51
|
Mehta M, Ahmed S, Dryden G. Immunopathophysiology of inflammatory bowel disease: how genetics link barrier dysfunction and innate immunity to inflammation. Innate Immun 2018; 23:497-505. [PMID: 28770665 DOI: 10.1177/1753425917722206] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic or relapsing immune activation and corresponding inflammation within the gastrointestinal (GI) tract. Diverse genetic mutations, encoding important aspects of innate immunity and mucosal homeostasis, combine with environmental triggers to create inappropriate, sustained inflammatory responses. Recently, significant advances have been made in understanding the interplay of the intestinal epithelium, mucosal immune system, and commensal bacteria as a foundation of the pathogenesis of inflammatory bowel disease. Complex interactions between specialized intestinal epithelial cells and mucosal immune cells determine different outcomes based on the environmental input: the development of tolerance in the presence of commensal bacterial or the promotion of inflammation upon recognition of pathogenic organisms. This article reviews key genetic abnormalities involved in inflammatory and homeostatic pathways that enhance susceptibility to immune dysregulation and combine with environmental triggers to trigger the development of chronic intestinal inflammation and IBD.
Collapse
Affiliation(s)
- Minesh Mehta
- 1 Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY, USA
| | - Shifat Ahmed
- 2 Department of Internal Medicine, University of Louisville, Louisville, KY, USA
| | - Gerald Dryden
- 1 Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY, USA
| |
Collapse
|
52
|
Manda B, Mir H, Gangwar R, Meena AS, Amin S, Shukla PK, Dalal K, Suzuki T, Rao R. Phosphorylation hotspot in the C-terminal domain of occludin regulates the dynamics of epithelial junctional complexes. J Cell Sci 2018; 131:jcs206789. [PMID: 29507118 PMCID: PMC5963837 DOI: 10.1242/jcs.206789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
The apical junctional complex (AJC), which includes tight junctions (TJs) and adherens junctions (AJs), determines the epithelial polarity, cell-cell adhesion and permeability barrier. An intriguing characteristic of a TJ is the dynamic nature of its multiprotein complex. Occludin is the most mobile TJ protein, but its significance in TJ dynamics is poorly understood. On the basis of phosphorylation sites, we distinguished a sequence in the C-terminal domain of occludin as a regulatory motif (ORM). Deletion of ORM and expression of a deletion mutant of occludin in renal and intestinal epithelia reduced the mobility of occludin at the TJs. ORM deletion attenuated Ca2+ depletion, osmotic stress and hydrogen peroxide-induced disruption of TJs, AJs and the cytoskeleton. The double point mutations T403A/T404A, but not T403D/T404D, in occludin mimicked the effects of ORM deletion on occludin mobility and AJC disruption by Ca2+ depletion. Both Y398A/Y402A and Y398D/Y402D double point mutations partially blocked AJC disruption. Expression of a deletion mutant of occludin attenuated collective cell migration in the renal and intestinal epithelia. Overall, this study reveals the role of ORM and its phosphorylation in occludin mobility, AJC dynamics and epithelial cell migration.
Collapse
Affiliation(s)
- Bhargavi Manda
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Hina Mir
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Ruchika Gangwar
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Avtar S Meena
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Shrunali Amin
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Pradeep K Shukla
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Kesha Dalal
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - Takuya Suzuki
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| | - RadhaKrishna Rao
- Departments of Physiology, University of Tennessee Health Science Center, 3 North Dunlap, Memphis, TN, 38103 USA
| |
Collapse
|
53
|
SLC26A3 (DRA) prevents TNF-alpha-induced barrier dysfunction and dextran sulfate sodium-induced acute colitis. J Transl Med 2018; 98:462-476. [PMID: 29330471 DOI: 10.1038/s41374-017-0005-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/08/2017] [Accepted: 11/15/2017] [Indexed: 12/24/2022] Open
Abstract
SLC26A3 encodes a Cl-/HCO3- ion transporter that is also known as downregulated in adenoma (DRA) and is involved in HCO3-/mucus formation. The role of DRA in the epithelial barrier has not been previously established. In this study, we investigated the in vivo and in vitro mechanisms of DRA in the colon epithelial barrier. Immunofluorescence (IF) and co-immunoprecipitation (co-IP) studies reveal that DRA binds directly to tight junction (TJ) proteins and affects the expression of TJ proteins in polarized Caco-2BBe cells. Similarly, DRA colocalizes with ZO-1 in the intestinal epithelium. Knockdown or overexpression of DRA leads to alterations in TJ proteins and epithelial permeability. In addition, TNF-α treatment downregulates DRA by activating NF-кB and subsequently affecting intestinal epithelial barrier integrity. Furthermore, overexpression of DRA partly reverses the TNF-α-induced damage by stabilizing TJ proteins. Neutralization of TNF-α in dextran sulfate sodium (DSS)-induced colitis mice demonstrates improved the outcomes, and the therapeutic effect of the TNF-α neutralizing mAb is mediated in part by the preservation of DRA expression. These data suggest that DRA may be one of the therapeutic targets of TNF-α. Moreover, DRA delivered by adenovirus vector significantly prevents the exacerbation of colitis and improves epithelial barrier function by promoting the recovery of TJ proteins in DSS-treated mice. In conclusion, DRA plays a role in protecting the epithelial barrier and may be a therapeutic target in gut homeostasis.
Collapse
|
54
|
Cartwright JA, Gow AG, Milne E, Drummond D, Smith S, Handel I, Mellanby RJ. Vitamin D Receptor Expression in Dogs. J Vet Intern Med 2018; 32:764-774. [PMID: 29469965 PMCID: PMC5866978 DOI: 10.1111/jvim.15052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/11/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023] Open
Abstract
Background There is growing evidence linking low blood vitamin D concentration to numerous diseases in people and in dogs. Vitamin D influences cellular function by signaling through the vitamin D receptor (VDR). Little is known about which non‐skeletal tissues express the VDR or how inflammation influences its expression in the dog. Objectives To define which non‐skeletal canine tissues express the VDR and to investigate expression in inflamed small intestine. Animals Thirteen non‐skeletal tissues were collected prospectively from 6 control dogs. Thirty‐five dogs diagnosed with a chronic enteropathy (CE) and 24 control dogs were prospectively enrolled and duodenal biopsies were evaluated for VDR expression. Methods Prospective; blinded assessment of canine intestinal VDR. Dogs with CE were included once other identifiable causes of intestinal disease were excluded. Age matched controls were included with no intestinal clinical signs. VDR expression was assessed immunohistochemically in all samples, using a Rat IgG VDR monoclonal antibody. Quantitative real‐time polymerase chain reaction (qPCR) was also used for duodenal biopsies. Results VDR expression as assessed by immunohistochemistry (IHC) was highest in the kidney, duodenum, skin, ileum and spleen, and weak in the colon, heart, lymph node, liver, lung, and ovary. Gastric and testicular tissue did not express the VDR. There was no statistical difference in duodenal VDR expression between the 24 healthy dogs and 34 dogs with CE when quantified by either qPCR (P = 0.87) or IHC (P = 0.099). Conclusions and Clinical Importance The lack of down regulation of VDR expression in inflamed intestine contrasts with previous studies in humans. Our findings support future studies to investigate whether vitamin D and its analogues can be used to modulate intestinal inflammation in the dog.
Collapse
Affiliation(s)
- J A Cartwright
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - A G Gow
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - E Milne
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - D Drummond
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - S Smith
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - I Handel
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| | - R J Mellanby
- Division of Veterinary Clinical Studies, Royal (Dick) School of Veterinary Studies and The Roslin Institute, Hospital for Small Animals, Easter Bush Veterinary Centre, The University of Edinburgh, Roslin, United Kingdom
| |
Collapse
|
55
|
Yu M, Wang Q, Ma Y, Li L, Yu K, Zhang Z, Chen G, Li X, Xiao W, Xu P, Yang H. Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity. Int J Biol Sci 2018; 14:69-77. [PMID: 29483826 PMCID: PMC5821050 DOI: 10.7150/ijbs.22259] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/14/2017] [Indexed: 12/19/2022] Open
Abstract
Activation of Aryl hydrocarbon receptor (AhR) is involved in the control of intestinal mucosal homeostasis. Intestinal barrier dysfunction contributes to the development of many intestinal diseases, such as inflammatory bowel disease (IBD). In this study, we investigated the mechanisms of AhR activation in the maintenance of intestinal barrier function. Adult C57BL/6 mice were treated with dextran sulphate sodium (DSS) for 7 days, with or without 6-Formylindolo(3,2-b)carbazole (FICZ), a ligand of AhR. We found that AhR activation by FICZ attenuated the decreased TJ protein expression in the colonic mucosa of the DSS-induced mice. Further, the increase of both MLC phosphorylation and MLCK expression in the mice with DSS-induced colitis was also significantly inhibited by FICZ induced AhR activation. For in vitro experiments, Caco-2 cells were treated with tumour necrosis factor alpha (TNF-α)/interferon gamma (IFN-γ) for 48 h, with or without FICZ. AhR activation prevented TNF-α/IFN-γ-induced decrease in TER and morphological disruption of the TJs in Caco-2 monolayers. It also inhibited TNF-α/IFN-γ-induced increase in MLCK expression and MLC phosphorylation by suppression of NF-κB p65 signaling pathway. Thus, AhR-activating factors might have potential as therapeutic agents for the treatment of patients with IBD.
Collapse
Affiliation(s)
- Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qimeng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuanhang Ma
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Liangzi Li
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Kun Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhicao Zhang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiangsheng Li
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Pengyuan Xu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
56
|
Kang T, Ding W, Zhang L, Ziemek D, Zarringhalam K. A biological network-based regularized artificial neural network model for robust phenotype prediction from gene expression data. BMC Bioinformatics 2017; 18:565. [PMID: 29258445 PMCID: PMC5735940 DOI: 10.1186/s12859-017-1984-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Stratification of patient subpopulations that respond favorably to treatment or experience and adverse reaction is an essential step toward development of new personalized therapies and diagnostics. It is currently feasible to generate omic-scale biological measurements for all patients in a study, providing an opportunity for machine learning models to identify molecular markers for disease diagnosis and progression. However, the high variability of genetic background in human populations hampers the reproducibility of omic-scale markers. In this paper, we develop a biological network-based regularized artificial neural network model for prediction of phenotype from transcriptomic measurements in clinical trials. To improve model sparsity and the overall reproducibility of the model, we incorporate regularization for simultaneous shrinkage of gene sets based on active upstream regulatory mechanisms into the model. RESULTS We benchmark our method against various regression, support vector machines and artificial neural network models and demonstrate the ability of our method in predicting the clinical outcomes using clinical trial data on acute rejection in kidney transplantation and response to Infliximab in ulcerative colitis. We show that integration of prior biological knowledge into the classification as developed in this paper, significantly improves the robustness and generalizability of predictions to independent datasets. We provide a Java code of our algorithm along with a parsed version of the STRING DB database. CONCLUSION In summary, we present a method for prediction of clinical phenotypes using baseline genome-wide expression data that makes use of prior biological knowledge on gene-regulatory interactions in order to increase robustness and reproducibility of omic-scale markers. The integrated group-wise regularization methods increases the interpretability of biological signatures and gives stable performance estimates across independent test sets.
Collapse
Affiliation(s)
- Tianyu Kang
- Department of Computer Science, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 02125 MA USA
| | - Wei Ding
- Department of Computer Science, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 02125 MA USA
| | - Luoyan Zhang
- Department of Computer Science, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 02125 MA USA
| | - Daniel Ziemek
- Inflammation and Immunology, Pfizer Worldwide Research & Development, Berlin, Germany
| | - Kourosh Zarringhalam
- Department of Mathematics, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 0212 MA USA
| |
Collapse
|
57
|
Sunuwar L, Medini M, Cohen L, Sekler I, Hershfinkel M. The zinc sensing receptor, ZnR/GPR39, triggers metabotropic calcium signalling in colonocytes and regulates occludin recovery in experimental colitis. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0420. [PMID: 27377730 DOI: 10.1098/rstb.2015.0420] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 12/22/2022] Open
Abstract
Impaired epithelial barrier function is a hallmark of inflammatory bowel diseases, such as colitis, contributing to diarrhoea and perpetuating inflammation. We show that the zinc sensing receptor, ZnR/GPR39, triggers intracellular Ca(2+) signalling in colonocytes thereby inducing occludin expression. Moreover, ZnR/GPR39 is essential for epithelial barrier recovery in the dextran sodium sulfate (DSS) ulcerative colitis model. Loss of ZnR/GPR39 results in increased susceptibility to DSS-induced inflammation, owing to low expression of the tight junction protein occludin and impaired epithelial barrier. Recovery of wild-type (WT) mice from the DSS insult was faster than that of ZnR/GPR39 knockout (KO) mice. Enhanced recovery of the epithelial layer and increased crypt regeneration were observed in WT mice compared with ZnR/GPR39 KO, suggesting that ZnR/GPR39 is promoting epithelial barrier integrity following DSS insult. Indeed, cell proliferation and apical expression of occludin, following the DSS-induced epithelial erosion, were increased in WT tissue but not in ZnR/GPR39 KO tissue. Importantly, survival following DSS treatment was higher in WT mice compared with ZnR/GPR39 KO mice. Our results support a direct role for ZnR/GPR39 in promoting epithelial renewal and barrier function following DSS treatment, thereby affecting the severity of the disease. We suggest ZnR/GPR39 as a novel therapeutic target that can improve epithelial barrier function in colitis.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Michal Medini
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Limor Cohen
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
58
|
Zeng Q, He X, Puthiyakunnon S, Xiao H, Gong Z, Boddu S, Chen L, Tian H, Huang SH, Cao H. Probiotic Mixture Golden Bifido Prevents Neonatal Escherichia coli K1 Translocation via Enhancing Intestinal Defense. Front Microbiol 2017; 8:1798. [PMID: 28979247 PMCID: PMC5611410 DOI: 10.3389/fmicb.2017.01798] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Escherichia coli (E. coli) K1 sepsis and meningitis is a severe infection characterized by high mortality in neonates. Successful colonization and translocation across the intestinal mucosa have been regarded as the critical steps for E. coli K1 sepsis and meningitis. We recently reported that the probiotic mixture, Golden Bifido (containing live Lactobacillus bulgaricus, Bifidobacterium, and Streptococcus thermophilus, LBS) has a preventive role against neonatal E. coli K1 bacteremia and meningitis. However, the interaction between the neonatal gut barrier, probiotics and E. coli K1 is still not elucidated. The present study aims to investigate how LBS exerts its protective effects on neonatal gut barrier during E. coli K1 infection. The beneficial effects of LBS were explored in vitro and in vivo using human colon carcinoma cell lines HT-29 and rat model of neonatal E. coli K1 infection, respectively. Our results showed that stimulation with E. coli K1 was able to cause intestinal barrier dysfunction, which were reflected by E. coli K1-induced intestinal damage and apoptosis of intestinal epithelial cells, reduction of mucin, immunoglobulin A (IgA) and tight junction proteins expression, as well as increase in intestinal permeability, all these changes facilitate E. coli K1 intestinal translocation. However, these changes were alleviated when HT-29 cells were treated with LBS before E. coli K1 infection. Furthermore, we found that LBS-treated neonatal rats (without E. coli K1 infection) have showed higher production of mucin, ZO-1, IgA, Ki67 in intestinal mucosa as well as lower intestinal permeability than that of non-treated rats, indicating that LBS could accelerate the development of neonatal intestinal defense. Taken together, our results suggest that enhancement of the neonatal intestinal defense to fight against E. coli K1 translocation could be the potential mechanism to elucidate how LBS confers a protective effect against neonatal E. coli K1 bacteremia and meningitis. This indirect mechanism makes LBS exert preventive effect on most of gut-derived pathogenic infections rather than only E. coli.
Collapse
Affiliation(s)
- Qing Zeng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Xiaolong He
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Santhosh Puthiyakunnon
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Hansen Xiao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Zelong Gong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Swapna Boddu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Lecheng Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Huiwen Tian
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China.,The First School of Clinical Medicine, Southern Medical UniversityGuangzhou, China
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China.,Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los AngelesCA, United States
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| |
Collapse
|
59
|
Jo H, Hwang D, Kim JK, Lim YH. Oxyresveratrol improves tight junction integrity through the PKC and MAPK signaling pathways in Caco-2 cells. Food Chem Toxicol 2017; 108:203-213. [PMID: 28780155 DOI: 10.1016/j.fct.2017.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022]
Abstract
Strengthening intestinal tight junctions (TJ) provides an effective barrier from the external environment and is important for recovery from inflammatory bowel disease. Oxyresveratrol (OXY), an isomer of hydroxylated resveratrol, is isolated from many plants. The aim of this study was to investigate the effect of OXY on intestinal TJ and to elucidate the mechanism underlying the OXY-mediated increase in TJ integrity in human intestinal Caco-2 cells. OXY-treated Caco-2 cell monolayers showed decreased monolayer permeability as evaluated by paracellular transport assay. The results showed that OXY significantly increased the levels of TJ-related genes and proteins (Claudin-1, Occludin and ZO-1) compared with those of the negative control. OXY activated protein kinase C (PKC) and increased expression levels of mitogen-activated protein kinase (MAPK) genes. OXY also increased gene and protein levels of the transcription factor Cdx-2. Expression levels of TJ, PKC and Cdx-2 proteins and transepithelial electrical resistance (TEER) value decreased in OXY-treated Caco-2 cells following treatment with a pan-PKC inhibitor compared with those of the untreated control. In conclusion, OXY strengthens the integrity of the intestinal TJ barrier via activation of the PKC and MAPK pathways.
Collapse
Affiliation(s)
- HyunA Jo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 136-701, Republic of Korea
| | - Dahyun Hwang
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan 31499, Republic of Korea
| | - Jeong-Keun Kim
- Department of Chemical Engineering and Biotechnology, Korea Polytechnic University, Shihung-si, Gyeonggi-do 429-793, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 136-701, Republic of Korea; Department of Public Health Science (Brain Korea 21 PLUS Program), Graduate School, Korea University, Seoul 136-701, Republic of Korea; Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 152-703, Republic of Korea.
| |
Collapse
|
60
|
Courthion H, Mugnier T, Rousseaux C, Möller M, Gurny R, Gabriel D. Self-assembling polymeric nanocarriers to target inflammatory lesions in ulcerative colitis. J Control Release 2017; 275:32-39. [PMID: 28774843 DOI: 10.1016/j.jconrel.2017.07.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 07/28/2017] [Accepted: 07/30/2017] [Indexed: 02/07/2023]
Abstract
We have developed a self-assembling polymeric nanocarrier to deliver the potent immunosuppressive drug Cyclosporine A (CsA) to inflammatory lesions in ulcerative colitis (UC) patients. Our nanocarrier has a high drug loading capacity and efficiently targets its CsA payload to the diseased tissue after local administration. Tissue drug levels were several orders of magnitude higher in animals suffering from a trinitrobenzene-sulfonic acid (TNBS) - induced colitis, compared to healthy control animals; no drug was detectable in the plasma, underlining the localized delivery strategy. An efficient reduction in inflammation score was obtained with a CsA dose of 1mg/mL. Therapeutic efficacy was comparable to 5-aminosalicylic acid (5-ASA), the positive control treatment in the TNBS-induced colitis model. Repetitive treatment of healthy animals with CsA nanocarriers for seven days was well tolerated with no alterations in colon histology.
Collapse
Affiliation(s)
| | | | - Christel Rousseaux
- Intestinal Biotech Development, Faculté de Médicine - Pole Recherche Amphi JK, Bd du Pr Jules Leclercq, 59045 Lille, France.
| | - Michael Möller
- Apidel SA, 29 Quai du Mont Blanc, 1201 Geneva, Switzerland
| | - Robert Gurny
- Apidel SA, 29 Quai du Mont Blanc, 1201 Geneva, Switzerland.
| | - Doris Gabriel
- Apidel SA, 29 Quai du Mont Blanc, 1201 Geneva, Switzerland.
| |
Collapse
|
61
|
Terciolo C, Dobric A, Ouaissi M, Siret C, Breuzard G, Silvy F, Marchiori B, Germain S, Bonier R, Hama A, Owens R, Lombardo D, Rigot V, André F. Saccharomyces boulardii CNCM I-745 Restores intestinal Barrier Integrity by Regulation of E-cadherin Recycling. J Crohns Colitis 2017; 11:999-1010. [PMID: 28333335 DOI: 10.1093/ecco-jcc/jjx030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Alteration in intestinal permeability is the main factor underlying the pathogenesis of many diseases affecting the gut, such as inflammatory bowel disease [IBD]. Characterization of molecules targeting the restoration of intestinal barrier integrity is therefore vital for the development of alternative therapies. The yeast Saccharomyces boulardii CNCM I-745 [Sb], used to prevent and treat antibiotic-associated infectious and functional diarrhea, may have a beneficial effect in the treatment of IBD. METHODS We analyzed the impact of Sb supernatant on tissue integrity and components of adherens junctions using cultured explants of colon from both IBD and healthy patients. To evaluate the pathways by which Sb regulates the expression of E-cadherin at the cell surface, we developed in vitro assays using human colonic cell lines, including cell aggregation, a calcium switch assay, real-time measurement of transepithelial electrical resistance [TEER] and pulse-chase experiments. RESULTS We showed that Sb supernatant treatment of colonic explants protects the epithelial morphology and maintains E-cadherin expression at the cell surface. In vitro experiments revealed that Sb supernatant enhances E-cadherin delivery to the cell surface by re-routing endocytosed E-cadherin back to the plasma membrane. This process, involving Rab11A-dependent recycling endosome, leads to restoration of enterocyte adherens junctions, in addition to the overall restoration and strengthening of intestinal barrier function. CONCLUSION These findings open new possibilities of discovering novel options for prevention and therapy of diseases that affect intestinal permeability.
Collapse
Affiliation(s)
- Chloé Terciolo
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Aurélie Dobric
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Mehdi Ouaissi
- Aix-Marseille Université, Inserm, AP-HM, UMR 911, CRO2, Marseille, France
| | - Carole Siret
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Gilles Breuzard
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Françoise Silvy
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | | | | | - Renaté Bonier
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Adel Hama
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | - Roisin Owens
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | | | - Véronique Rigot
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Frédéric André
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| |
Collapse
|
62
|
Stewart AS, Pratt-Phillips S, Gonzalez LM. Alterations in Intestinal Permeability: The Role of the "Leaky Gut" in Health and Disease. J Equine Vet Sci 2017; 52:10-22. [PMID: 31000910 DOI: 10.1016/j.jevs.2017.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
All species, including horses, suffer from alterations that increase intestinal permeability. These alterations, also known as "leaky gut," may lead to severe disease as the normal intestinal barrier becomes compromised and can no longer protect against harmful luminal contents including microbial toxins and pathogens. Leaky gut results from a variety of conditions including physical stressors, decreased blood flow to the intestine, inflammatory disease, and pathogenic infections, among others. Several testing methods exist to diagnose these alterations in both a clinical and research setting. To date, most research has focused on regulation of the host immune response due to the wide variety of factors that can potentially influence the intestinal barrier. This article serves to review the normal intestinal barrier, measurement of barrier permeability, pathogenesis and main causes of altered permeability, and highlight potential alternative therapies of leaky gut in horses while relating what has been studied in other species. Conditions resulting in barrier dysfunction and leaky gut can be a major cause of decreased performance and also death in horses. A better understanding of the intestinal barrier in disease and ways to optimize the function of this barrier is vital to the long-term health and maintenance of these animals.
Collapse
Affiliation(s)
- Amy Stieler Stewart
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | | | - Liara M Gonzalez
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Center for Gastrointestinal Biology and Disease, Large Animal Models Core, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| |
Collapse
|
63
|
Secretions of Bifidobacterium infantis and Lactobacillus acidophilus Protect Intestinal Epithelial Barrier Function. J Pediatr Gastroenterol Nutr 2017; 64:404-412. [PMID: 28230606 DOI: 10.1097/mpg.0000000000001310] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The secreted metabolites of probiotics are cytoprotective to intestinal epithelium and have been shown to attenuate inflammation and reduce gut permeability. The present study was designed to determine the protective effects of probiotic conditioned media (PCM) from Bifidobacterium infantis (BCM) and Lactobacillus acidophilus (LCM) on interleukin (IL)-1β-induced intestinal barrier compromise. METHODS The epithelial barrier was determined by measuring the transepithelial electrical resistance (TER) across a Caco-2 cell monolayer using a Transwell model. The paracellular permeability was determined by fluorescein isothiocyanate-labeled dextran flux. The expression of tight junction (TJ) proteins and nuclear factor-kappa B (NF-κB) p65 were determined using Western blot and the distribution of NF-κB p65 was determined by immunofluorescence staining. RESULTS BCM and LCM induced a dose-dependent increase in Caco-2 TER after 4 and 24 hours of incubation (P < 0.05). The maximal increase of Caco-2 TER occurred at 4 hours of treatment with a PCM concentration of 15%. Preincubation with BCM and LCM for 4 hours significantly prevented the decrease of Caco-2 TER induced by 24 hours of stimulation with 10 ng/mL IL-1β. BCM and LCM decreased paracellular permeability in both stimulated and unstimulated Caco-2 monolayers (P < 0.05). IL-1β stimulation decreased occludin expression and increased claudin-1 expression in Caco-2 cells (P < 0.05), which was prevented in cells treated with BCM or LCM. The changes of claudin-1 expression in H4 cells were similar to Caco-2 cells in response to PCM treatment and IL-1β stimulation; however, a similar response in occludin was not demonstrated. The IL-1β-induced nuclear translocation of NF-κB p65 in Caco-2 cells was prevented by pretreatment with both PCMs. CONCLUSIONS BCM and LCM protected the intestinal barrier against IL-1β stimulation by normalizing the protein expression of occludin and claudin-1 and preventing IL-1β-induced NF-κB activation in Caco-2 cells, which may be partly responsible for the preservation of intestinal permeability.
Collapse
|
64
|
Calcium-mediated oxidative stress: a common mechanism in tight junction disruption by different types of cellular stress. Biochem J 2017; 474:731-749. [PMID: 28057718 DOI: 10.1042/bcj20160679] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/10/2016] [Accepted: 01/05/2017] [Indexed: 02/07/2023]
Abstract
The role of reactive oxygen species (ROS) in osmotic stress, dextran sulfate sodium (DSS) and cyclic stretch-induced tight junction (TJ) disruption was investigated in Caco-2 cell monolayers in vitro and restraint stress-induced barrier dysfunction in mouse colon in vivo Live cell imaging showed that osmotic stress, cyclic stretch and DSS triggered rapid production of ROS in Caco-2 cell monolayers, which was blocked by depletion of intracellular Ca2+ by 1,2-bis-(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. Knockdown of CaV1.3 or TRPV6 channels blocked osmotic stress and DSS-induced ROS production and attenuated TJ disruption and barrier dysfunction. N-Acetyl l-cysteine (NAC) and l-NG-Nitroarginine methyl ester (l-NAME) blocked stress-induced TJ disruption and barrier dysfunction. NAC and l-NAME also blocked stress-induced activation of c-Jun N-terminal kinase (JNK) and c-Src. ROS was colocalized with the mitochondrial marker in stressed cells. Cyclosporin A blocked osmotic stress and DSS-induced ROS production, barrier dysfunction, TJ disruption and JNK activation. Mitochondria-targeted Mito-TEMPO blocked osmotic stress and DSS-induced barrier dysfunction and TJ disruption. Chronic restraint stress in mice resulted in the elevation of intracellular Ca2+, activation of JNK and c-Src, and disruption of TJ in the colonic epithelium. Furthermore, corticosterone administration induced JNK and c-Src activation, TJ disruption and protein thiol oxidation in colonic mucosa. The present study demonstrates that oxidative stress is a common signal in the mechanism of TJ disruption in the intestinal epithelium by different types of cellular stress in vitro and bio behavioral stress in vivo.
Collapse
|
65
|
Arsenic downregulates tight junction claudin proteins through p38 and NF-κB in intestinal epithelial cell line, HT-29. Toxicology 2017; 379:31-39. [PMID: 28115242 DOI: 10.1016/j.tox.2017.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 11/24/2022]
Abstract
Arsenic is a naturally occurring metalloid that often is found in foods and drinking water. Human exposure to arsenic is associated with the development of gastrointestinal problems such as fluid loss, diarrhea and gastritis. Arsenic is also known to induce toxic responses including oxidative stress in cells of the gastrointestinal track. Tight junctions (TJs) regulate paracellular permeability and play a barrier role by inhibiting the movement of water, solutes and microorganisms in the paracellular space. Since oxidative stress and TJ damage are known to be associated, we examined whether arsenic produces TJ damage such as downregulation of claudins in the human colorectal cell line, HT-29. To confirm the importance of oxidative stress in arsenic-induced TJ damage, effects of the antioxidant compound (e.g., N-acetylcysteine (NAC)) were also determined in cells. HT-29 cells were treated with arsenic trioxide (40μM, 12h) to observe the modified expression of TJ proteins. Arsenic decreased expression of TJ proteins (i.e., claudin-1 and claudin-5) and transepithelial electrical resistance (TEER) whereas pretreatment of NAC (5-10mM, 1h) attenuated the observed claudins downregulation and TEER. Arsenic treatment produced cellular oxidative stress via superoxide generation and lowering glutathione (GSH) levels, while NAC restored cellular GSH levels and decreased oxidative stress. Arsenic increased phosphorylation of p38 and nuclear translocation of nuclear factor-kappa B (NF-κB) p65, while NAC attenuated these intracellular events. Results demonstrated that arsenic can damage intestinal epithelial cells by proinflammatory process (oxidative stress, p38 and NF-κB) which resulted in the downregulation of claudins and NAC can protect intestinal TJs from arsenic toxicity.
Collapse
|
66
|
Shawki A, McCole DF. Mechanisms of Intestinal Epithelial Barrier Dysfunction by Adherent-Invasive Escherichia coli. Cell Mol Gastroenterol Hepatol 2017; 3:41-50. [PMID: 28174756 PMCID: PMC5247418 DOI: 10.1016/j.jcmgh.2016.10.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/14/2016] [Indexed: 12/12/2022]
Abstract
Pathobiont expansion, such as that of adherent-invasive Escherichia coli (AIEC), is an emerging factor associated with inflammatory bowel disease. The intestinal epithelial barrier is the first line of defense against these pathogens. Inflammation plays a critical role in altering the epithelial barrier and is a major factor involved in promoting the expansion and pathogenesis of AIEC. AIEC in turn can exacerbate intestinal epithelial barrier dysfunction by targeting multiple elements of the barrier. One critical element of the epithelial barrier is the tight junction. Increasing evidence suggests that AIEC may selectively target protein components of tight junctions, leading to increased barrier permeability. This may represent one mechanism by which AIEC could contribute to the development of inflammatory bowel disease. This review article discusses potential mechanisms by which AIEC can disrupt epithelial tight junction function and intestinal barrier function.
Collapse
Key Words
- AIEC, adherent-invasive Escherichia coli
- AJ, adherens junction
- AJC, apical junctional complex
- BP, bacterial peptidoglycans
- CD, Crohn’s disease
- CEACAM6, carcinoembryonic antigen–related cell-adhesion molecule
- IBD, inflammatory bowel disease
- IEC, intestinal epithelial cell
- IFN, interferon
- IL, interleukin
- Inflammatory Bowel Disease
- Intestinal Permeability
- JAM-A, junctional adhesion molecule-A
- LPF, long polar fimbriae
- MLC, myosin light chain
- MLCK, myosin light chain kinase
- NF-κB, nuclear factor-κB
- NOD2, nucleotide-binding oligomerization domain 2
- PDZ, PSD95-DlgA-zonula occludens-1 homology domain
- TJ, tight junction
- TNF, tumor necrosis factor
- Tight Junctions
- UC, ulcerative colitis
- ZO, zonula occludens
Collapse
Affiliation(s)
| | - Declan F. McCole
- Division of Biomedical Sciences, University of California Riverside, Riverside, California
| |
Collapse
|
67
|
Abstract
IQGAP1 is a scaffold protein involved in the assembly of adherens junctions. Our work has recently revealed a novel role for IQGAP1 in the regulation of tight junctions (TJ) through differential recruitment of claudins to the nascent TJ. Here, we discuss the potential mechanisms of this regulation, including IQGAP1 effects on CDC42, and IQGAP1 interactions with sorting/trafficking molecules (e.g. Exo70). Given the many roles of IQGAP1 and the large number of interacting partners, we focus our discussion of these functions in the context of junction formation, trafficking, growth factor signaling and cancer. We also propose a potential role for IQGAP1 in regulating epithelial integrity and compartmentalized signaling in epithelia.
Collapse
Affiliation(s)
- Barbara E Tanos
- a Division of Cancer Therapeutics, The Institute of Cancer Research , London , UK
| | - Charles Yeaman
- b Department of Anatomy and Cell Biology , The University of Iowa , Iowa City , IA , USA
| | - Enrique Rodriguez-Boulan
- c Department of Ophthalmology , Margaret Dyson Vision Research Institute, Weill Cornell Medical College , New York , NY , USA.,d Department of Cell and Developmental Biology , Weill Cornell Medical College , New York , NY , USA
| |
Collapse
|
68
|
Tian Z, Liu X, Dai R, Xiao Y, Wang X, Bi D, Shi D. Enterococcus faecium HDRsEf1 Protects the Intestinal Epithelium and Attenuates ETEC-Induced IL-8 Secretion in Enterocytes. Mediators Inflamm 2016; 2016:7474306. [PMID: 27890970 PMCID: PMC5116501 DOI: 10.1155/2016/7474306] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/21/2016] [Accepted: 09/28/2016] [Indexed: 12/25/2022] Open
Abstract
The probiotic Enterococcus faecium HDRsEf1 (Ef1) has been shown to have positive effects on piglet diarrhoea, but the mechanism has not yet been elucidated. In this study, using the IPEC-J2 cell line to mimic intestinal epithelial cells and enterotoxigenic Escherichia coli (ETEC) K88ac as a representative intestinal pathogen, the mechanism underlying Ef1 protection against an enteropathogen was investigated. The results demonstrated that Ef1 was effective in displacing K88ac from the IPEC-J2 cell layer. Moreover, Ef1 and its cell-free supernatant (S-Ef1) modulate IL-8 released by IPEC-J2 cells. Ef1 and its cell-free supernatant showed the potential to protect enterocytes from an acute inflammatory response. In addition, Ef1 and its cell-free supernatant increased the transepithelial electrical resistance (TEER) of the enterocyte monolayer, thus strengthening the intestinal barrier against ETEC. These results may contribute to the development of therapeutic interventions using Ef1 in intestinal disorders of piglets.
Collapse
Affiliation(s)
- Zhongyuan Tian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiaofang Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ran Dai
- College of Animal Science and Technology, Agricultural University of Hebei, Baoding 071000, China
| | - Yuncai Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Deshi Shi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
69
|
Cazarin CBB, Rodriguez-Nogales A, Algieri F, Utrilla MP, Rodríguez-Cabezas ME, Garrido-Mesa J, Guerra-Hernández E, Braga PADC, Reyes FGR, Maróstica MR, Gálvez J. Intestinal anti-inflammatory effects of Passiflora edulis peel in the dextran sodium sulphate model of mouse colitis. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.08.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
70
|
Fermented Pueraria Lobata extract ameliorates dextran sulfate sodium-induced colitis by reducing pro-inflammatory cytokines and recovering intestinal barrier function. Lab Anim Res 2016; 32:151-159. [PMID: 27729931 PMCID: PMC5057003 DOI: 10.5625/lar.2016.32.3.151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/26/2022] Open
Abstract
Inflammatory bowel disease is a chronic inflammatory disorder occurring in the gastrointestinal track. However, the efficacy of current therapeutic strategies has been limited and accompanied by side effects. In order to eliminate the limitations, herbal medicines have recently been developed for treatment of IBD. Peuraria Lobata (Peuraria L.) is one of the traditional herbal medicines that have anti-inflammatory effects. Bioavailability of Peuraria L., which is rich in isoflavones, is lower than that of their fermented forms. In this study, we generated fermented Peuraria L. extracts (fPue) and investigated the role of fPue in inflammation and intestinal barrier function in vitro and in vivo. As the mice or intestinal epithelial cells were treated with DSS/fPue, mRNA expression of pro-inflammatory cytokines was reduced and the architecture and expression of tight junction proteins were recovered, compared to the DSS-treated group. In summary, fPue treatment resulted in amelioration of DSS-induced inflammation in the colon, and the disrupted intestinal barrier was recovered as the expression and architecture of tight junction proteins were retrieved. These results suggest that use of fPue could be a new therapeutic strategy for treatment of IBD.
Collapse
|
71
|
van den Nieuwboer M, van Hemert S, Claassen E, de Vos WM. Lactobacillus plantarum WCFS1 and its host interaction: a dozen years after the genome. Microb Biotechnol 2016; 9:452-65. [PMID: 27231133 PMCID: PMC4919987 DOI: 10.1111/1751-7915.12368] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus plantarum WCFS1 is one of the best studied Lactobacilli, notably as its genome was unravelled over 12 years ago. L. plantarum WCFS1 can be grown to high densities, is amenable to genetic transformation and highly robust with a relatively high survival rate during the gastrointestinal passage. In this review, we present and discuss the main insights provided by the functional genomics research on L. plantarum WCFS1 with specific attention for the molecular mechanisms related to its interaction with the human host and its potential to modify the immune system, and induce other health-related benefits. Whereas most insight has been gained in mouse and other model studies, only five human studies have been reported with L. plantarum WCFS1. Hence NCIMB 8826 (the parental strain of L. plantarum WCFS1) in human trials as to capitalize on the wealth of knowledge that is summarized here.
Collapse
Affiliation(s)
| | | | - Eric Claassen
- Athena Institute, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology and Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
72
|
DiGuilio KM, Mercogliano CM, Born J, Ferraro B, To J, Mixson B, Smith A, Valenzano MC, Mullin JM. Sieving characteristics of cytokine- and peroxide-induced epithelial barrier leak: Inhibition by berberine. World J Gastrointest Pathophysiol 2016; 7:223-234. [PMID: 27190695 PMCID: PMC4867402 DOI: 10.4291/wjgp.v7.i2.223] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To study whether the inflammatory bowel disease (IBD) colon which exhibits varying severity and cytokine levels across its mucosa create varying types of transepithelial leak.
METHODS: We examined the effects of tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin-1-β (IL1β) and hydrogen peroxide (H2O2) - singly and in combinations - on barrier function of CACO-2 cell layers. Our focus was on the type (not simply the magnitude) of transepithelial leak generated by these agents as measured by transepithelial electrical resistance (TER) and transepithelial flux of 14C-D-mannitol, 3H-Lactulose and 14C-Polyethylene glycol as radiolabeled probe molecules. The isoquinoline alkaloid, berberine, was then examined for its ability to reduce specific types of transepithelial leak.
RESULTS: Exposure to TNF-α alone (200 ng/mL; 48 h) induced a 50% decrease in TER, i.e., increased leak of Na+ and Cl- - with only a marginal but statistically significant increase in transepithelial leak of 14C-mannitol (Jm). Exposure to TNF-α + IFN-γ (200 ng/mL; 48 h) + IL1β (50 ng/mL; 48 h) did not increase the TER change (from TNF-α alone), but there was now a 100% increase in Jm. There however was no increase in transepithelial leak of two larger probe molecules, 3H-lactulose and 14C-polyethylene glycol (PEG). However, exposure to TNF-α + IFN-γ + IL1β followed by a 5 h exposure to 2 mmol/L H2O2 resulted in a 500% increase in 14C-PEG leak as well as leak to the luminal mitogen, epidermal growth factor.
CONCLUSION: This model of graded transepithelial leak is useful in evaluating therapeutic agents reducing IBD morbidity by reducing barrier leak to various luminal substances.
Collapse
|
73
|
Impact of gut microbiota on diabetes mellitus. DIABETES & METABOLISM 2016; 42:303-315. [PMID: 27179626 DOI: 10.1016/j.diabet.2016.04.004] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 02/07/2023]
Abstract
Various functions of the gut are regulated by sophisticated interactions among its functional elements, including the gut microbiota. These microorganisms play a crucial role in gastrointestinal mucosa permeability. They control the fermentation and absorption of dietary polysaccharides to produce short-chain fatty acids, which may explain their importance in the regulation of fat accumulation and the subsequent development of obesity-related diseases, suggesting that they are a crucial mediator of obesity and its consequences. In addition, gut bacteria play a crucial role in the host immune system, modulation of inflammatory processes, extraction of energy from the host diet and alterations of human gene expression. Dietary modulation of the human colonic microbiota has been shown to confer a number of health benefits to the host. Simple therapeutic strategies targeted at attenuating the progression of chronic low-grade inflammation and insulin resistance are urgently required to prevent or slow the development of diabetes in susceptible individuals. The main objective of this review is to address the pathogenic association between gut microbiota and diabetes, and to explore any novel related therapeutic targets. New insights into the role of the gut microbiota in diabetes could lead to the development of integrated strategies using probiotics to prevent and treat these metabolic disorders.
Collapse
|
74
|
Landy J, Ronde E, English N, Clark SK, Hart AL, Knight SC, Ciclitira PJ, Al-Hassi HO. Tight junctions in inflammatory bowel diseases and inflammatory bowel disease associated colorectal cancer. World J Gastroenterol 2016; 22:3117-3126. [PMID: 27003989 PMCID: PMC4789987 DOI: 10.3748/wjg.v22.i11.3117] [Citation(s) in RCA: 338] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/19/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases are characterised by inflammation that compromises the integrity of the epithelial barrier. The intestinal epithelium is not only a static barrier but has evolved complex mechanisms to control and regulate bacterial interactions with the mucosal surface. Apical tight junction proteins are critical in the maintenance of epithelial barrier function and control of paracellular permeability. The characterisation of alterations in tight junction proteins as key players in epithelial barrier function in inflammatory bowel diseases is rapidly enhancing our understanding of critical mechanisms in disease pathogenesis as well as novel therapeutic opportunities. Here we give an overview of recent literature focusing on the role of tight junction proteins, in particular claudins, in inflammatory bowel diseases and inflammatory bowel disease associated colorectal cancer.
Collapse
|
75
|
Gut Microbiota Dysbiosis as Risk and Premorbid Factors of IBD and IBS Along the Childhood-Adulthood Transition. Inflamm Bowel Dis 2016; 22:487-504. [PMID: 26588090 DOI: 10.1097/mib.0000000000000602] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal disorders, although clinically heterogeneous, share pathogenic mechanisms, including genetic susceptibility, impaired gut barrier function, altered microbiota, and environmental triggers (infections, social and behavioral factors, epigenetic control, and diet). Gut microbiota has been studied for inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) in either children or adults, while modifiable gut microbiota features, acting as risk and premorbid factors along the childhood-adulthood transition, have not been thoroughly investigated so far. Indeed, the relationship between variations of the entire host/microbiota/environmental scenario and clinical phenotypes is still not fully understood. In this respect, tracking gut dysbiosis grading may help deciphering host phenotype-genotype associations and microbiota shifts in an integrated top-down omics-based approach within large-scale pediatric and adult case-control cohorts. Large-scale gut microbiota signatures and host inflammation patterns may be integrated with dietary habits, under genetic and epigenetic constraints, providing gut dysbiosis profiles acting as risk predictors of IBD or IBS in preclinical cases. Tracking dysbiosis supports new personalized/stratified IBD and IBS prevention programmes, generating Decision Support System tools. They include (1) high risk or flare-up recurrence -omics-based dysbiosis profiles; (2) microbial and molecular biomarkers of health and disease; (3) -omics-based pipelines for laboratory medicine diagnostics; (4) health apps for self-management of score-based dietary profiles, which can be shared with clinicians for nutritional habit and lifestyle amendment; (5) -omics profiling data warehousing and public repositories for IBD and IBS profile consultation. Dysbiosis-related indexes can represent novel laboratory and clinical medicine tools preventing or postponing the disease, finally interfering with its natural history.
Collapse
|
76
|
Garg S, Zheng J, Wang J, Authier S, Pouliot M, Hauer-Jensen M. Segmental Differences in Radiation-Induced Alterations of Tight Junction-Related Proteins in Non-Human Primate Jejunum, Ileum and Colon. Radiat Res 2015; 185:50-9. [PMID: 26720804 DOI: 10.1667/rr14157.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dysfunction of the intestinal epithelial barrier and leakage of luminal antigens and bacteria across the barrier have been linked to various human diseases. Intestinal permeability is regulated by intercellular structures, termed "tight junction" (Tj), which are disrupted after total-body irradiation (TBI). In this study, we investigated radiation-induced alterations in Tj-related proteins in the jejunum, ileum and colon of a non-human primate (NHP) model. NHPs were total-body irradiated with 6.7 and 7.4 Gy and intestines were procured at day 4, 7 and 12. Radiation exposure was found to induce significant increases in claudin-10 mRNA early (day 4) in all three gut segments and claudin-4 mRNA levels were repressed through day 12. TNF-alpha was highly induced in the jejunum and colon at early time points, but little induction was found in the ileum. Claudin-1 was induced only in the colon on day 4 postirradiation. Unlike the colon and jejunum, the ileum levels of claudin-7 were significantly downregulated through day 12 postirradiation. Western blot analysis revealed increased levels of claudin-2 on day 4 and of JAM-1 on day 7 postirradiation in all three gut segments. E-cadherin was downregulated on day 4 postirradiation in all segments, but remained reduced in the jejunum only until day 12. Taken together, these data suggest that exposure to radiation causes segment-specific alterations in the expression of Tj-related proteins. Interruption of Tjs may be a key factor contributing to injury to the intestinal mucosal barrier and increased intestinal permeability.
Collapse
Affiliation(s)
- Sarita Garg
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junying Zheng
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junru Wang
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | - Martin Hauer-Jensen
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas;,c Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
77
|
Mullin JM, Skrovanek SM, Ramalingam A, DiGuilio KM, Valenzano MC. Methionine restriction fundamentally supports health by tightening epithelial barriers. Ann N Y Acad Sci 2015; 1363:59-67. [PMID: 26646941 DOI: 10.1111/nyas.12955] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/16/2015] [Accepted: 10/16/2015] [Indexed: 12/21/2022]
Abstract
Dietary methionine restriction (MR) has been found to affect one of the most primary tissue-level functions of an organism: the efficiency with which the epithelial linings of major organs separate the fluid compartments that they border. This process, epithelial barrier function, is basic for proper function of all organs, including the lung, liver, gastrointestinal tract, reproductive tract, blood-brain barrier, and kidney. Specifically, MR has been found to modify the protein composition of tight junctional complexes surrounding individual epithelial cells in a manner that renders the complexes less leaky. This has been observed in both a renal epithelial cell culture model and in gastrointestinal tissue. In both cases, MR increased the transepithelial electrical resistance across the epithelium, while decreasing passive leak of small nonelectrolytes. However, the specific target protein modifications involved were unique to each case. Overall, this provides an example of the primary level on which MR functions to modify, and improve, an organism.
Collapse
Affiliation(s)
- James M Mullin
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.,Division of Gastroenterology, Lankenau Medical Center, Wynnewood, Pennsylvania
| | | | | | | | - Mary C Valenzano
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| |
Collapse
|
78
|
Rodríguez-Feo JA, Puerto M, Fernández-Mena C, Verdejo C, Lara JM, Díaz-Sánchez M, Álvarez E, Vaquero J, Marín-Jiménez I, Bañares R, Menchén L. A new role for reticulon-4B/NOGO-B in the intestinal epithelial barrier function and inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 2015; 308:G981-93. [PMID: 25907690 DOI: 10.1152/ajpgi.00309.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 04/17/2015] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel disease (IBD) is characterized by an impaired intestinal barrier function. We aimed to investigate the role of reticulon-4B (RTN-4B/NOGO-B), a structural protein of the endoplasmic reticulum, in intestinal barrier function and IBD. We used immunohistochemistry, confocal microscopy, real-time PCR, and Western blotting to study tissue distribution and expression levels of RTN-4B/NOGO-B in control and IBD samples from mouse and humans. We also targeted RTN-4B/NOGO-B using siRNAs in cultured human intestinal epithelial cell (IECs). Epithelial barrier permeability was assessed by transepithelial electrical resistance (TEER) measurement. RTN-4B/NOGO-B is expressed in the intestine mainly by IECs. Confocal microscopy revealed a colocalization of RTN-4B, E-cadherin, and polymerized actin fibers in tissue and cultured IECs. RTN-4B mRNA and protein expression were lower in the colon of IL-10(-/-) compared with wild-type mice. Colocalization of RTN-4B/E-cadherin/actin was reduced in the colon of IL-10(-/-) mice. Analysis of endoscopic biopsies from IBD patients showed a significant reduction of RTN-4B/NOGO-B expression in inflamed mucosa compared with control. Treatment of IECs with H2O2 reduced TEER values and triggered phosphorylation of RTN-4B in serine 107 residues as well as downregulation of RTN-4B expression. Acute RTN-4B/NOGO-B knockdown by siRNAs resulted in a decreased TEER values and reduction of E-cadherin and α-catenin expression and in the amount of F-actin-rich filaments in IECs. Epithelial RTN-4B/NOGO-B was downregulated in human and experimental IBD. RTN-4B participates in the intestinal epithelial barrier function, most likely via its involvement in E-cadherin, α-catenin expression, and actin cytoskeleton organization at sites of cell-to-cell contacts.
Collapse
Affiliation(s)
- Juan Antonio Rodríguez-Feo
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Marta Puerto
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Carolina Fernández-Mena
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Cristina Verdejo
- Servicio de Aparato Digestivo, Hospital General de Ciudad Real, Ciudad Real, Spain
| | - José Manuel Lara
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, Madrid, Spain; and
| | - María Díaz-Sánchez
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, Madrid, Spain; and
| | - Emilio Álvarez
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, Madrid, Spain; and
| | - Javier Vaquero
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Ignacio Marín-Jiménez
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Luis Menchén
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
79
|
Lau WL, Liu SM, Pahlevan S, Yuan J, Khazaeli M, Ni Z, Chan JY, Vaziri ND. Role of Nrf2 dysfunction in uremia-associated intestinal inflammation and epithelial barrier disruption. Dig Dis Sci 2015; 60:1215-22. [PMID: 25399330 DOI: 10.1007/s10620-014-3428-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gut inflammation is prevalent in chronic kidney disease (CKD) and likely contributes to systemic inflammation via disruption of the epithelial tight junction with subsequent endotoxin and bacterial translocation. AIMS To study the expression profile of inflammatory and tight junction proteins in the colon from CKD rats compared to healthy controls, and demonstrate the role of Nrf2 (transcription factor nuclear factor erythroid 2-related factor 2) using a potent Nrf2 activator. METHODS CKD was induced via 5/6 nephrectomy in Sprague-Dawley rats, and dh404 (2 mg/kg/day) was used to study the effects of systemic Nrf2 activation. The experimental groups included sham, CKD and CKD+ dh404 rats. Blood and colon tissues were analyzed after a 10-week study period. RESULTS Colon from CKD rats showed histological evidence of colitis, depletion of epithelial tight junction proteins, significant reduction of Nrf2 and its measured target gene products (NQO1, catalase, and CuZn SOD), activation of NFkB, and upregulation of pro-inflammatory molecules (COX-2, MCP-1, iNOS, and gp91(phox)). Treatment with dh404 attenuated colonic inflammation, restored Nrf2 activity and levels of NQO1, catalase and CuZn SOD, decreased NFkB and lowered expression of COX-2, MCP-1, iNOS, and gp91(phox). This was associated with restoration of colonic epithelial tight junction proteins (occludin and claudin-1). CONCLUSIONS CKD rats exhibited colitis, disruption of colonic epithelial tight junction, activation of inflammatory mediators, and impairment of Nrf2 pathway. Treatment with an Nrf2 activator restored Nrf2 activity, attenuated colonic inflammation, and restored epithelial tight junction proteins.
Collapse
Affiliation(s)
- Wei Ling Lau
- Division of Nephrology and Hypertension, Department of Medicine, University of California-Irvine, C362 Medical Sciences I, Irvine, CA, 92697, USA,
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Cravo AS, Carter E, Erkan M, Harvey E, Furutani-Seiki M, Mrsny R. Hippo pathway elements Co-localize with Occludin: A possible sensor system in pancreatic epithelial cells. Tissue Barriers 2015; 3:e1037948. [PMID: 26451343 PMCID: PMC4574897 DOI: 10.1080/21688370.2015.1037948] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/22/2015] [Accepted: 03/27/2015] [Indexed: 12/14/2022] Open
Abstract
External adherens junction-based cell-cell contacts involving E-cadherin interactions function to sense planar cell status and modulate epithelial cell proliferation through Hippo (Hpo) and non-canonical Wnt pathways signaling. We hypothesized these regulatory processes should also be sensitive to a similar cell-cell contact sensor associated with apical-basal polarity events at epithelial surfaces. We used 2 human pancreatic cancer cell lines to explore this hypothesis: one with the capacity to form functional tight junction structures and polarize (HPAFII) and one lacking this capacity (AsPc1). Occludin (Ocln), a tetraspanning protein associated with TJ structures and capable of establishing external cell-cell contacts, was observed to partially co-localize with Hpo elements YAP (c-yes associated protein) and TEAD (TEA-dependent), which function to drive a proliferative transcription program. Treatment with dobutamine, known to affect YAP, was shown to suppress proliferation in an Ocln-dependent manner. Blockade of protein kinase C-zeta (PKC-ζ) diminished transepithelial electrical resistance (TER) of HPAFII monolayers that was not corrected by dobutamine treatment while the loss of TER resulting from inhibition of ROCK1 could be partially recovered. Examination of normal and cancerous human pancreatic biopsies showed that the cellular localization of Ocln, c-Yes, YAP, and TEAD were similar to HPAFII for normal cells and AsPc1 for cancerous cells. Together, these results suggest a link between Hpo and signals emanating from cell-cell contacts involving Ocln that may regulate pancreatic cell proliferation through the coordination of planar cell polarity with apical-basal polarity events.
Collapse
Affiliation(s)
- Ana Santos Cravo
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | - Edward Carter
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | - Mert Erkan
- Department of Surgery; Koc University School of Medicine ; Istanbul, Turkey
| | - Emma Harvey
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | | | - Randall Mrsny
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| |
Collapse
|
81
|
Morampudi V, Conlin VS, Dalwadi U, Wu X, Marshall KC, Nguyen C, Vallance BA, Jacobson K. Vasoactive intestinal peptide prevents PKCε-induced intestinal epithelial barrier disruption during EPEC infection. Am J Physiol Gastrointest Liver Physiol 2015; 308:G389-402. [PMID: 25501546 DOI: 10.1152/ajpgi.00195.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We previously showed that vasoactive intestinal peptide (VIP) protects against bacterial pathogen-induced epithelial barrier disruption and colitis, although the mechanisms remain poorly defined. The aim of the current study was to identify cellular pathways of VIP-mediated protection with use of pharmacological inhibitors during enteropathogenic Escherichia coli (EPEC) infection of Caco-2 cell monolayers and during Citrobacter rodentium-induced colitis. EPEC-induced epithelial barrier disruption involved the PKC pathway but was independent of functional cAMP, Rho, and NF-κB pathways. VIP mediated its protective effects by inhibiting EPEC-induced PKC activity and increasing expression of the junctional protein claudin-4. Short-term treatment with TPA, which is known to activate PKC, was inhibited by VIP pretreatment, while PKC degradation via long-term treatment with TPA mimicked the protective actions of VIP. Immunostaining for specific PKC isotypes showed upregulated expression of PKCθ and PKCε during EPEC infection. Treatment with specific inhibitors revealed a critical role for PKCε in EPEC-induced barrier disruption. Furthermore, activation of PKCε and loss of barrier integrity correlated with claudin-4 degradation. In contrast, inhibition of PKCε by VIP pretreatment or the PKCε inhibitor maintained membrane-bound claudin-4 levels, along with barrier function. Finally, in vivo treatment with the PKCε inhibitor protected mice from C. rodentium-induced colitis. In conclusion, EPEC infection increases intracellular PKCε levels, leading to decreased claudin-4 levels and compromising epithelial barrier integrity. VIP inhibits PKCε activation, thereby attenuating EPEC-induced barrier disruption.
Collapse
Affiliation(s)
- V Morampudi
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - V S Conlin
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - U Dalwadi
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - X Wu
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - K C Marshall
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - C Nguyen
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - B A Vallance
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - K Jacobson
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
82
|
Tanos BE, Perez Bay AE, Salvarezza S, Vivanco I, Mellinghoff I, Osman M, Sacks DB, Rodriguez-Boulan E. IQGAP1 controls tight junction formation through differential regulation of claudin recruitment. J Cell Sci 2015; 128:853-62. [PMID: 25588839 DOI: 10.1242/jcs.118703] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IQGAP1 is a scaffolding protein previously implicated in adherens junction formation. However, its role in the establishment or maintenance of tight junctions (TJs) has not been explored. We hypothesized that IQGAP1 could regulate TJ formation by modulating the expression and/or localization of junctional proteins, and we systematically tested this hypothesis in the model Madin-Darby canine kidney (MDCK) cell line. We find that IQGAP1 silencing enhances a transient increase in transepithelial electrical resistance (TER) observed during the early stages of TJ formation (Cereijido et al., 1978). Quantitative microscopy and biochemical experiments suggest that this effect of IQGAP1 on TJ assembly is accounted for by reduced expression and TJ recruitment of claudin 2, and increased TJ recruitment of claudin 4. Furthermore, we show that IQGAP1 also regulates TJ formation through its interactor CDC42, because IQGAP1 knockdown increases the activity of the CDC42 effector JNK and dominant-negative CDC42 prevents the increase in TER caused by IQGAP1 silencing. Hence, we provide evidence that IQGAP1 modulates TJ formation by a twofold mechanism: (1) controlling the expression and recruitment of claudin 2 and recruitment of claudin 4 to the TJ, and (2) transient inhibition of the CDC42-JNK pathway.
Collapse
Affiliation(s)
- Barbara E Tanos
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Andres E Perez Bay
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Susana Salvarezza
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Igor Vivanco
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Ingo Mellinghoff
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Mahasin Osman
- Department of Molecular Pharmacology, Physiology and Biotechnology, Division of Biology and Medicine, Alpert School of Medicine, Brown University, Providence, RI 02912, USA
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Enrique Rodriguez-Boulan
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065, USA Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
83
|
|
84
|
Effect of surface chemistry on nanoparticle interaction with gastrointestinal mucus and distribution in the gastrointestinal tract following oral and rectal administration in the mouse. J Control Release 2014; 197:48-57. [PMID: 25449804 DOI: 10.1016/j.jconrel.2014.10.026] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/13/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023]
Abstract
It is believed that mucoadhesive surface properties on particles delivered to the gastrointestinal (GI) tract improve oral absorption or local targeting of various difficult-to-deliver drug classes. To test the effect of nanoparticle mucoadhesion on distribution of nanoparticles in the GI tract, we orally and rectally administered nano- and microparticles that we confirmed possessed surfaces that were either strongly mucoadhesive or non-mucoadhesive. We found that mucoadhesive particles (MAP) aggregated in mucus in the center of the GI lumen, far away from the absorptive epithelium, both in healthy mice and in a mouse model of ulcerative colitis (UC). In striking contrast, water absorption by the GI tract rapidly and uniformly transported non-mucoadhesive mucus-penetrating particles (MPP) to epithelial surfaces, including reaching the surfaces between villi in the small intestine. When using high gavage fluid volumes or injection into ligated intestinal loops, common methods for assessing oral drug and nanoparticle absorption, we found that both MAP and MPP became well-distributed throughout the intestine, indicating that the barrier properties of GI mucus were compromised. In the mouse colorectum, MPP penetrated into mucus in the deeply in-folded surfaces to evenly coat the entire epithelial surface. Moreover, in a mouse model of UC, MPP were transported preferentially into the disrupted, ulcerated tissue. Our results suggest that delivering drugs in non-mucoadhesive MPP is likely to provide enhanced particle distribution, and thus drug delivery, in the GI tract, including to ulcerated tissues.
Collapse
|
85
|
Matricon J, Barnich N, Ardid D. Immunopathogenesis of inflammatory bowel disease. SELF NONSELF 2014; 1:299-309. [PMID: 21487504 DOI: 10.4161/self.1.4.13560] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 09/06/2010] [Accepted: 09/07/2010] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is a group of idiopathic, chronic and relapsing inflammatory conditions of the gastrointestinal tract. Familial and epidemiological studies have stressed the involvement of genetic factors and have also shown the critical role of environmental factors such as sanitation and hygiene in the development of IBD. However, the molecular mechanisms of intestinal inflammation in IBD have long remained unknown. In recent years, the study of susceptibility genes involved in the detection of bacterial components and in the regulation of the host immune response has shed light onto the potential role of intestinal pathogens and gut flora in IBD immunobiology. This review presents current knowledge on intestinal epithelial barrier alterations and on dysfunction of mucosal innate and acquired immune responses in IBD. The data support the etiological hypothesis which argues that pathogenic intestinal bacteria and/or infectious agents initiate and perpetuate the inflammation of the gut through disruption of tolerance towards the commensal microbiota in an individual with genetic vulnerability.
Collapse
Affiliation(s)
- Julien Matricon
- Clermont Université; Université d'Auvergne; Pharmacologie Fondamentale et Clinique de la Douleur; Laboratoire de Pharmacologie Médicale; Inserm U 766; Clermont-Ferrand, France
| | | | | |
Collapse
|
86
|
Ohta H, Sunden Y, Yokoyama N, Osuga T, Lim SY, Tamura Y, Morishita K, Nakamura K, Yamasaki M, Takiguchi M. Expression of apical junction complex proteins in duodenal mucosa of dogs with inflammatory bowel disease. Am J Vet Res 2014; 75:746-51. [PMID: 25061706 DOI: 10.2460/ajvr.75.8.746] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the expression of tight junction and adherens junction proteins in duodenal mucosa samples of dogs with inflammatory bowel disease (IBD). ANIMALS 12 dogs with IBD and 6 healthy control Beagles. PROCEDURES Duodenal mucosa biopsy samples were endoscopically obtained from dogs with IBD and healthy control Beagles. The expression of claudin-1, -2, -3, -4, -5, -7, and -8; E-cadherin; and β-catenin in the duodenal mucosa samples was determined by means of immunoblotting. The subcellular localization of E-cadherin in the duodenal mucosa samples was determined with immunofluorescence microscopy. RESULTS The expression of each claudin and β-catenin was not significantly different between control dogs and dogs with IBD. However, expression of E-cadherin was significantly lower in duodenal mucosa samples of dogs with IBD than it was in samples obtained from healthy control dogs. Results of immunofluorescence microscopy indicated decreased intensity of E-cadherin labeling in the tips of villi in duodenal mucosa samples obtained from 6 dogs with IBD, compared with staining intensity for other dogs. CONCLUSIONS AND CLINICAL RELEVANCE Results of this study indicated expression of claudin-1, -2, -3, -4, -5, -7, and -8 and β-catenin was not significantly different between duodenal mucosa samples obtained from control dogs and those obtained from dogs with IBD. However, E-cadherin expression was significantly lower in the villus epithelium in duodenal mucosa samples obtained from dogs with IBD versus samples obtained from control dogs, which suggested that decreased expression of that protein has a role in the pathogenesis of IBD in dogs.
Collapse
Affiliation(s)
- Hiroshi Ohta
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Sziksz E, Pap D, Veres G, Fekete A, Tulassay T, Vannay &A. Involvement of heat shock proteins in gluten-sensitive enteropathy. World J Gastroenterol 2014; 20:6495-6503. [PMID: 24914370 PMCID: PMC4047334 DOI: 10.3748/wjg.v20.i21.6495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/12/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023] Open
Abstract
Gluten-sensitive enteropathy, also known as coeliac disease (CD), is an autoimmune disorder occurring in genetically susceptible individuals that damages the small intestine and interferes with the absorption of other nutrients. As it is triggered by dietary gluten and related prolamins present in wheat, rye and barley, the accepted treatment for CD is a strict gluten-free diet. However, a complete exclusion of gluten-containing cereals from the diet is often difficult, and new therapeutic strategies are urgently needed. A class of proteins that have already emerged as drug targets for other autoimmune diseases are the heat shock proteins (HSPs), which are highly conserved stress-induced chaperones that protect cells against harmful extracellular factors. HSPs are expressed in several tissues, including the gastrointestinal tract, and their levels are significantly increased under stress circumstances. HSPs exert immunomodulatory effects, and also play a crucial role in the maintenance of epithelial cell structure and function, as they are responsible for adequate protein folding, influence the degradation of proteins and cell repair processes after damage, and modulate cell signalling, cell proliferation and apoptosis. The present review discusses the involvement of HSPs in the pathophysiology of CD. Furthermore, HSPs may represent a useful therapeutic target for the treatment of CD due to the cytoprotective, immunomodulatory, and anti-apoptotic effects in the intestinal mucosal barrier.
Collapse
|
88
|
Si-jun-zi decoction treatment promotes the restoration of intestinal function after obstruction by regulating intestinal homeostasis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:928579. [PMID: 24876882 PMCID: PMC4020469 DOI: 10.1155/2014/928579] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 03/23/2014] [Accepted: 03/23/2014] [Indexed: 11/17/2022]
Abstract
Intestinal obstruction is a common disease requiring abdominal surgery with significant morbidity and mortality. Currently, an effective medical treatment for obstruction, other than surgical resection or decompression, does not exist. Si-Jun-Zi Decoction is a famous Chinese medicine used to replenish qi and invigorate the functions of the spleen. Modern pharmacological studies show that this prescription can improve gastrointestinal function and strengthen immune function. In this study, we investigated the effects of a famous Chinese herbal formula, Si-Jun-Zi Decoction, on the restoration of intestinal function after the relief of obstruction in a rabbit model. We found that Si-Jun-Zi Decoction could reduce intestinal mucosal injury while promoting the recovery of the small intestine. Further, Si-Jun-Zi Decoction could regulate the intestinal immune system. Our results suggest that Si-Jun-Zi Decoction promotes the restoration of intestinal function after obstruction by regulating intestinal homeostasis. Our observations indicate that Si-Jun-Zi Decoction is potentially a therapeutic drug for intestinal obstruction.
Collapse
|
89
|
Wilcz-Villega E, McClean S, O'Sullivan M. Reduced E-cadherin expression is associated with abdominal pain and symptom duration in a study of alternating and diarrhea predominant IBS. Neurogastroenterol Motil 2014; 26:316-25. [PMID: 24286617 DOI: 10.1111/nmo.12262] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/23/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Increased intestinal permeability and altered expression of tight junction (TJ) proteins may be implicated in the pathogenesis of irritable bowel syndrome (IBS). This study aimed to investigate the expression of adherens junction (AJ) protein E-cadherin and TJ proteins zonula occludens (ZO)-1 and claudin (CLD)-1 and associations with IBS symptoms. METHODS Junctional proteins were immunostained in cecal biopsy tissue of Rome II IBS patients (n = 34) comprising both alternating (IBS-A) and diarrhea predominant (IBS-D) subtypes, and controls (n = 12). IBS symptom duration, abdominal pain severity and stool frequency were assessed for IBS patients. Protein expression was determined by immunofluorescence. KEY RESULTS E-cadherin and ZO-1 protein expression was significantly lower (p = 0.03 and p = 0.016, respectively) in the cecal surface epithelium of the IBS group comprising both IBS-A and IBS-D subtypes. CLD-1 expression was not significantly altered compared with controls. On subtype analysis, ZO-1 expression was significantly reduced in both IBS-A and IBS-D compared with controls, whereas E-cadherin was reduced only in IBS-A. Lower E-cadherin expression was associated with longer symptoms duration specifically in IBS-A patients (rs = -0.76, p = 0.004). Reduced E-cadherin associated with abdominal pain severity in the overall IBS group (rs = -0.36, p = 0.041), but this association was unrelated to IBS subtype. CONCLUSIONS & INFERENCES E-cadherin protein expression in the cecum was significantly lower in IBS-A compared with controls and associated with longstanding symptoms. E-cadherin was further associated with abdominal pain severity in the IBS group overall, but unrelated to IBS subtype. Altered E-cadherin expression may provide novel insights into mechanisms underlying intestinal barrier dysfunction in IBS.
Collapse
Affiliation(s)
- E Wilcz-Villega
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Adelaide & Meath Hospital, Dublin, Ireland
| | | | | |
Collapse
|
90
|
Abstract
BACKGROUND/AIMS Endoscopic radiofrequency ablation of dysplastic Barrett's esophagus (BE) combined with proton pump inhibitor therapy is commonly utilized for preventing progression of dysplastic BE to esophageal adenocarcinoma. Fundamental to the success of this and all ablative approaches is the healing of the ablated areas of BE with a stratified squamous epithelium referred to as 'neosquamous epithelium' (NSE). Although NSE appears 'normal' endoscopically, the reemergence of BE over time in the previously ablated segments raises the question of the health and integrity of NSE. METHODS The health of NSE was recently investigated in endoscopic biopsies in vitro in a group of patients after ablation while on proton pump inhibitors. Biopsies of NSE were compared to upper squamous epithelium (USE) from the same patients morphologically (light microscopy) and with respect to barrier function by measuring electrical resistance and fluorescein flux in mini-Ussing chambers. RESULTS Compared to USE, NSE exhibited dilated intercellular spaces and inflammation and defective barrier function by low electrical resistance and high fluorescein flux. Moreover, NSE exhibited downregulation of claudin-4, a highly expressed protein in squamous tight junctions. CONCLUSION NSE has defective barrier function in part due to downregulation of claudin-4. Since downregulation of claudin-4 increases paracellular permeability to cations, e.g. hydrogen ions, NSE is more vulnerable to attack and damage by acidic and weakly acidic refluxates--a phenomenon that may contribute in part to the reemergence of BE.
Collapse
Affiliation(s)
- Roy C Orlando
- University of North Carolina at Chapel Hill, Chapel Hill, N.C., USA
| |
Collapse
|
91
|
Fischer A, Gluth M, Weege F, Pape UF, Wiedenmann B, Baumgart DC, Theuring F. Glucocorticoids regulate barrier function and claudin expression in intestinal epithelial cells via MKP-1. Am J Physiol Gastrointest Liver Physiol 2014; 306:G218-28. [PMID: 24309183 DOI: 10.1152/ajpgi.00095.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Barrier dysfunction is pivotal to the pathogenesis of inflammatory bowel diseases (IBD) and collagenous colitis. Glucocorticoids restore barrier function in Crohn's disease, but whether this reflects attenuated inflammation or an epithelial-specific action has not yet been addressed. Using filter-grown Caco-2 monolayers as an in vitro model of the intestinal epithelial barrier, we observed that glucocorticoids induced a time- and dose-dependent increase in transepithelial electrical resistance (TEER) in a glucocorticoid receptor-dependent manner without altering flux of larger solutes or changing principal tight junction architecture. This was accompanied by reduced paracellular cation flux, reduced expression of the pore-forming tight junction component claudin-2, and upregulation of the sealing tight junction protein claudin-4. In contrast, expression of occludin, claudin-1, -7, or -8 was not altered. Dexamethasone increased expression and activity of MAPK phosphatase-1 and inhibition of this phosphatase prevented the glucocorticoid-induced changes in TEER and claudin expression, whereas inhibiting p38 or MEK1/2 was not sufficient to replicate the glucocorticoid effects. Upon exposure to IFN-γ, TNF-α, or IL-1β, TEERs declined in dexamethasone-treated cells but remained consistently higher than in cells not receiving glucocorticoids. Treatment with IFN/TNF resulted in an upregulation of claudin-2 that was significantly attenuated by dexamethasone, whereas increased claudin-2 expression upon IL-1β stimulation was not affected by glucocorticoids. Taken together, barrier augmentation might represent a previously unrecognized mechanism of action, potentially contributing to the therapeutic efficacy of glucocorticoids in IBD and collagenous colitis.
Collapse
Affiliation(s)
- Andreas Fischer
- Department of Medicine, Division of Gastroenterology and Hepatology Campus Virchow Klinikum and
| | | | | | | | | | | | | |
Collapse
|
92
|
Vanheel H, Vicario M, Vanuytsel T, Van Oudenhove L, Martinez C, Keita ÅV, Pardon N, Santos J, Söderholm JD, Tack J, Farré R. Impaired duodenal mucosal integrity and low-grade inflammation in functional dyspepsia. Gut 2014; 63:262-71. [PMID: 23474421 DOI: 10.1136/gutjnl-2012-303857] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Functional dyspepsia (FD) is an extremely common functional gastrointestinal disorder, the pathophysiology of which is poorly understood. We hypothesised that impaired intestinal barrier function is involved in the onset and persistence of this disorder by inducing low-grade inflammation. Therefore, our aim was to evaluate duodenal mucosal integrity and low-grade inflammation in patients with FD. DESIGN Duodenal biopsy specimens were obtained from 15 patients with FD fulfilling the Rome III criteria and 15 age- and gender-matched healthy volunteers. Transepithelial electrical resistance (TEER) and paracellular permeability were measured in Ussing chambers. Expression of cell-to-cell adhesion proteins was evaluated by real-time PCR, western blot and/or immunofluorescence. Numbers of mast cells, eosinophils and intraepithelial lymphocytes were assessed by immunohistochemistry. RESULTS Patients with FD displayed lower TEER and increased paracellular passage compared with healthy controls, which is indicative of impaired mucosal integrity. In addition, abnormal expression of cell-to-cell adhesion proteins at the level of tight junctions, adherens junctions and desmosomes was shown. Furthermore, patients were characterised by the presence of low-grade inflammation, as demonstrated by increased infiltration of mucosal mast cells and eosinophils. A significant association between the expression level of several cell-to-cell adhesion proteins, the extent of increased permeability and the severity of low-grade inflammation was found. CONCLUSIONS These findings challenge the classical paradigm that patients with FD show no structural changes in the gastrointestinal tract. We suggest that impaired intestinal barrier function is a pathophysiological mechanism in FD. Thus, restoration of intestinal barrier integrity may be a potential therapeutic target for treating patients with FD.
Collapse
Affiliation(s)
- Hanne Vanheel
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders, KU Leuven, , Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Muise A, Rotin D. Apical junction complex proteins and ulcerative colitis: a focus on thePTPRSgene. Expert Rev Mol Diagn 2014; 8:465-77. [DOI: 10.1586/14737159.8.4.465] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
94
|
Anderson RC, Young W, Clerens S, Cookson AL, McCann MJ, Armstrong KM, Roy NC. Human oral isolate Lactobacillus fermentum AGR1487 reduces intestinal barrier integrity by increasing the turnover of microtubules in Caco-2 cells. PLoS One 2013; 8:e78774. [PMID: 24244356 PMCID: PMC3828418 DOI: 10.1371/journal.pone.0078774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/16/2013] [Indexed: 02/06/2023] Open
Abstract
Lactobacillus fermentum is found in fermented foods and thought to be harmless. In vivo and clinical studies indicate that some L. fermentum strains have beneficial properties, particularly for gastrointestinal health. However, L. fermentum AGR1487 decreases trans-epithelial electrical resistance (TEER), a measure of intestinal barrier integrity. The hypothesis was that L. fermentum AGR1487 decreases the expression of intestinal cell tight junction genes and proteins, thereby reducing barrier integrity. Transcriptomic and proteomic analyses of Caco-2 cells (model of human intestinal epithelial cells) treated with L. fermentum AGR1487 were used to obtain a global view of the effect of the bacterium on intestinal epithelial cells. Specific functional characteristics by which L. fermentum AGR1487 reduces intestinal barrier integrity were examined using confocal microscopy, cell cycle progression and adherence bioassays. The effects of TEER-enhancing L. fermentum AGR1485 were investigated for comparison. L. fermentum AGR1487 did not alter the expression of Caco-2 cell tight junction genes (compared to L. fermentum AGR1485) and tight junction proteins were not able to be detected. However, L. fermentum AGR1487 increased the expression levels of seven tubulin genes and the abundance of three microtubule-associated proteins, which have been linked to tight junction disassembly. Additionally, Caco-2 cells treated with L. fermentum AGR1487 did not have defined and uniform borders of zona occludens 2 around each cell, unlike control or AGR1485 treated cells. L. fermentum AGR1487 cells were required for the negative effect on barrier integrity (bacterial supernatant did not cause a decrease in TEER), suggesting that a physical interaction may be necessary. Increased adherence of L. fermentum AGR1487 to Caco-2 cells (compared to L. fermentum AGR1485) was likely to facilitate this cell-to-cell interaction. These findings illustrate that bacterial strains of the same species can cause contrasting host responses and suggest that food-safe status should be given to individual strains not species.
Collapse
Affiliation(s)
- Rachel C. Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- * E-mail:
| | - Wayne Young
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Stefan Clerens
- Proteins & Biomaterials Team, Food & Bio-based Products Group, AgResearch Lincoln, Lincoln, New Zealand
| | - Adrian L. Cookson
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Rumen Microbiology Team, Animal Health & Nutrition Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Mark J. McCann
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Gravida: National Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| | - Kelly M. Armstrong
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Nicole C. Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Rumen Microbiology Team, Animal Health & Nutrition Group, AgResearch Grasslands, Palmerston North, New Zealand
- Gravida: National Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
95
|
Barnich N, Denizot J, Darfeuille-Michaud A. E. coli-mediated gut inflammation in genetically predisposed Crohn's disease patients. ACTA ACUST UNITED AC 2013; 61:e65-9. [DOI: 10.1016/j.patbio.2010.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 01/18/2010] [Indexed: 12/12/2022]
|
96
|
Rodríguez-Lagunas M, Storniolo C, Ferrer R, Moreno J. 5-Hydroxyeicosatetraenoic acid and leukotriene D4 increase intestinal epithelial paracellular permeability. Int J Biochem Cell Biol 2013; 45:1318-26. [DOI: 10.1016/j.biocel.2013.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 03/31/2013] [Accepted: 04/03/2013] [Indexed: 01/18/2023]
|
97
|
Fischer A, Gluth M, Pape UF, Wiedenmann B, Theuring F, Baumgart DC. Adalimumab prevents barrier dysfunction and antagonizes distinct effects of TNF-α on tight junction proteins and signaling pathways in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2013; 304:G970-9. [PMID: 23538493 DOI: 10.1152/ajpgi.00183.2012] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intestinal barrier dysfunction is pivotal in the etiology of inflammatory bowel diseases. Combined clinical and endoscopic remission ("mucosal healing") in patients who received anti-TNF-α therapies suggests restitution of the intestinal barrier, but the mechanisms involved are largely unknown. We therefore investigated the impact of the anti-TNF-α antibody adalimumab on barrier function in two in vitro models. Combined stimulation of Caco-2 and T-84 cells with interferon-γ and TNF-α resulted in a significant decrease of transepithelial electrical resistance (TEER) within 6 h that was prevented by adalimumab in concentrations down to 100 ng/ml. Adalimumab furthermore antagonized the appearance of irregular membrane undulations and prevented internalization of tight junction proteins upon cytokine exposure. In addition, TNF-α induced a downregulation of claudin-1, claudin-2, claudin-4, and occludin as well as activation of phosphatidylinositol 3-kinase signaling in T-84 but not Caco-2 cells, which was reversed by adalimumab. At the signaling level, adalimumab prevented increased phosphorylation of myosin light chain as well as activation of p38 MAPK and NF-κB accompanying the decline in TEER in both model systems. Pharmacological inhibition of NF-κB signaling partially prevented the TNF-α-induced TEER loss, whereas inhibition of p38 worsened barrier dysfunction in Caco-2 but not T-84 cells. Taken together, these data demonstrate that adalimumab prevents barrier dysfunction induced by TNF-α both functionally and structurally as well as at the level of signal transduction. Barrier protection might therefore constitute a novel mechanism how anti-TNF-α therapy contributes to epithelial restitution and tissue repair in inflammatory bowel diseases.
Collapse
Affiliation(s)
- Andreas Fischer
- Department of Medicine, Division of Gastroenterology and Hepatology, Humboldt-University of Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
98
|
Early life stress triggers persistent colonic barrier dysfunction and exacerbates colitis in adult IL-10-/- mice. Inflamm Bowel Dis 2013; 19:712-9. [PMID: 23446335 PMCID: PMC4114389 DOI: 10.1097/mib.0b013e3182802a4e] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND It has become increasingly evident that disease flares in the human inflammatory bowel diseases are influenced by life stress. It is known that life stress can trigger disturbances in intestinal barrier function and activate proinflammatory signaling pathways, which are important contributors to intestinal inflammation and clinical disease; however, the exact mechanisms of stress-induced inflammatory bowel disease exacerbations remain to be elucidated. Here, we presented a model of early life stress-induced exacerbation of colitis in interleukin (IL)-10 mice. METHODS C57Bl/6 wild-type and IL-10 mice were exposed to neonatal maternal separation (NMS) stress on postnatal days 1 to 18 and reared under normal conditions until 10 to 12 weeks of age. At this time, histopathology, colitis scores, intestinal barrier function, proinflammatory cytokine expression, and mast cell activity were evaluated. RESULTS NMS increased the severity of colitis IL-10 mice indicated by greater colitis scores and colonic proinflammatory cytokine concentrations. NMS and IL-10 increased colonic permeability; however, NMS alone did not induce colitis. Increased mast cell activation and colonic tryptase release were observed in IL-10 mice exposed to NMS, indicating mast cell activation. CONCLUSIONS This study demonstrates that colitis in IL-10 mice can be exacerbated by NMS stress. The precise mechanisms of enhanced colitis severity in NMS IL10 mice are unclear but persistent defects in intestinal barrier function likely play a contributing role. NMS serves as a novel model to investigate the mechanisms by which early life stress influences the development and course of inflammatory bowel disease in adulthood.
Collapse
|
99
|
Bermudez-Brito M, Plaza-Díaz J, Muñoz-Quezada S, Gómez-Llorente C, Gil A. Probiotic mechanisms of action. ANNALS OF NUTRITION AND METABOLISM 2012; 61:160-74. [PMID: 23037511 DOI: 10.1159/000342079] [Citation(s) in RCA: 662] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 12/28/2022]
Abstract
Probiotics are live microorganisms that provide health benefits to the host when ingested in adequate amounts. The strains most frequently used as probiotics include lactic acid bacteria and bifidobacteria. Probiotics have demonstrated significant potential as therapeutic options for a variety of diseases, but the mechanisms responsible for these effects have not been fully elucidated yet. Several important mechanisms underlying the antagonistic effects of probiotics on various microorganisms include the following: modification of the gut microbiota, competitive adherence to the mucosa and epithelium, strengthening of the gut epithelial barrier and modulation of the immune system to convey an advantage to the host. Accumulating evidence demonstrates that probiotics communicate with the host by pattern recognition receptors, such as toll-like receptors and nucleotide-binding oligomerization domain-containing protein-like receptors, which modulate key signaling pathways, such as nuclear factor-ĸB and mitogen-activated protein kinase, to enhance or suppress activation and influence downstream pathways. This recognition is crucial for eliciting measured antimicrobial responses with minimal inflammatory tissue damage. A clear understanding of these mechanisms will allow for appropriate probiotic strain selection for specific applications and may uncover novel probiotic functions. The goal of this systematic review was to explore probiotic modes of action focusing on how gut microbes influence the host.
Collapse
Affiliation(s)
- Miriam Bermudez-Brito
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology José Mataix, Biomedical Research Center, University of Granada, Armilla, Spain
| | | | | | | | | |
Collapse
|
100
|
Alhamoruni A, Wright KL, Larvin M, O'Sullivan SE. Cannabinoids mediate opposing effects on inflammation-induced intestinal permeability. Br J Pharmacol 2012; 165:2598-610. [PMID: 21745190 DOI: 10.1111/j.1476-5381.2011.01589.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of cannabinoid receptors decreases emesis, inflammation, gastric acid secretion and intestinal motility. The ability to modulate intestinal permeability in inflammation may be important in therapy aimed at maintaining epithelial barrier integrity. The aim of the present study was to determine whether cannabinoids modulate the increased permeability associated with inflammation in vitro. EXPERIMENTAL APPROACH Confluent Caco-2 cell monolayers were treated for 24 h with IFNγ and TNFα (10 ng·mL(-1) ). Monolayer permeability was measured using transepithelial electrical resistance and flux measurements. Cannabinoids were applied either apically or basolaterally after inflammation was established. Potential mechanisms of action were investigated using antagonists for CB(1) , CB(2) , TRPV1, PPARγ and PPARα. A role for the endocannabinoid system was established using inhibitors of the synthesis and degradation of endocannabinoids. KEY RESULTS Δ(9) -Tetrahydrocannabinol (THC) and cannabidiol accelerated the recovery from cytokine-induced increased permeability; an effect sensitive to CB(1) receptor antagonism. Anandamide and 2-arachidonylglycerol further increased permeability in the presence of cytokines; this effect was also sensitive to CB(1) antagonism. No role for the CB(2) receptor was identified in these studies. Co-application of THC, cannabidiol or a CB(1) antagonist with the cytokines ameliorated their effect on permeability. Inhibiting the breakdown of endocannabinoids worsened, whereas inhibiting the synthesis of endocannabinoids attenuated, the increased permeability associated with inflammation. CONCLUSIONS AND IMPLICATIONS These findings suggest that locally produced endocannabinoids, acting via CB(1) receptors play a role in mediating changes in permeability with inflammation, and that phytocannabinoids have therapeutic potential for reversing the disordered intestinal permeability associated with inflammation. LINKED ARTICLES This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.
Collapse
Affiliation(s)
- A Alhamoruni
- School of Graduate Entry Medicine & Health, Derby City General Hospital, University of Nottingham, Derby, UK
| | | | | | | |
Collapse
|