronic diseases, including inflammatory bowel disease (IBD), namely Crohn disease (CD). OBJECTIVES This study investigated which of the 3 major macronutrients (protein, fat, carbohydrates) in the AD has the greatest impact on preventing chronic inflammation in experimental IBD mouse models. METHODS We compared 5 rodent diets designed to mirror the 2011-2012 "What We Eat in America" NHANES. Each diet had 1 macronutrient dietary source replaced. The formulated diets were AD, AD-soy-pea (animal protein replaced by soy + pea protein), AD-CHO ("refined carbohydrate" by polysaccharides), AD-fat [redistribution of the ω-6:ω-3 (n-6:n-3) PUFA ratio; ∼10:1 to 1:1], and AD-mix (all 3 "healthier" macronutrients combined). In 3 separate experiments, 8-wk-old germ-free SAMP1/YitFC mice (SAMP) colonized with human gut microbiota ("hGF-SAMP") from CD or healthy donors were fed an AD, an AD-"modified," or laboratory rodent diet for 24 wk. Two subsequent dextran sodium sulfate-colitis experiments in hGF-SAMP (12-wk-old) and specific-pathogen-free (SPF) C57BL/6 (20-wk-old) mice, and a 6-wk feeding trial in 24-wk-old SPF SAMP were performed. Intestinal inflammation, gut metagenomics, and MS profiles were assessed. RESULTS The AD-soy-pea diet resulted in lower histology scores [mean ± SD (56.1% ± 20.7% reduction)] in all feeding trials and IBD mouse models than did other diets (P < 0.05). Compared with the AD, the AD-soy-pea correlated with increased abundance in Lactobacillaceae and Leuconostraceae (1.5-4.7 log2 and 3.0-5.1 log2 difference, respectively), glutamine (6.5 ± 0.8 compared with 3.9 ± 0.3 ng/μg stool, P = 0.0005) and butyric acid (4:0; 3.3 ± 0.5 compared with 2.54 ± 0.4 ng/μg stool, P = 0.006) concentrations, and decreased linoleic acid (18:2n-6; 5.4 ± 0.4 compared with 8.6 ± 0.3 ng/μL plasma, P = 0.01). CONCLUSIONS Replacement of animal protein in an AD by plant-based sources reduced the severity of experimental IBD in all mouse models studied, suggesting that similar, feasible adjustments to the daily human diet could help control/prevent IBD in humans.
Collapse
Affiliation(s)
- Abigail Raffner Basson
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Adrian Gomez-Nguyen
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexandria LaSalla
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ludovica Buttó
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Danielle Kulpins
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alexandra Warner
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Luca Di Martino
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Gina Ponzani
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Abdullah Osme
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexander Rodriguez-Palacios
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Fabio Cominelli
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|