1
|
Bhat MA, Usman I, Dhaneshwar S. Application of Drug Repurposing Approach for Therapeutic Intervention of Inflammatory Bowel Disease. Curr Rev Clin Exp Pharmacol 2024; 19:234-249. [PMID: 37859409 DOI: 10.2174/0127724328245156231008154045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/07/2023] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
Inflammatory bowel disease (IBD), represented by Crohn's disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the gastrointestinal tract (GIT) characterized by chronic relapsing intestinal inflammation, abdominal pain, cramping, loss of appetite, fatigue, diarrhoea, and weight loss. Although the etiology of IBD remains unclear, it is believed to be an interaction between genes, and environmental factors, such as an imbalance of the intestinal microbiota, changing food habits, an ultra-hygiene environment, and an inappropriate immune system. The development of novel effective therapies is stymied by a lack of understanding of the aetiology of IBD. The current therapy involves the use of aminosalicylates, immunosuppressants, and corticosteroids that can effectively manage symptoms, induce and sustain remission, prevent complications, modify the course of the disease, provide diverse treatment options, showcase advancements in biologic therapies, and enhance the overall quality of life. However, the efficacy of current therapy is overshadowed by a plethora of adverse effects, such as loss of weight, mood swings, skin issues, loss of bone density, higher vulnerability to infections, and elevated blood pressure. Biologicals, like anti-tumour necrosis factor agents, can stimulate an autoimmune response in certain individuals that may diminish the effectiveness of the medication over time, necessitating a switch to alternative treatments. The response of IBD patients to current drug therapy is quite varied, which can lead to disease flares that underlines the urgent need to explore alternative treatment option to address the unmet need of developing new treatment strategies for IBD with high efficacy and fewer adverse effects. Drug repurposing is a novel strategy where existing drugs that have already been validated safe in patients for the management of certain diseases are redeployed to treat other, unindicated diseases. The present narrative review focuses on potential drug candidates that could be repurposed for the management of IBD using on-target and off-target strategies. It covers their preclinical, clinical assessment, mechanism of action, and safety profiles, and forecasts their appropriateness in the management of IBD. The review presents useful insights into the most promising candidates for repurposing, like anti-inflammatory and anti-apoptotic troxerutin, which has been found to improve the DSS-induced colitis in rats, an antiosteoarthritic drug diacetylrhein that has been found to have remarkable ameliorating effects on DSS-induced colitis via anti-oxidant and anti- inflammatory properties and by influencing both apoptosis and pyroptosis. Topiramate, an antiepileptic and anticonvulsant drug, has remarkably decreased overall pathophysiological and histopathological events in the experimental model of IBD in rodents by its cytokine inhibitory action.
Collapse
Affiliation(s)
- Mohammad Aadil Bhat
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, UP, Noida, India
| | - Iqra Usman
- Department of Pharmacology, Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, UP, Noida, India
| | - Suneela Dhaneshwar
- Department of Pharmaceutical Chemistry, Amity Institute of Pharmacy, Amity University Maharashtra, Mumbai, Maharashtra, India
| |
Collapse
|
2
|
Lee JY, Ma HW, Kim JH, Park IS, Son M, Ryu KH, Shin J, Kim SW, Cheon JH. Novel Histone Deacetylase 6 Inhibitor Confers Anti-inflammatory Effects and Enhances Gut Barrier Function. Gut Liver 2023; 17:766-776. [PMID: 36167345 PMCID: PMC10502503 DOI: 10.5009/gnl220159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/07/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022] Open
Abstract
Background/Aims The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function. Methods Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay. Results Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner. Conclusions The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.
Collapse
Affiliation(s)
- Jae-Young Lee
- Department of Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Mijeong Son
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Keun Ho Ryu
- Department of Non-Clinical Study, CKD Research Institute, CKD Pharmaceutical Co., Yongin, Korea
| | - Jieun Shin
- Department of Non-Clinical Study, CKD Research Institute, CKD Pharmaceutical Co., Yongin, Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Ding R, Ma Y, Li T, Sun M, Sun Z, Duan J. The detrimental effects of micro-and nano-plastics on digestive system: An overview of oxidative stress-related adverse outcome pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:163144. [PMID: 37003332 DOI: 10.1016/j.scitotenv.2023.163144] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 05/13/2023]
Abstract
With the massive manufacture and use of plastics, plastic pollution-related environmental impacts have raised great concern in recent years. As byproducts of plastic fragmentation and degradation, microplastics (MPs) and nanoplastics (NPs) have been identified as novel pollutants that posed a threat to the ecosystem and humans. Since MPs/NPs could be transported via the food chain and retained in the water, the digestive system should be one of the major targets of MPs/NPs-related toxicity. Although considerable evidence has supported the digestive toxicity of MPs/NPs, the proposed mechanisms remained ambiguous due to the variety of study types, models, and endpoints. This review provided a mechanism-based perspective on MPs/NPs-induced digestive effects by adopting the adverse outcome pathway framework as a promising tool. The overproduction of reactive oxygen species was identified as the molecular initiating event in MPs/NPs-mediated injury to the digestive system. A series of detrimental effects including oxidative stress, apoptosis, inflammation, dysbiosis, and metabolic disorders were summarized as key events. Finally, the occurrence of these effects eventually led to an adverse outcome, suggesting a possible increase in the incidence of digestive morbidity and mortality.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yiming Ma
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Mengqi Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
4
|
Zhao C, Han Y, Wang C, Ren M, Hu Q, Gu Y, Ye P, Li S, Jin E. Transcriptome Profiling of Duodenum Reveals the Importance of Boron Supplementation in Modulating Immune Activities in Rats. Biol Trace Elem Res 2022; 200:3762-3773. [PMID: 34773147 DOI: 10.1007/s12011-021-02983-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022]
Abstract
As an essential trace element, appropriate boron supplementation can promote immune function of animals. To illustrate the effects of boron in a rat model, RNA-Seq was conducted for the RNA from duodenum after treatment with different concentration of boron in which boron was given in the form of boric acid. More than 47 million reads were obtained in 0, 10, and 320 mg/L boron (0, 57.21, and 1830.66 mg/L boric acid) treatment groups that produced 58 965 402, 48 607 328, and 46 760 660 clean reads, respectively. More than 95% of the clean reads were successfully matched to the rat reference genome and assembled to generate 32 662 transcripts. A total of 624 and 391 differentially expressed candidate genes (DEGs) were found between 0 vs.10 and 0 vs. 320 mg/L boron comparison groups. We also identified transcription start site, transcription terminal site, and skipped exons as the main alternative splicing events. GO annotations revealed most of DEGs were involved in the regulation of immune activity. The DEGs were enriched in influenza A, herpes simplex infection, cytosolic DNA-sensing pathway, and antigen processing and presentation signaling pathways. The expression levels of genes enriched in these signaling pathways indicate that lower doses of boron could achieve better effects on promoting immune response in the duodenum. These effects on the immune system appear to be mediated via altering the expression patterns of genes involved in the related signaling pathways in a dose-dependent pattern. These data provide more insights into the molecular mechanisms of immune regulation in rats in response to dietary boron treatment.
Collapse
Affiliation(s)
- Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Yujiao Han
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Chenfang Wang
- College of Life and Health Science, Anhui Science and Technology University, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Man Ren
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Qianqian Hu
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Pengfei Ye
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, No. 9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No. 9, Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| |
Collapse
|
5
|
El-Sherbiny M, Eisa NH, Abo El-Magd NF, Elsherbiny NM, Said E, Khodir AE. Anti-inflammatory/anti-apoptotic impact of betulin attenuates experimentally induced ulcerative colitis: An insight into TLR4/NF-kB/caspase signalling modulation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 88:103750. [PMID: 34597787 DOI: 10.1016/j.etap.2021.103750] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 06/13/2023]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease with limited therapeutic management approaches. The present study evaluated the potential therapeutic impact of betulin on acetic acid (AA)-induced UC in rats. UC was induced by intracolonic instillation of AA (3% v/v). Rats were treated with betulin (8 mg/kg, I.P., once daily) four days post AA instillation and for 14 consecutive days. Betulin attenuated AA-induced UC as evidenced by retracted macroscopic scores, serum CRP titre and LDH activity, attenuated histopathological hallmarks of UC including mucosal necrosis, haemorrhage, congestion and inflammatory cells infiltration. Moreover, betulin dampened UC-associated colonic inflammatory load with modulation of TLR4/NF-kB axis and reduction in colonic inflammatory cytokines; TNF-α, IL1β and IL-6. Nevertheless, betulin suppressed colonic apoptosis with reduced colonic caspase-3 and caspase-8 expression. The current findings confirm a beneficial therapeutic impact of betulin against UC. The prospective underlying mechanisms include down-regulation of TLR4/NF-κB and the subsequent downstream signalling pathways.
Collapse
Affiliation(s)
- Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, 71666, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Nada F Abo El-Magd
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Nehal M Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Ahmed E Khodir
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| |
Collapse
|
6
|
Liu T, Wang C, Wu X, Ren M, Hu Q, Jin E, Gu Y. Effect of Boron on Microstructure, Immune Function, Expression of Tight Junction Protein, Cell Proliferation and Apoptosis of Duodenum in Rats. Biol Trace Elem Res 2021; 199:205-215. [PMID: 32319072 DOI: 10.1007/s12011-020-02123-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/18/2020] [Indexed: 10/24/2022]
Abstract
Boron is an essential trace element for animals. Appropriate boron supplementation can produce beneficial effects on the animal body, while a high dose of boron has adverse and even toxic effects. Our aim was to investigate the impact of different doses of boron on the microstructure of duodenum in rats, expression of secretory immunoglobulin A (SIgA) and tight junction protein, cell proliferation and apoptosis. Eighty newly weaned clean Sprague-Dawley (SD) rats were given distilled water supplemented with 0, 10, 20, 40, 80, 160, 320, and 640 mg/L of boron for 60 days. We found that supplementation of 40 and 80 mg/L boron could increase the height of duodenal villi and the crypt depth, the number of intraepithelial lymphocytes (IELs) and goblet cells, the expression of SIgA, Zonula occludens-1 (ZO-1) and occludin, and proliferating cell nuclear antigen (PCNA) in duodenum of rats; decrease expression of Caspase-3 mRNA and the number of Caspase-3-positive cells, but supplementation of 320 and 640 mg/L boron could have the opposite effect in these indexes. The results showed that supplemented with 40 and 80 mg/L of boron could improve the structure and function of duodenum, while supplemented with 320-640 mg/L had a significant inhibitory effect.
Collapse
Affiliation(s)
- Ting Liu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Chenfang Wang
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Xiaoshuang Wu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Man Ren
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Qianqian Hu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, China.
| |
Collapse
|
7
|
Solà-Tapias N, Vergnolle N, Denadai-Souza A, Barreau F. The Interplay Between Genetic Risk Factors and Proteolytic Dysregulation in the Pathophysiology of Inflammatory Bowel Disease. J Crohns Colitis 2020; 14:1149-1161. [PMID: 32090263 DOI: 10.1093/ecco-jcc/jjaa033] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Crohn's disease [CD] and ulcerative colitis [UC] are the two main forms of inflammatory bowel disease [IBD]. Previous studies reported increased levels of proteolytic activity in stool and tissue samples from IBD patients, whereas the re-establishment of the proteolytic balance abrogates the development of experimental colitis. Furthermore, recent data suggest that IBD occurs in genetically predisposed individuals who develop an abnormal immune response to intestinal microbes once exposed to environmental triggers. In this review, we highlight the role of proteases in IBD pathophysiology, and we showcase how the main cellular pathways associated with IBD influence proteolytic unbalance and how functional proteomics are allowing the unambiguous identification of dysregulated proteases in IBD, paving the way to the development of new protease inhibitors as a new potential treatment.
Collapse
Affiliation(s)
- Núria Solà-Tapias
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Alexandre Denadai-Souza
- Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Leuven, Belgium
| | - Frédérick Barreau
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| |
Collapse
|
8
|
Loss of Bcl-G, a Bcl-2 family member, augments the development of inflammation-associated colorectal cancer. Cell Death Differ 2019; 27:742-757. [PMID: 31296963 PMCID: PMC7206067 DOI: 10.1038/s41418-019-0383-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal epithelial cells provide a selective barrier that segregates the host immune system from luminal microorganisms, thereby contributing directly to the regulation of homeostasis. We have shown that from early embryonic development Bcl-G, a Bcl-2 protein family member with unknown function, was highly expressed in gastrointestinal epithelial cells. While Bcl-G was dispensable for normal growth and development in mice, the loss of Bcl-G resulted in accelerated progression of colitis-associated cancer. A label-free quantitative proteomics approach revealed that Bcl-G may contribute to the stability of a mucin network, which when disrupted, is linked to colon tumorigenesis. Consistent with this, we observed a significant reduction in Bcl-G expression in human colorectal tumors. Our study identifies an unappreciated role for Bcl-G in colon cancer.
Collapse
|
9
|
Zhou J, Wu LY, Chen L, Guo YJ, Sun Y, Li T, Zhao JM, Bao CH, Wu HG, Shi Y. Herbs-partitioned moxibustion alleviates aberrant intestinal epithelial cell apoptosis by upregulating A20 expression in a mouse model of Crohn’s disease. World J Gastroenterol 2019; 25:2071-2085. [PMID: 31114134 PMCID: PMC6506586 DOI: 10.3748/wjg.v25.i17.2071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A20 inhibits intestinal epithelial cell apoptosis in Crohn’s disease, and herbs-partitioned moxibustion (HPM) has been demonstrated to be an effective treatment for Crohn’s disease. However, the mechanism by which HPM reduces intestinal epithelial cell apoptosis in Crohn’s disease has not been thoroughly elucidated to date.
AIM To elucidate whether HPM exerts its effects by upregulating A20 to affect intestinal epithelial cell apoptosis in a Crohn’s disease mouse model.
METHODS In this study, mice with A20 deletion in intestinal epithelial cells (A20IEC-KO) were utilized to establish a Crohn’s disease mouse model with 2,4,6-trinitrobenzene sulfonic acid (TNBS) administration, as well as wild-type mice. Mice were randomly divided into normal control (NC), model control (MC), mesalazine (MESA), and HPM groups. The morphology of the colonic mucosa was observed by hematoxylin-eosin staining, and serum endotoxin and apoptosis of epithelial cells were evaluated by enzyme-linked immunosorbent assay and terminal dUTP nick-end labeling assay accordingly. The protein expression levels of A20 and tumor necrosis factor receptor 1 (TNFR1)-related signaling molecules were evaluated by Western blot, and co-expression of A20 and TNFR1-associated death domain (TRADD) and co-expression of A20 and receptor-interacting protein 1 (RIP1) were observed by double immunofluorescence staining.
RESULTS The intestinal epithelial barrier was noted to have an improvement in the HPM group of wild-type (WT) mice compared with that in A20IEC-KO mice. Compared with A20 IEC-KO HPM mice, serum endotoxin levels and apoptosis percentages were decreased (P < 0.01), A20 expression levels were increased (P < 0.01), and expression of TNFR1, TRADDD, and RIP1 was decreased in the HPM group of WT mice (PTNFR1 < 0.05, PTRADD < 0.01, PRIP1 < 0.01). Both of the co-expression of A20/TRADD and A20/RIP1 showed a predominantly yellow fluorescence in the HPM group of WT mice, while a predominantly red fluorescence was noted in the HPM group of A20IEC-KO mice.
CONCLUSION Our findings suggest that HPM in treating Crohn’s disease functions possibly via upregulation of the A20 expression level, resulting in downregulation of TNFR1, TRADD, and RIP1 to alleviate increased cell apoptosis in the intestinal epithelial barrier in Crohn's disease.
Collapse
Affiliation(s)
- Jing Zhou
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lu-Yi Wu
- Qigong Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Liu Chen
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ya-Jing Guo
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi Sun
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tao Li
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ji-Meng Zhao
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai Institute of Acupuncture-Moxibustion and Meridian, Shanghai 200030, China
| | - Chun-Hui Bao
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai Institute of Acupuncture-Moxibustion and Meridian, Shanghai 200030, China
| | - Huan-Gan Wu
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai Institute of Acupuncture-Moxibustion and Meridian, Shanghai 200030, China
| | - Yin Shi
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Outpatient Department, Shanghai Institute of Acupuncture-Moxibustion and Meridian, Shanghai 200030, China
| |
Collapse
|
10
|
Sébert M, Sola-Tapias N, Mas E, Barreau F, Ferrand A. Protease-Activated Receptors in the Intestine: Focus on Inflammation and Cancer. Front Endocrinol (Lausanne) 2019; 10:717. [PMID: 31708870 PMCID: PMC6821688 DOI: 10.3389/fendo.2019.00717] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
Protease-activated receptors (PARs) belong to the G protein-coupled receptor (GPCR) family. Compared to other GPCRs, the specificity of the four PARs is the lack of physiologically soluble ligands able to induce their activation. Indeed, PARs are physiologically activated after proteolytic cleavage of their N-terminal domain by proteases. The resulting N-terminal end becomes a tethered activation ligand that interact with the extracellular loop 2 domain and thus induce PAR signal. PARs expression is ubiquitous and these receptors have been largely described in chronic inflammatory diseases and cancer. In this review, after describing their discovery, structure, mechanisms of activation, we then focus on the roles of PARs in the intestine and the two main diseases affecting the organ, namely inflammatory bowel diseases and cancer.
Collapse
|
11
|
Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med 2018; 50:1-9. [PMID: 30115904 PMCID: PMC6095905 DOI: 10.1038/s12276-018-0126-x] [Citation(s) in RCA: 1051] [Impact Index Per Article: 150.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is a specialized organ in which dynamic interactions between host cells and the complex environment occur in addition to food digestion. Together with the chemical barrier of the mucosal layer and the cellular immune system, the epithelial cell layer performs a pivotal role as the first physical barrier against external factors and maintains a symbiotic relationship with commensal bacteria. The tight junction proteins, including occludin, claudins, and zonula occludens, are crucial for the maintenance of epithelial barrier integrity. To allow the transport of essential molecules and restrict harmful substances, the intracellular signaling transduction system and a number of extracellular stimuli such as cytokines, small GTPases, and post-translational modifications dynamically modulate the tight junction protein complexes. An imbalance in these regulations leads to compromised barrier integrity and is linked with pathological conditions. Despite the obscurity of the causal relationship, the loss of barrier integrity is considered to contribute to inflammatory bowel disease, obesity, and metabolic disorders. The elucidation of the role of diseases in barrier integrity and the underlying regulatory mechanisms have improved our understanding of the intestinal barrier to allow the development of novel and potent therapeutic approaches. A better understanding of how the cells that line the inside of the intestines allow nutrients in, while keeping harmful substances and pathogens out could lead to new therapies for inflammatory bowel disease, obesity, and other conditions. A team from South Korea led by Sung Ho Ryu from Pohang University of Science and Technology review the regulatory mechanisms that help maintain the intestinal epithelial barrier. They discuss the role of tight junction proteins in forming a seal between adjacent cells and the various signaling pathways that loosen or tighten these junctions to enable limited transport. Loss of barrier integrity because of genetics, gut microbes, auto-immunity, diet, or other factors is often implicated in disease, and restoring barrier function with drugs or probiotics could help ameliorate many health problems.
Collapse
|
12
|
Protective effect of cardamonin against acetic acid-induced ulcerative colitis in rats. Pharmacol Rep 2017; 69:268-275. [DOI: 10.1016/j.pharep.2016.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/06/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022]
|
13
|
Van Spaendonk H, Ceuleers H, Witters L, Patteet E, Joossens J, Augustyns K, Lambeir AM, De Meester I, De Man JG, De Winter BY. Regulation of intestinal permeability: The role of proteases. World J Gastroenterol 2017; 23:2106-2123. [PMID: 28405139 PMCID: PMC5374123 DOI: 10.3748/wjg.v23.i12.2106] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/20/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal barrier is - with approximately 400 m2 - the human body’s largest surface separating the external environment from the internal milieu. This barrier serves a dual function: permitting the absorption of nutrients, water and electrolytes on the one hand, while limiting host contact with noxious luminal antigens on the other hand. To maintain this selective barrier, junction protein complexes seal the intercellular space between adjacent epithelial cells and regulate the paracellular transport. Increased intestinal permeability is associated with and suggested as a player in the pathophysiology of various gastrointestinal and extra-intestinal diseases such as inflammatory bowel disease, celiac disease and type 1 diabetes. The gastrointestinal tract is exposed to high levels of endogenous and exogenous proteases, both in the lumen and in the mucosa. There is increasing evidence to suggest that a dysregulation of the protease/antiprotease balance in the gut contributes to epithelial damage and increased permeability. Excessive proteolysis leads to direct cleavage of intercellular junction proteins, or to opening of the junction proteins via activation of protease activated receptors. In addition, proteases regulate the activity and availability of cytokines and growth factors, which are also known modulators of intestinal permeability. This review aims at outlining the mechanisms by which proteases alter the intestinal permeability. More knowledge on the role of proteases in mucosal homeostasis and gastrointestinal barrier function will definitely contribute to the identification of new therapeutic targets for permeability-related diseases.
Collapse
|
14
|
Arya A, Majumdar DK, Pathak DP, Sharma AK, Ray AR. Design and evaluation of acrylate polymeric carriers for fabrication of pH-sensitive microparticles. Drug Dev Ind Pharm 2016; 43:305-318. [DOI: 10.1080/03639045.2016.1239629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amit Arya
- Delhi Institute of Pharmaceutical Sciences and Research, (formerly College of Pharmacy), University of Delhi, Pushp Vihar, Sector III, New Delhi, India
| | - Dipak K. Majumdar
- Delhi Institute of Pharmaceutical Sciences and Research, (formerly College of Pharmacy), University of Delhi, Pushp Vihar, Sector III, New Delhi, India
- School of Pharmaceutical Sciences, Apeejay Stya University, Sohna, Haryana, India
| | - Dharam Pal Pathak
- Delhi Institute of Pharmaceutical Sciences and Research, (formerly College of Pharmacy), University of Delhi, Pushp Vihar, Sector III, New Delhi, India
| | - Anil K. Sharma
- Delhi Institute of Pharmaceutical Sciences and Research, (formerly College of Pharmacy), University of Delhi, Pushp Vihar, Sector III, New Delhi, India
| | - Alok R. Ray
- Centre for Biomedical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi, India
| |
Collapse
|
15
|
The suppressor of cytokine signaling SOCS1 promotes apoptosis of intestinal epithelial cells via p53 signaling in Crohn's disease. Exp Mol Pathol 2016; 101:1-11. [PMID: 27236107 DOI: 10.1016/j.yexmp.2016.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/17/2022]
|
16
|
γ-Glutamyl valine supplementation-induced mitigation of gut inflammation in a porcine model of colitis. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
17
|
Tang Q, Ji Q, Xia W, Li L, Bai J, Ni R, Qin Y. Pyruvate kinase M2 regulates apoptosis of intestinal epithelial cells in Crohn's disease. Dig Dis Sci 2015; 60:393-404. [PMID: 24817408 DOI: 10.1007/s10620-014-3189-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 04/25/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Pyruvate kinase M2 (PKM2), a key glycolytic enzyme, is involved in multiple cellular processes including apoptosis. Recently increased fecal PKM2 has been found in Crohn's disease (CD), but little is known regarding its function in the pathophysiology of the disease. AIM The intestinal expression of PKM2 and its involvement in CD was investigated. METHODS Pyruvate kinase M2 expression in mucosal biopsies from patients with CD and normal controls was detected by immunohistochemistry. A murine model of colitis induced by trinitrobenzenesulphonic acid (TNBS) was established and expression of PKM2, B cell lymphoma-extra large (Bcl-xl), active caspase-3 as well as cleaved poly (ADP-ribose) polymerase (PARP) was examined for association of PKM2 with intestinal epithelial cell (IEC) apoptosis. Furthermore, we treated human IEC line HT-29 by tumor necrosis factor-α (TNF-α) and used RNA interference to analyze the role of PKM2 in IEC apoptosis. RESULTS Intestinal expression of PKM2 was higher in patients with CD compared with normal controls mainly locating in IECs. In TNBS-induced colitis, up-regulation of PKM2 was accompanied by the elevated expression of Bcl-xl, active caspase-3, and cleaved PARP. PKM2 was co-localized with active caspase-3 in IECs marked by E-cadherin, suggesting its role in IEC apoptosis. Expression of PKM2 and Bcl-xl in TNF-α-induced HT-29 cells was increased, while TNF-α had no effect on cellular localization of PKM2. Furthermore, knockdown of PKM2 by siRNA could inhibit expression of Bcl-xl but enhance apoptosis in TNF-α-treated HT-29 cells. CONCLUSION The up-regulation of PKM2 might protect IECs against apoptosis possibly through Bcl-xl in CD, indicating its important role in the pathophysiology of CD.
Collapse
Affiliation(s)
- Qiyun Tang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Arab HH, Salama SA, Eid AH, Omar HA, Arafa ESA, Maghrabi IA. Camel's milk ameliorates TNBS-induced colitis in rats via downregulation of inflammatory cytokines and oxidative stress. Food Chem Toxicol 2014; 69:294-302. [PMID: 24788059 DOI: 10.1016/j.fct.2014.04.032] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 04/12/2014] [Accepted: 04/18/2014] [Indexed: 02/06/2023]
Abstract
Current treatment strategies for inflammatory bowel diseases (IBD) are associated with several adverse effects, and thus, the search for effective agents with minimal side effects merits attention. Camel's milk (CM) is endowed with antioxidant/anti-inflammatory features and has been reported to protect against diabetes and hepatic injury, however, its effects on IBD have not been previously explored. In the current study, we aimed to investigate the potential alleviating effects of CM against TNBS-induced colitis in rats. CM (10 ml/kg b.i.d. by oral gavage) effectively suppressed the severity of colon injury as evidenced by amelioration of macroscopic damage, colon weight/length ratio, histopathological alterations, leukocyte influx and myeloperoxidase activity. Administration of CM mitigated the colonic levels of TNF-α and IL-10 cytokines. The attenuation of CM to colon injury was also associated with suppression of oxidative stress via reduction of lipid peroxides and nitric oxide along with boosting the antioxidant defenses through restoration of colon glutathione and total anti-oxidant capacity. In addition, caspases-3 activity, an apoptotic marker, was inhibited. Together, our study highlights evidences for the promising alleviating effects of CM in colitis. Thus, CM may be an interesting complementary approach for the management of IBD.
Collapse
Affiliation(s)
- Hany H Arab
- Biochemistry Division and GTMR Unit, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Samir A Salama
- Biochemistry Division and GTMR Unit, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Cairo 11751, Egypt
| | - Ahmed H Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia; Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Dokki, Cairo, Egypt
| | - Hany A Omar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia; Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - El-Shaimaa A Arafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia; Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ibrahim A Maghrabi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Taif University, Taif 21974, Saudi Arabia
| |
Collapse
|
19
|
Witaicenis A, Luchini A, Hiruma-Lima C, Felisbino S, Justulin L, Garrido-Mesa N, Utrilla P, Gálvez J, Di Stasi L. Mechanism and Effect of Esculetin in an Experimental Animal Model of Inflammatory Bowel Disease. EUR J INFLAMM 2013. [DOI: 10.1177/1721727x1301100213] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- A. Witaicenis
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - A.C. Luchini
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - C.A. Hiruma-Lima
- Department of Physiology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - S.L. Felisbino
- Department of Morphology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - L.A. Justulin
- Department of Morphology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - N. Garrido-Mesa
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - P. Utrilla
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - J. Gálvez
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - L.C. Di Stasi
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| |
Collapse
|
20
|
Silva MA, Menezes J, Dionne S, Levy E, Amre DK, Seidman EG. Herpes simplex virus-1 infection of colonic explants as a model of viral-induced activation of Crohn's disease. J Crohns Colitis 2012; 6:454-63. [PMID: 22398063 DOI: 10.1016/j.crohns.2011.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 09/20/2011] [Accepted: 09/29/2011] [Indexed: 02/08/2023]
Abstract
The exogenous triggers responsible for Crohn's disease (CD) relapses are not often identified. Cytomegalovirus and other members of the herpesvirus family have been implicated in precipitating relapses. However, the role of viral infections in the immunopathogenesis of CD remains poorly understood. We describe an ex-vivo model of primary viral infection of CD tissue with Herpes Simplex Virus type I (HSV-1). IL-6 and CD68 served as markers for CD inflammation, type I IFNs for viral infection. Colonic explants obtained from CD resections were infected via the luminal or the submucosal compartments with HSV-1 or mock virus solution, at varying concentrations for up to 20 h. Serial tissue sections were assayed for expression of HSV-1 specific antigens, CD-68, IL-6 and DC-SIGN. Culture supernatants were tested for IL-6 and type I IFN production. Positive immunostaining for HSV-1 specific antigens was consistently detectable using 11×10(6)PFU from 13 h onwards, mainly on cells located in the submucosa, and in the perivascular area. CD68 was up-regulated in lamina propria macrophages from mildly and non-inflamed CD tissue after HSV-1 infection. IL-6+ cells in the infected tissues were mainly submucosal DC-SIGN+ dendritic cells. IL-6 and IFN-β levels were higher in the supernatants from HSV-1-infected explants compared to controls after 20 h of culture (p<0.01). These data show increased expression of inflammatory markers during the initial stages of HSV-1 primary infection using CD colonic explants. This in vitro model appears promising to study the immunoregulatory changes induced by microbial infection in reactivation of CD.
Collapse
Affiliation(s)
- Manuel A Silva
- Sainte-Justine Hospital Research Center, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Witaicenis A, Luchini AC, Hiruma-Lima CA, Felisbino SL, Garrido-Mesa N, Utrilla P, Gálvez J, Di Stasi LC. Suppression of TNBS-induced colitis in rats by 4-methylesculetin, a natural coumarin: comparison with prednisolone and sulphasalazine. Chem Biol Interact 2011; 195:76-85. [PMID: 22119283 DOI: 10.1016/j.cbi.2011.11.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 11/03/2011] [Accepted: 11/08/2011] [Indexed: 12/20/2022]
Abstract
The aim of the present study was to compare the effects of the 4-methylesculetin with those produced by prednisolone and sulphasalazine and to elucidate the mechanisms involved in its action. Colitis was induced in rat by instillation of trinitrobenzenesulphonic acid (TNBS). The colon damage was evaluated using macroscopic, microscopic and biochemical analysis. In addition, in vitro studies were performed to evaluate cytokine production in cell cultures using the murine macrophage cell line RAW264.7, mouse splenocytes and the human colonic epithelial cell line Caco-2. 4-Methylesculetin produced a reduction of the macroscopic damage score and the recovery of the intestinal cytoarchitecture. These effects were associated with a prevention of the GSH depletion and an inhibition in AP activity. After colitis relapse, 4-methylesculetin improved the colonic inflammatory status as evidenced by histological findings, with a reduction in apoptosis, as well as biochemically by inhibition of colonic myeloperoxidase, alkaline phosphatase and metalloproteinase 9 activities. Paired with this inhibitive activity, there was a decrease in malondialdehyde content and in IL-1β levels. In vitro assays revealed that 4-methylesculetin promoted an inhibition in IL-1β, IL-8, IL-2 and IFN-γ production in cell cultures. In conclusion, 4-methylesculetin showed similar efficacy to that obtained with either prednisolone or sulphasalazine, both in the acute phase of colitis as well as following a curative protocol. The intestinal anti-inflammatory activity by 4-methylesculetin is likely related to its ability in reduce colonic oxidative stress and inhibit pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Aline Witaicenis
- Univ. Estadual Paulista - UNESP, Institute of Biosciences, Department of Pharmacology, Botucatu, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
22
|
DIRISINA RAMANARAO, KATZMAN REBECCAB, GORETSKY TATIANA, MANAGLIA ELIZABETH, MITTAL NAVDHA, WILLIAMS DAVIDB, QIU WEI, YU JIAN, CHANDEL NAVDEEPS, ZHANG LIN, BARRETT TERRENCEA. p53 and PUMA independently regulate apoptosis of intestinal epithelial cells in patients and mice with colitis. Gastroenterology 2011; 141:1036-45. [PMID: 21699775 PMCID: PMC3736614 DOI: 10.1053/j.gastro.2011.05.032] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 05/06/2011] [Accepted: 05/16/2011] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease (IBD) is associated with increased apoptosis of intestinal epithelial cells (IECs). Mutations in the tumor suppressor p53 appear during early stages of progression from colitis to cancer. We investigated the role of p53 and its target, p53-upregulated modulator of apoptosis (PUMA), in inflammation-induced apoptosis of IECs. METHODS Apoptosis was induced in mouse models of mucosal inflammation. Responses of IECs to acute, T-cell activation were assessed in wild-type, p53⁻/⁻, Bid⁻/⁻, Bim⁻/⁻, Bax3⁻/⁻, Bak⁻/⁻, PUMA⁻/⁻, and Noxa⁻/⁻ mice. Responses of IECs to acute and chronic colitis were measured in mice following 1 or 3 cycles of dextran sulfate sodium (DSS), respectively. Apoptosis was assessed by TUNEL staining and measuring activity of caspases 3 and 9; levels of p53 and PUMA were assessed in colon tissue from patients with and without ulcerative colitis. RESULTS Apoptosis of IECs occurred in the lower crypts of colitic tissue from humans and mice. Colitis induction with anti-CD3 or 3 cycles of DSS increased apoptosis and protein levels of p53 and PUMA in colonic crypt IECs. In p53⁻/⁻ and PUMA⁻/⁻ mice, apoptosis of IECs was significantly reduced but inflammation was not. Levels of p53 and PUMA were increased in inflamed mucosal tissues of mice with colitis and in patients with UC, compared with controls. Induction of PUMA in IECs of p53⁻/⁻ mice indicated that PUMA-mediated apoptosis was independent of p53. CONCLUSIONS In mice and humans, colon inflammation induces apoptosis of IECs via p53-dependent and - independent mechanisms; PUMA also activates an intrinsic apoptosis pathway associated with colitis.
Collapse
Affiliation(s)
- RAMANARAO DIRISINA
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - REBECCA B. KATZMAN
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - TATIANA GORETSKY
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - ELIZABETH MANAGLIA
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - NAVDHA MITTAL
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - DAVID B. WILLIAMS
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - WEI QIU
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Departments of Pharmacology and Chemical Biology, and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - JIAN YU
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Departments of Pharmacology and Chemical Biology, and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - NAVDEEP S. CHANDEL
- Division of Pulmonary & Critical Care Medicine, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - LIN ZHANG
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Departments of Pharmacology and Chemical Biology, and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - TERRENCE A. BARRETT
- Division of Gastroenterology, Department of Medicine Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
23
|
Bussuan LAM, Fagundes DJ, Marks G, Bussuan PM, Teruya R. The role of Fas ligand protein in the oxidative stress induced by azoxymethane on crypt colon of rats. Acta Cir Bras 2010; 25:501-6. [DOI: 10.1590/s0102-86502010000600008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/21/2010] [Indexed: 11/21/2022] Open
Abstract
PURPOSE: To study the protein Fas ligand (FasL) on the expression of apoptosis, using a model of oxidative stress induced by azoxymethane (AOM), in the crypt of colon in rats. METHODS: Wistar rats (n=14) were assigned into two groups: control (n=7) and AOM (n=7). A single subcutaneous administration of AOM (5mg/kg) or saline solution was performed at the beginning of third week and after three hours samples of proximal colon were collected. The expression of FasL was quantified (Software ImageLab) in percentage of areas in the top, base and all crypt. Results were expressed as mean ± sd (Shapiro-Wilks test and t Student test) (p < 0.05). RESULTS: In the animals of CG there was no significant difference between the FasL expression of the top (10.75±3.33) and basal (11.14±3.53) colon crypt (p=0.34293740). In the animals of AOM there was no significant difference between the FasL expression of the top (8.86±4.19) and basal (8.99±4.08) colon crypt (p=0.78486003). In the animals of CG (10.95±3.43) and AOM (8.92±4.13) there was a significant difference of the FasL expression (p=0.026466821). A significantly decrease on the FasL expression was observed in the animals of CG (10.75±3.33) and AOM (8.86±4.19) in the top crypt (p=0.00003755*). A significant decrease was also observed in the animals of CG (11.14±3.53) and AOM (8.99±4.08) in the basal colon crypt (p=0.00000381**). CONCLUSION: Azoxymethane induce the oxidative stress and the significantly decrease of FasL expression, although there is no significant difference between basal and top of colon crypt linked to consumption-activation of Fas ligand.
Collapse
|
24
|
Darsigny M, Babeu JP, Dupuis AA, Furth EE, Seidman EG, Lévy É, Verdu EF, Gendron FP, Boudreau F. Loss of hepatocyte-nuclear-factor-4alpha affects colonic ion transport and causes chronic inflammation resembling inflammatory bowel disease in mice. PLoS One 2009; 4:e7609. [PMID: 19898610 PMCID: PMC2764139 DOI: 10.1371/journal.pone.0007609] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 10/02/2009] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hnf4alpha, an epithelial specific transcriptional regulator, is decreased in inflammatory bowel disease and protects against chemically-induced colitis in mice. However, the precise role of this factor in maintaining normal inflammatory homeostasis of the intestine remains unclear. The aim of this study was to evaluate the sole role of epithelial Hnf4alpha in the maintenance of gut inflammatory homeostasis in mice. METHODOLOGY/PRINCIPAL FINDINGS We show here that specific epithelial deletion of Hnf4alpha in mice causes spontaneous chronic intestinal inflammation leading to focal areas of crypt dropout, increased cytokines and chemokines secretion, immune cell infiltrates and crypt hyperplasia. A gene profiling analysis in diseased Hnf4alpha null colon confirms profound genetic changes in cell death and proliferative behaviour related to cancer. Among the genes involved in the immune protection through epithelial barrier function, we identify the ion transporter claudin-15 to be down-modulated early in the colon of Hnf4alpha mutants. This coincides with a significant decrease of mucosal ion transport but not of barrier permeability in young animals prior to the manifestation of the disease. We confirm that claudin-15 is a direct Hnf4alpha gene target in the intestinal epithelial context and is down-modulated in mouse experimental colitis and inflammatory bowel disease. CONCLUSION Our results highlight the critical role of Hnf4alpha to maintain intestinal inflammatory homeostasis during mouse adult life and uncover a novel function for Hnf4alpha in the regulation of claudin-15 expression. This establishes Hnf4alpha as a mediator of ion epithelial transport, an important process for the maintenance of gut inflammatory homeostasis.
Collapse
Affiliation(s)
- Mathieu Darsigny
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Philippe Babeu
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Andrée-Anne Dupuis
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ernest G. Seidman
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Research Institute, McGill University Health Center, Montréal, Quebec, Canada
| | - Émile Lévy
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Nutrition, CHU Ste-Justine, Université de Montréal, Quebec, Canada
| | - Elena F. Verdu
- Division of Gastroenterology, McMaster University, Hamilton, Ontario, Canada
| | - Fernand-Pierre Gendron
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - François Boudreau
- Canadian Institutes of Health Research Team on Digestive Epithelium, Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
25
|
L-cysteine supplementation attenuates local inflammation and restores gut homeostasis in a porcine model of colitis. Biochim Biophys Acta Gen Subj 2009; 1790:1161-9. [DOI: 10.1016/j.bbagen.2009.05.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Revised: 05/02/2009] [Accepted: 05/27/2009] [Indexed: 01/01/2023]
|
26
|
Wang L, Luo HS, Xia H. Sodium butyrate induces human colon carcinoma HT-29 cell apoptosis through a mitochondrial pathway. J Int Med Res 2009; 37:803-11. [PMID: 19589263 DOI: 10.1177/147323000903700323] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Some tumours respond favourably to tumour necrosis factor-alpha (TNF-alpha). Despite this preferential sensitivity, resistance to TNF-alpha remains a clinical problem and more interest is now being focused on finding compounds that induce apoptosis through other pathways. Sodium butyrate (NaBt) has anti-tumour effects on colon cancer cells, inhibiting cell growth and promoting differentiation and apoptosis. In this study we investigated whether NaBt induced apoptosis in the human colon cancer cell line HT-29 and examined the intracellular mechanisms involved. Pre-incubation of cells with NaBt significantly increased apoptosis as measured by fluorescence activated cell sorter analysis and mitochondrial membrane potential determination. This effect could be blocked with the caspase inhibitors, z-VAD-fmk (pan-caspase inhibitor), z-DEVD-fmk (caspase-3 inhibitor) and z-LEHD-fmk (caspase-9 inhibitor), but not with z-IETD-fmk (caspase-8 inhibitor). Enhancement of caspase-3 and caspase-9 activities suggests that NaBt induces apoptosis via mitochondrial pathways not involving TNF-alpha.
Collapse
Affiliation(s)
- L Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | |
Collapse
|
27
|
Bouchard V, Harnois C, Demers MJ, Thibodeau S, Laquerre V, Gauthier R, Vézina A, Noël D, Fujita N, Tsuruo T, Arguin M, Vachon PH. B1 integrin/Fak/Src signaling in intestinal epithelial crypt cell survival: integration of complex regulatory mechanisms. Apoptosis 2008; 13:531-42. [PMID: 18322799 DOI: 10.1007/s10495-008-0192-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular determinants which dictate survival and apoptosis/anoikis in human intestinal crypt cells remain to be fully understood. To this effect, the roles of beta1 integrin/Fak/Src signaling to the PI3-K/Akt-1, MEK/Erk, and p38 pathways, were investigated. The regulation of six Bcl-2 homologs (Bcl-2, Mcl-1, Bcl-X(L), Bax, Bak, Bad) was likewise analyzed. We report that: (1) Anoikis causes a down-activation of Fak, Src, Akt-1 and Erk1/2, a loss of Fak-Src association, and a sustained/enhanced activation of p38beta, which is required as apoptosis/anoikis driver; (2) PI3-K/Akt-1 up-regulates the expression of Bcl-X(L) and Mcl-1, down-regulates Bax and Bak, drives Bad phosphorylation (both serine112/136 residues) and antagonizes p38beta activation; (3) MEK/Erk up-regulates Bcl-2, drives Bad phosphorylation (serine112 residue), but does not antagonize p38bactivation; (4) PI3-K/Akt-1 is required for survival, whereas MEK/Erk is not; (5) Src acts as a cornerstone in the engagement of both pathways by beta1 integrins/Fak, and is crucial for survival; and (6) beta1 integrins/Fak and/or Src regulate Bcl-2 homologs as both PI3-K/Atk-1 and MEK/Erk combined. Hence, beta1 integrin/Fak/Src signaling translates into integrated mediating functions of p38beta activation and regulation of Bcl-2 homologs by PI3-K/Akt-1 and MEK/Erk, consequently determining their requirement (or not) for survival.
Collapse
Affiliation(s)
- Véronique Bouchard
- Département d'Anatomie et de Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Larson SD, Li J, Chung DH, Mark Evers B. Molecular mechanisms contributing to glutamine-mediated intestinal cell survival. Am J Physiol Gastrointest Liver Physiol 2007; 293:G1262-71. [PMID: 17916648 PMCID: PMC2432018 DOI: 10.1152/ajpgi.00254.2007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glutamine, the most abundant amino acid in the bloodstream, is the preferred fuel source for enterocytes and plays a vital role in the maintenance of mucosal growth. The molecular mechanisms regulating the effects of glutamine on intestinal cell growth and survival are poorly understood. Here, we show that addition of glutamine (1 mmol/l) enhanced rat intestinal epithelial (RIE)-1 cell growth; conversely, glutamine deprivation increased apoptosis as noted by increased DNA fragmentation and caspase-3 activity. To delineate signaling pathways involved in the effects of glutamine on intestinal cells, we assessed activation of extracellular signal-related kinase (ERK), protein kinase D (PKD), and phosphatidylinositol 3-kinase (PI3K)/Akt, which are important pathways in cell growth and survival. Addition of glutamine activated ERK and PKD in RIE-1 cells after a period of glutamine starvation; inhibition of ERK, but not PKD, increased cell apoptosis. Conversely, glutamine starvation alone increased phosphorylated Akt; inhibition of Akt enhanced RIE-1 cell DNA fragmentation. The role of ERK was further delineated using RIE-1 cells stably transfected with an inducible Ras. Apoptosis was significantly increased following ERK inhibition, despite Ras activation. Taken together, these results identify a critical role for the ERK signaling pathways in glutamine-mediated intestinal homeostasis. Furthermore, activation of PI3K/Akt during periods of glutamine deprivation likely occurs as a protective mechanism to limit apoptosis associated with cellular stress. Importantly, our findings provide novel mechanistic insights into the antiapoptotic effects of glutamine in the intestine.
Collapse
Affiliation(s)
- Shawn D. Larson
- Department of Surgery The University of Texas Medical Branch, Galveston, Texas
| | - Jing Li
- Department of Surgery The University of Texas Medical Branch, Galveston, Texas,The Sealy Center for Cancer Cell Biology The University of Texas Medical Branch, Galveston, Texas
| | - Dai H. Chung
- Department of Surgery The University of Texas Medical Branch, Galveston, Texas,The Sealy Center for Cancer Cell Biology The University of Texas Medical Branch, Galveston, Texas
| | - B. Mark Evers
- Department of Surgery The University of Texas Medical Branch, Galveston, Texas,The Sealy Center for Cancer Cell Biology The University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
29
|
Hu S, Ciancio MJ, Lahav M, Fujiya M, Lichtenstein L, Anant S, Musch MW, Chang EB. Translational inhibition of colonic epithelial heat shock proteins by IFN-gamma and TNF-alpha in intestinal inflammation. Gastroenterology 2007; 133:1893-904. [PMID: 18054561 PMCID: PMC2180161 DOI: 10.1053/j.gastro.2007.09.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 09/06/2007] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Inducible heat shock proteins (iHsp), Hsp25/27 and Hsp70, play essential roles in protecting cells against stress and, in intestinal mucosal inflammation, potentially lessening the extent and severity of injury. We examined the expression and regulation of iHsp in human and experimental inflammatory bowel diseases (IBD) and in vitro. METHODS iHsp expression and regulation were assessed in normal and IBD colonic biopsy specimens, IL-10(-/-) mice, and young adult mouse colonic epithelial cells by immunohistochemistry, Western blot, and real-time polymerase chain reaction (PCR). Phosphorylation of double-stranded RNA-dependent protein kinase (PKR) and eukaryotic initiation factor-2alpha (eIF-2alpha) was determined by Western blot. RESULTS Hsp25/27 and Hsp70 levels were selectively reduced in areas of active mucosal inflammation associated with human IBD and IL-10(-/-) mice with colitis. Wild-type mice treated in vivo with interferon (IFN)-gamma + tumor necrosis factor (TNF)-alpha also demonstrated reduced colonic Hsp25/27 and Hsp70. In young adult mouse colonic epithelial cells, IFN-gamma+TNF-alpha inhibited heat induction of Hsp25/27 and Hsp70, an effect not associated with changes in iHsp messenger RNA or protein half-lives but caused by suppressed de novo iHsp synthesis. IFN-gamma+TNF-alpha cotreatment activated PKR, resulting in phosphorylation and inactivation of eIF-2alpha, an essential factor in protein translation. These effects were not due to induced apoptosis and could be negated by PKR-inhibitor and short interfering RNA to PKR. Increased phosphorylation of PKR and eIF-2alpha were also observed in active IBD tissues. CONCLUSIONS Mucosal inflammation is associated with iHsp down-regulation, an effect that appears mediated by translational down-regulation by proinflammatory cytokines. In the context of IBD, we propose that this mechanism contributes to the severity, extent, and persistence of inflammation-induced mucosal injury.
Collapse
Affiliation(s)
- Shien Hu
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL
| | - Mae J. Ciancio
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL
| | - Maor Lahav
- Department of Gastroenterology, Chaim Sheba Medical Center, Affiliated to the Tel-Aviv University, Tel-Hashomer, Israel
| | - Mikihiro Fujiya
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL, Third Department of Internal Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Lev Lichtenstein
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL, Department of Gastroenterology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Shrikant Anant
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Mark W. Musch
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL
| | - Eugene B. Chang
- The Martin Boyer Laboratories, Department of Medicine, University of Chicago IBD Research Center, Chicago, IL
| |
Collapse
|
30
|
Chapkin RS, Davidson LA, Ly L, Weeks BR, Lupton JR, McMurray DN. Immunomodulatory effects of (n-3) fatty acids: putative link to inflammation and colon cancer. J Nutr 2007; 137:200S-204S. [PMID: 17182826 DOI: 10.1093/jn/137.1.200s] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammation and colorectal cancer are closely linked. Although the overall mechanisms of inflammation-associated gastrointestinal carcinogenesis are complex, it is clear that antiinflammatory therapy is efficacious against neoplastic progression and malignant conversion. From a dietary perspective, fish oil containing (n-3) polyunsaturated fatty acids (PUFAs) has antiinflammatory properties, but for years the mechanism has remained obscure. Of relevance to the immune system in the intestine, we showed that (n-3) PUFA feeding alters the balance between CD4+ T-helper (Th1 and Th2) subsets by directly suppressing Th1 cell development (i.e., clonal expansion). This is noteworthy because Th1 cells mediate inflammatory diseases and resistance to intracellular pathogens or allergic hypersensitivity, and Th2 cells mediate resistance to extracellular pathogens. Therefore, any changes induced by (n-3) PUFAs in T-cell subset balance and function are important because the outcome is expected to suppress the development of autoimmune diseases and possibly the occurrence of colon cancer. Precisely how the immunomodulatory effects of (n-3) PUFAs influence inflammation-associated colonic tumor development is the subject of an ongoing investigation.
Collapse
Affiliation(s)
- Robert S Chapkin
- Faculty of Nutrition, Center for Environmental and Rural Health, Texas A&M University, College Station, TX 77843, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Evans ME, Jones DP, Ziegler TR. Glutamine inhibits cytokine-induced apoptosis in human colonic epithelial cells via the pyrimidine pathway. Am J Physiol Gastrointest Liver Physiol 2005; 289:G388-96. [PMID: 15878985 DOI: 10.1152/ajpgi.00072.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glutamine (Gln) prevents apoptosis in intestinal epithelial cells, but the mechanism(s) remain unknown. Gln-derived metabolites include ammonia, glutamate (Glu), glutathione (GSH), and nucleotides. We previously showed that Gln potently inhibited apoptosis in cytokine-treated human colonic HT-29 cells; this effect was specific to Gln, unaffected by Glu, and unrelated to intracellular GSH. The current research examines mechanism(s) for Gln-induced antiapoptotic effects in HT-29 cells treated with TNF-alpha-related apoptosis-inducing ligand (TRAIL). Proliferating cells were treated with Gln or selected Gln metabolites for 24 h. Cells were then treated with TRAIL and Gln or its downstream metabolites, and apoptosis was assessed at 8 h after treatment. The purine and pyrimidine precursors inosine and orotate inhibited TRAIL-induced apoptosis. However, inhibition of purine synthesis with azaserine did not alter the potent antiapoptotic effect of Gln. In contrast, the pyrimidine synthesis inhibitor, acivicin, completely prevented this response. Supplementation with the pyrimidine uracil or the pyrimidine precursor orotate rescued the acivicin-induced blockade of Gln antiapoptotic action. Removal of bicarbonate, a substrate for pyrimidine synthesis, also inhibited the antiapoptotic effects of Gln. Uracil and thymine alone also significantly decreased TRAIL-induced apoptosis. The antiapoptotic effects of Gln were independent of DNA/RNA synthesis as measured by flow cytometry and bromodeoxyuridine incorporation. In conclusion, Gln prevents TRAIL-induced apoptosis in HT-29 cells through a mechanism involving the pyrimidine pathway. Our data also demonstrate the novel antiapoptotic effects of pyrimidine bases and their precursor orotate in these human intestinal cells.
Collapse
Affiliation(s)
- Mary E Evans
- Department of Medicine, Center for Clinical and Molecular Nutrition, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
32
|
Abstract
BACKGROUND Interleukin (IL)-15 is overexpressed in intestinal tissue with active Crohn's disease (CD). However, its role in the pathogenesis of the disease remains uncertain. We studied the effects of IL-15 on colonic mucosal proinflammatory cytokine response in vitro using organ culture of human colonic explants. METHODS Colonic tissue was obtained from (1) resections in pediatric CD patients (inflamed and noninflamed) and (2) rectal biopsies in patients with CD undergoing colonoscopy (n = 31) and controls (n = 9). In preliminary experiments, explants from the resections were cultured in the presence or absence of a simulated T(H)1 stimulation using ionomycin (Io) and phorbol-myristate-acetate (PMA), with or without IL-15, or in medium alone. Rectal biopsies were cultured in the same conditions as above, with or without adding a monoclonal anti-IL-15 neutralizing antibody (mAb). Levels of interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, and IL-2R alpha were measured by enzyme-linked immunosorbent assay. RESULTS IL-15, in the absence of Io + PMA, did not induce the expression of IFN-gamma, TNF-alpha, or IL-2R alpha. Only inflamed explants from resections stimulated with Io + PMA expressed IFN-gamma, TNF-alpha, and IL-2R alpha. This T(H)1 stimulatory effect was inhibited by IL-15 in a dose-dependent fashion. In rectal biopsy explants, inflamed, noninflamed CD, and control tissue responded to stimulation with Io + PMA (P < 0.05) with increased IFN-gamma and TNF-alpha (P < 0.05). This response was again inhibited by IL-15. The inhibitory effect of IL-15 was specifically reversed by anti-IL-15 mAb (P < 0.05). The data for the CD group were also analyzed according to the severity of colonic inflammation and medication use. CONCLUSIONS Our results suggest a possible anti-inflammatory role for IL-15 in CD. We postulate that its overexpression in CD potentially represents a protective mechanism against the exaggerated T(H)1 immune response.
Collapse
Affiliation(s)
- Manuel A Silva
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
33
|
Kitazawa H, Nishihara T, Nambu T, Nishizawa H, Iwaki M, Fukuhara A, Kitamura T, Matsuda M, Shimomura I. Intectin, a Novel Small Intestine-specific Glycosylphosphatidylinositol-anchored Protein, Accelerates Apoptosis of Intestinal Epithelial Cells. J Biol Chem 2004; 279:42867-74. [PMID: 15292182 DOI: 10.1074/jbc.m408047200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Intestinal epithelial cells undergo rapid turnover and exfoliation especially at the villus tips. This process is modulated by various nutrients especially fat. Apoptosis is one of the important regulatory mechanisms of this turnover. Therefore, identification of the factors that control epithelial cell apoptosis should help us understand the mechanism of intestinal mucosal turnover. Here, we report the identification of a novel small intestine-specific member of the Ly-6 family, intectin, by signal sequence trap method. Intectin mRNA expression was exclusively identified in the intestine and localized at the villus tips of intestinal mucosa, which is known to undergo apoptosis. Intectin mRNA expression was modulated by nutrition. Intestinal epithelial cells expressing intectin were more sensitive to palmitate-induced apoptosis, compared with control intestinal epithelial cells, and such effect was accompanied by increased activity of caspase-3. Intectin expression also reduced cell-cell adhesion of intestinal epithelial cells.
Collapse
Affiliation(s)
- Hidefumi Kitazawa
- Department of Medicine and Pathophysiology, Graduate School of Frontier Bioscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sýkora J, Varvarovská J, Stozický F, Vondráková R, Svecová M, Siala K, Schwarz J. Adolescent herpes simplex viral infection related Ludwig's angina in ulcerative colitis. J Pediatr Gastroenterol Nutr 2004; 38:221-3. [PMID: 14734889 DOI: 10.1097/00005176-200402000-00023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- Josef Sýkora
- Department of Paediatrics, Charles University Hospital, Pilsen, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
35
|
Evans ME, Jones DP, Ziegler TR. Glutamine Prevents Cytokine-Induced Apoptosis in Human Colonic Epithelial Cells. J Nutr 2003; 133:3065-71. [PMID: 14519785 DOI: 10.1093/jn/133.10.3065] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epithelial cell apoptosis is an important regulator of normal gut mucosal turnover; however, excessive apoptosis may inhibit mucosal restitution during pathophysiologic states. Apoptosis is induced by oxidative stress and cytokines, but regulation by specific nutrients has been infrequently studied under these conditions. Glutamine (Gln) is an important metabolic fuel for intestinal epithelial cells and a precursor to the antioxidant glutathione (GSH), which has antiapoptotic effects. In cultured intestinal epithelial cells, Gln depletion increases oxidant-induced apoptosis. This study examined whether Gln protects against apoptosis induced by the cytokine tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) in the human colon carcinoma cell line, HT-29. TRAIL-induced apoptosis in HT-29 cells was characterized by an increase in the percentage of cells in the sub-G1 fraction by flow cytometry, nuclear condensation and the activation of caspase-8 and caspase-3. TRAIL-induced apoptosis was completely prevented by Gln, but not inhibited by other amino acids, including the GSH constituents, glutamate, cysteine and glycine. Similar antiapoptotic effects of Gln occurred when apoptosis was induced by a combination of tumor necrosis factor-alpha and interferon-gamma. Cellular GSH was oxidized during TRAIL-induced apoptosis. This effect was completely blocked by Gln, however, inhibition of GSH synthesis with buthionine sulfoximine did not alter Gln antiapoptotic effects. Furthermore, glutamate prevented GSH oxidation in response to TRAIL but did not protect against TRAIL-induced apoptosis. These results show that Gln specifically protects intestinal epithelial cells against cytokine-induced apoptosis, and that this occurs by a mechanism that is distinct from the protection against oxidative stress mediated by cellular GSH.
Collapse
Affiliation(s)
- Mary E Evans
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
36
|
Groos S, Reale E, Luciano L. General suitability of techniques for in situ detection of apoptosis in small intestinal epithelium. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 272:503-13. [PMID: 12740944 DOI: 10.1002/ar.a.10063] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The present study was designed to evaluate different techniques for the in situ detection of apoptosis in human and rat small intestinal epithelium. The techniques included light microscopy (LM) and transmission electron microscopy (TEM) observation of epoxy resin-embedded tissue, scanning electron microscopy (SEM), TUNEL assay, and antibodies directed against caspase cleavage products of caspase 3, cytokeratin 18 (CK 18), and apoptotic single-strand DNA (ssDNA). All techniques, if the labeling was positive, showed apoptotic cells exclusively at the villus tip. LM and TEM were the most reliable and revealed morphological signs typical of cells that have died via apoptosis. SEM indicated the extension of the process. The antibody recognizing cleaved caspase 3 could be considered an appropriate marker for apoptotic epithelial cells in human and rat small intestine. However, the majority of epithelial cells lining the proximal small intestinal villus contained only low levels of intact CK 18. Therefore, sufficient amounts of cleaved CK 18 for immunohistochemical detection were not generated during apoptosis, rendering the application of the antibody inappropriate. The antibody detecting formamide-denatured ssDNA in apoptotic cells was both suitable and reliable; however, the particular staining procedure used compromised the tissue preservation. In comparison to this, the TUNEL assay was less reliable. Although it was performed with a commercially available ready-to-use kit, its application conditions had to be adjusted for each specimen on the basis of the findings produced by other techniques.
Collapse
Affiliation(s)
- Stephanie Groos
- Department of Microscopic Anatomy, Center of Anatomy, Hannover Medical School, Hannover, Germany.
| | | | | |
Collapse
|