201
|
Yang Y, Huang C, Lin X, Wu Y, Ouyang W, Tang L, Ye S, Wang Y, Li W, Zhang X, Liu Z. 0.005% Preservative-Free Latanoprost Induces Dry Eye-Like Ocular Surface Damage via Promotion of Inflammation in Mice. Invest Ophthalmol Vis Sci 2019; 59:3375-3384. [PMID: 30025085 DOI: 10.1167/iovs.18-24013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the side effects of preservative-free 0.005% latanoprost on the murine ocular surface. Methods We applied 0.005% latanoprost or vehicle in mice in two patterns for 14 to 28 days. Tear production was measured by phenol red cotton test, and corneal epithelial barrier function was assessed by Oregon-green-dextran (OGD) staining. Periodic acid-Schiff (PAS) staining was used to quantify conjunctival goblet cells (GCs). The expression of matrix metalloproteinase (MMP)-3 and -9, occludin-1 and zonula occludens (ZO)-1 in corneal epithelium was assessed by immunofluorescent staining and/or quantitative real-time PCR (qRT-PCR). Inflammation in conjunctiva was assessed by activation of P38 and NF-κB, infiltration of CD4+ T cells, and production inflammatory cytokines including TNF-α, IL-1β, IFN-γ, IL-17A, and IL-13. Apoptosis in ocular surface was assessed by TUNEL and immunofluorescent staining for activated caspase-3 and -8. Cell viability assay was performed in human corneal epithelial cells. Results Topical latanoprost treatment decreased tear production, induced conjunctival GC loss, disrupted the corneal epithelial barrier, and promoted cell apoptosis in the ocular surface. Topical latanoprost treatment increased the expression of MMP-3 and -9, and decreased the expression of ZO-1 and occludin-1 in the corneal epithelium. Topical application of latanoprost promoted activation of P38-NF-κB signaling and production of TNF-α and IL-1β in conjunctiva. Topical application of latanoprost increased CD4+ T cells infiltration, with increased production of IFN-γ and IL-17A and decreased production of IL-13 in conjunctiva. Conclusion 0.005% latanoprost induced dry eye-like ocular surface damage via promotion of inflammation in mice.
Collapse
Affiliation(s)
- Yiran Yang
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Caihong Huang
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Xiang Lin
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Yang Wu
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Weijie Ouyang
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Liying Tang
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Sihao Ye
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Yuhong Wang
- Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Wei Li
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China.,Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaobo Zhang
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China.,Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Eye Institute of Xiamen University & Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China.,Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
202
|
Shakoori M, Hoseinifar SH, Paknejad H, Jafari V, Safari R, Van Doan H, Torfi Mozanzadeh M. Enrichment of rainbow trout (Oncorhynchus mykiss) fingerlings diet with microbial lysozyme: Effects on growth performance, serum and skin mucus immune parameters. FISH & SHELLFISH IMMUNOLOGY 2019; 86:480-485. [PMID: 30513385 DOI: 10.1016/j.fsi.2018.11.077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
A two-month study was conducted to determine the influence of different levels of microbial lysozyme (LZ) contents (0, 0.5, 1.0, and 1.5 g kg-1 of diet) on growth performance, serum and skin mucus immune parameters as well as intestinal immune-related genes expression in rainbow trout Oncorhynchus mykiss fingerlings (5.5 ± 0.1 g). Growth performance and feed utilization were not affected significantly by dietary LZ. Fish fed LZ-supplemented diets had higher serum total immunoglobulins concentration than the control group. In addition, fish fed 1.5 g LZ kg-1 diet had the highest skin mucosal total protein and immunoglobulin contents compared to other experimental groups. Furthermore, skin mucosal lysozyme and alkaline phosphatase activities as well as intestinal tumor necrosis factor-α and interlukine-1β relative genes expression were higher in fish fed 1.0 and 1.5 g LZ kg-1 diets than the other groups. Overall, the present results clearly showed that LZ powder can be considered as a potential immunostimulant in O. mykiss fingerlings, but in the long term period it may result in negative effects on intestinal health as a consequence of inducing pro-inflammatory cytokines gene expression in the intestine.
Collapse
Affiliation(s)
- Meysam Shakoori
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Seyed Hossein Hoseinifar
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Hamed Paknejad
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Valiollah Jafari
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Roghieh Safari
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Hien Van Doan
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Mansour Torfi Mozanzadeh
- South Iran Aquaculture Research Centre, Iranian Fisheries Science Institute (IFSRI), Agricultural Research Education and Extension Organization (AREEO), Ahwaz, Iran
| |
Collapse
|
203
|
Du L, Long Y, Kim JJ, Chen B, Zhu Y, Dai N. Protease Activated Receptor-2 Induces Immune Activation and Visceral Hypersensitivity in Post-infectious Irritable Bowel Syndrome Mice. Dig Dis Sci 2019; 64:729-739. [PMID: 30446929 DOI: 10.1007/s10620-018-5367-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 11/07/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND The role of protease activated receptor-2 (PAR-2) in the pathogenesis of abdominal pain in irritable bowel syndrome (IBS) is not well defined. AIMS To investigate the role of PAR-2-mediated visceral hypersensitivity in a post-infectious IBS (PI-IBS) mouse model. METHODS T. spiralis-infected PI-IBS mouse model was used. Fecal serine protease activity and intestinal mast cells were evaluated. Intestinal permeability was assessed by urine lactulose/mannitol ratio, and colonic expressions of PAR-2 and tight junction (TJ) proteins were examined by Western blot. Intestinal immune profile was assessed by measuring Th (T helper) 1/Th2 cytokine expression. Visceral sensitivity was evaluated by abdominal withdrawal reflex in response to colorectal distention. RESULTS Colonic PAR-2 expression as well as fecal serine protease activity and intestinal mast cell counts were elevated in PI-IBS compared to the control mice. Decreased colonic TJ proteins expression, increased lactulose/mannitol ratio, elevated colonic Th1/Th2 cytokine ratio, and visceral hypersensitivity were observed in PI-IBS compared to the control mice. Administration of PAR-2 agonist in control mice demonstrated similar changes observed in PI-IBS mice, while PAR-2 antagonist normalized the increased intestinal permeability and reduced visceral hypersensitivity observed in PI-IBS mice. CONCLUSIONS PAR-2 activation increases intestinal permeability leading to immune activation and visceral hypersensitivity in PI-IBS mouse model.
Collapse
Affiliation(s)
- Lijun Du
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yanqin Long
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
| | - John J Kim
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
- Division of Gastroenterology & Hepatology, Loma Linda University Health, Loma Linda, CA, 92354, USA
| | - Binrui Chen
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yubin Zhu
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Ning Dai
- Department of Gastroenterology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| |
Collapse
|
204
|
Khare V, Krnjic A, Frick A, Gmainer C, Asboth M, Jimenez K, Lang M, Baumgartner M, Evstatiev R, Gasche C. Mesalamine and azathioprine modulate junctional complexes and restore epithelial barrier function in intestinal inflammation. Sci Rep 2019; 9:2842. [PMID: 30809073 PMCID: PMC6391397 DOI: 10.1038/s41598-019-39401-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
Disruption of mucosal structure and barrier function contribute to the pathogenesis of inflammatory bowel disease (IBD). Efficacy of therapy in IBD is based on endoscopic mucosal healing, which occurs by a dynamic interplay of epithelial cell regeneration, migration and differentiation. Both mesalamine (5-ASA) and azathioprine (AZTP) promote this process through mechanisms not clearly understood. We examined molecular pathways implicated in epithelial barrier function that were altered by 5-ASA and AZTP. Paracellular permeability induced by inflammatory mediators was mitigated by both compounds through restoration of cellular anchoring complexes. 5-ASA and AZTP induced rearrangement and membranous localization of junctional proteins and modulated genes involved in tight junctions. Intestinal organoids from wildtype-mice treated with TNF-α and IL-10- deficient-mice displayed impaired epithelial barrier with loss of membranous E-cadherin and reduced Desmoglein-2 expression. These effects were counteracted by 5-ASA and AZTP. Unlike AZTP that exhibited antiproliferative effects, 5-ASA promoted wound healing in colon epithelial cells. Both affected cellular senescence, cell cycle distribution and restricted cells in G1 or S phase without inducing apoptosis. This study provides mechanistic evidence that molecular actions of 5-ASA and AZTP on intestinal epithelia are fundamental in the resolution of barrier dysfunction.
Collapse
Affiliation(s)
- Vineeta Khare
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Anita Krnjic
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Adrian Frick
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Christina Gmainer
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Mario Asboth
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Kristine Jimenez
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Michaela Lang
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Maximilian Baumgartner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Rayko Evstatiev
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
205
|
Zhai Z, Torres-Fuentes C, Heeney DD, Marco ML. Synergy between Probiotic Lactobacillus casei and Milk to Maintain Barrier Integrity of Intestinal Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1955-1962. [PMID: 30629420 DOI: 10.1021/acs.jafc.8b06657] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We hypothesized that Lactobacillus casei BL23 and milk work synergistically to prevent damage to epithelial barrier integrity induced by pro-inflammatory cytokines. To test this, barrier disruption was induced in polarized Caco-2 monolayers by sequential, basolateral treatment with IFN-γ and TNF-α. Apical application of either 25% v/v reconstituted skim milk (RSM) or ultra high temperature (UHT) milk (2% fat) prior to cytokine exposure reduced losses to transepithelial electrical resistance (TER). Permeability to fluorescein isothiocyanate-dextran (FD-4; 4 kDa) was also significantly reduced in the presence of 25% v/v UHT milk ( P < 0.05) but not RSM. Protection against increases in paracellular permeability was even greater when cell-free preparations of L. casei BL23 fermented UHT milk or fermented RSM were applied. The permeability coefficients of cells incubated with BL23 fermented UHT milk were equivalent to the untreated controls ( P = 0.12) and those cells also produced 247.6 ± 35.5 pg/mL IL-8, quantities significantly lower than found for cytokine-treated controls (353.9 ± 40.0 pg/mL). The benefits of the fermented milk were also confirmed by the reduced expression of TNF receptor 2 (TNFR2), myosin light-chain kinase (MLCK), and claudin-encoding genes relative to the controls. By comparison, apical application of viable L. casei onto the Caco-2 cells did not result in protection from the barrier-disruptive actions of IFN-γ and TNF-α. These results indicate that milk can maintain intestinal barrier integrity during pro-inflammatory cytokine exposure and that this is enhanced by modifications to milk matrix caused by prior incubation with L. casei BL23.
Collapse
Affiliation(s)
- Zhengyuan Zhai
- Department of Food Science & Technology , University of California , Davis , California 95616 , United States of America
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality , China Agricultural University , Beijing , 100083 China
| | - Cristina Torres-Fuentes
- Department of Food Science & Technology , University of California , Davis , California 95616 , United States of America
| | - Dustin D Heeney
- Department of Food Science & Technology , University of California , Davis , California 95616 , United States of America
| | - Maria L Marco
- Department of Food Science & Technology , University of California , Davis , California 95616 , United States of America
| |
Collapse
|
206
|
Nalle SC, Zuo L, Ong MLDM, Singh G, Worthylake AM, Choi W, Manresa MC, Southworth AP, Edelblum KL, Baker GJ, Joseph NE, Savage PA, Turner JR. Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability. J Clin Invest 2019; 129:902-914. [PMID: 30667372 PMCID: PMC6355225 DOI: 10.1172/jci98554] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a complication of hematopoietic stem cell transplantation (HSCT) that affects multiple organs. GVHD-associated intestinal damage can be separated into two distinct phases, initiation and propagation, which correspond to conditioning-induced damage and effector T cell activation and infiltration, respectively. Substantial evidence indicates that intestinal damage induced by pretransplant conditioning is a key driver of GVHD initiation. Here, we aimed to determine the impact of dysregulated intestinal permeability on the subsequent GVHD propagation phase. The initiation phase of GVHD was unchanged in mice lacking long MLCK (MLCK210), an established regulator of epithelial tight junction permeability. However, MLCK210-deficient mice were protected from sustained barrier loss and exhibited limited GVHD propagation, as indicated by reduced histopathology, fewer CD8+ effector T cells in the gut, and improved overall survival. Consistent with these findings, intestinal epithelial MLCK210 expression and enzymatic activity were similarly increased in human and mouse GVHD biopsies. Intestinal epithelial barrier loss mediated by MLCK210 is therefore a key driver of the GVHD propagation. These data suggest that inhibition of MLCK210-dependent barrier regulation may be an effective approach to limiting GVHD progression.
Collapse
Affiliation(s)
- Sam C. Nalle
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Li Zuo
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Anhui Medical University, Hefei, Anhui, China
| | - Ma. Lora Drizella M. Ong
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alicia M. Worthylake
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wangsun Choi
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mario Cabrero Manresa
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna P. Southworth
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Karen L. Edelblum
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Cancer Center, Newark, New Jersey, USA
| | - Gregory J. Baker
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts, USA
| | - Nora E. Joseph
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Peter A. Savage
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
207
|
Wang Y, Li Y, Zou J, Polster SP, Lightle R, Moore T, Dimaano M, He TC, Weber CR, Awad IA, Shen L. The cerebral cavernous malformation disease causing gene KRIT1 participates in intestinal epithelial barrier maintenance and regulation. FASEB J 2019; 33:2132-2143. [PMID: 30252535 PMCID: PMC6338648 DOI: 10.1096/fj.201800343r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 08/27/2018] [Indexed: 01/15/2023]
Abstract
Epithelial barrier maintenance and regulation requires an intact perijunctional actomyosin ring underneath the cell-cell junctions. By searching for known factors affecting the actin cytoskeleton, we identified Krev interaction trapped protein 1 (KRIT1) as a major regulator for epithelial barrier function through multiple mechanisms. KRIT1 is expressed in both small intestinal and colonic epithelium, and KRIT1 knockdown in differentiated Caco-2 intestinal epithelium decreases epithelial barrier function and increases cation selectivity. KRIT1 knockdown abolished Rho-associated protein kinase-induced and myosin II motor inhibitor-induced barrier loss by limiting both small and large molecule permeability but did not affect myosin light chain kinase-induced increases in epithelial barrier function. These data suggest that KRIT1 participates in Rho-associated protein kinase- and myosin II motor-dependent (but not myosin light chain kinase-dependent) epithelial barrier regulation. KRIT1 knockdown exacerbated low-dose TNF-induced barrier loss, along with increased cleaved caspase-3 production. Both events are blocked by pan-caspase inhibition, indicating that KRIT1 regulates TNF-induced barrier loss through limiting epithelial apoptosis. These data indicate that KRIT1 controls epithelial barrier maintenance and regulation through multiple pathways, suggesting that KRIT1 mutation in cerebral cavernous malformation disease may alter epithelial function and affect human health.-Wang, Y., Li, Y., Zou, J., Polster, S. P., Lightle, R., Moore, T., Dimaano, M., He, T.-C., Weber, C. R., Awad, I. A., Shen, L. The cerebral cavernous malformation disease causing gene KRIT1 participates in intestinal epithelial barrier maintenance and regulation.
Collapse
Affiliation(s)
- Yitang Wang
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Ye Li
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Jinjing Zou
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Sean P. Polster
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Rhonda Lightle
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Thomas Moore
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Matthew Dimaano
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA; and
| | - Tong-Chuan He
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Issam A. Awad
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Le Shen
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
208
|
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders affecting the gastrointestinal tract. The incidence of IBD is increasing, with more cases occurring in developed countries. Multiple factors such as genetics, environmental changes, gut microbiota, and immune abnormalities have been associated with development of IBD. In recent years, it has become increasingly apparent that epigenetic modifications of chromatin and the manner in which chromatin is organized in the nucleus are additionally important elements that can influence responses induced by the factors described above, and may therefore contribute to the onset and pathogenesis of IBD. Epigenetics and chromatin organization regulate diverse functions that include maintenance of homeostasis in the intestinal epithelium, the development and differentiation of immune cells, and modulation of responses generated by the immune system to defend against potential pathogens. Furthermore, changes in epigenetic chromatin marks and in chromatin organization have now been linked to differential gene expression in IBD patient cells. Although direct evidence for a role of histone modifications in IBD is currently very limited, in this review, we summarize the links between various epigenetic modifications, the proteins that catalyze or recognize these modifications, and the development or progression of IBD in human and experimental IBD. We also discuss how epigenetics influence the organization of DNA contacts to regulate gene expression and the implications this may have for diagnosing and treating IBD.
Collapse
Affiliation(s)
- Greeshma Ray
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michelle S Longworth
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA,Address correspondence to: Michelle S. Longworth, 9500 Euclid Ave NC22, Cleveland, OH 44195 ()
| |
Collapse
|
209
|
Chen Y, Zhang M, Ren F. A Role of Exopolysaccharide Produced by Streptococcus thermophilus in the Intestinal Inflammation and Mucosal Barrier in Caco-2 Monolayer and Dextran Sulphate Sodium-Induced Experimental Murine Colitis. Molecules 2019; 24:molecules24030513. [PMID: 30708992 PMCID: PMC6384629 DOI: 10.3390/molecules24030513] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Exopolysaccharide (EPS) produced by probiotics may play an important role in gastrointestinal disease prevention, including ulcerative colitis. However, there is no literature reporting on the intervention effects of purified EPS. The aim of this study was to investigate the alleviating effect of the purified EPS produced by Streptococcus thermophilus MN-BM-A01 on murine model of colitis induced by dextran sulphate sodium (DSS). A water-soluble heteropolysaccharide (EPS-1) isolated from MN-BM-A01 was composed of rhamnose, glucose, galactose, and mannose in a molar ratio of 12.9:26.0:60.9:0.25, with molecular weight of 4.23 × 105 Da. After EPS-1 administration, the disease severity of mouse colitis was significantly alleviated, mainly manifesting as the decrease of disease activity index and mitigated colonic epithelial cell injury. Meanwhile, pro-inflammatory cytokines levels (tumor necrosis factor-α, interleukin-6, and interferon-γ) were significantly suppressed, the reduced expressions of tight junction protein (claudin-1, occludin, and E-canherin) were counteracted. In addition, the results in vitro showed that EPS-1 protected intestinal barrier integrity from the disruption by lipopolysaccharide in Caco-2 monolayer, increased expression of tight junction and alleviated pro-inflammatory response. Collectively, our study confirmed the protective effects of purified EPS produced by Streptococcus thermophilus on acute colitis via alleviating intestinal inflammation and improving mucosal barrier function.
Collapse
Affiliation(s)
- Yun Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Ming Zhang
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing 100048, China.
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
210
|
Delgado ME, Brunner T. The many faces of tumor necrosis factor signaling in the intestinal epithelium. Genes Immun 2019; 20:609-626. [DOI: 10.1038/s41435-019-0057-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023]
|
211
|
Maes E, Sadovskaya I, Lévêque M, Elass-Rochard E, Payré B, Grard T, Théodorou V, Guérardel Y, Mercier-Bonin M. Structure and biological activities of a hexosamine-rich cell wall polysaccharide isolated from the probiotic Lactobacillus farciminis. Glycoconj J 2019; 36:39-55. [PMID: 30637506 DOI: 10.1007/s10719-018-09854-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022]
Abstract
Lactobacillus farciminis CIP 103136 is a bacterial strain with recognized probiotic properties. However, the mechanisms underlying such properties have only been partially elucidated. In this study, we isolated and purified a cell-wall associated polysaccharide (CWPS), and evaluated its biological role in vitro. The structure of CWPS and responses from stimulation of (i) human macrophage-like THP-1 cells, (ii) human embryonal kidney (HEK293) cells stably transfected with Toll-like receptors (TLR2 or TLR4) and (iii) human colonocyte-like T84 intestinal epithelial cells, upon exposure to CWPS were studied. The structure of the purified CWPS from L. farciminis CIP 103136 was analyzed by nuclear magnetic resonance (NMR), MALDI-TOF-TOF MS, and methylation analyses in its native form and following Smith degradation. It was shown to be a novel branched polysaccharide, composed of linear backbone of trisaccharide repeating units of: [→6αGlcpNAc1 → 4βManpNAc1 → 4βGlcpNAc1→] highly substituted with single residues of αGlcp, αGalp and αGlcpNAc. Subsequently, the lack of pro- or anti-inflammatory properties of CWPS was established on macrophage-like THP-1 cells. In addition, CWPS failed to modulate cell signaling pathways dependent of TLR2 and TLR4 in transfected HEK-cells. Finally, in T84 cells, CWPS neither influenced intestinal barrier integrity under basal conditions nor prevented TNF-α/IFN-γ cytokine-mediated epithelium impairment.
Collapse
Affiliation(s)
- Emmanuel Maes
- CNRS UMR 8576, UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Univ Lille, 59 000, Lille, France
| | - Irina Sadovskaya
- Equipe Biochimie des Produits Aquatiques BPA, Institut Régional Charles Violette EA 7394, USC Anses-ULCO, Université du Littoral-Côte d'Opale, Bassin Napoléon, 62327, Boulogne-sur-mer cedex, France
| | - Mathilde Lévêque
- Toxalim (Research Centre in Food Toxicology), INRA, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Elisabeth Elass-Rochard
- CNRS UMR 8576, UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Univ Lille, 59 000, Lille, France
| | - Bruno Payré
- Faculté de Médecine Rangueil, Centre de Microscopie Electronique Appliquée à la Biologie (CMEAB), Toulouse Cedex, France
| | - Thierry Grard
- Equipe Biochimie des Produits Aquatiques BPA, Institut Régional Charles Violette EA 7394, USC Anses-ULCO, Université du Littoral-Côte d'Opale, Bassin Napoléon, 62327, Boulogne-sur-mer cedex, France
| | - Vassilia Théodorou
- Toxalim (Research Centre in Food Toxicology), INRA, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Yann Guérardel
- CNRS UMR 8576, UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Univ Lille, 59 000, Lille, France
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), INRA, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France.
| |
Collapse
|
212
|
Cocetta V, Catanzaro D, Borgonetti V, Ragazzi E, Giron MC, Governa P, Carnevali I, Biagi M, Montopoli M. A Fixed Combination of Probiotics and Herbal Extracts Attenuates Intestinal Barrier Dysfunction from Inflammatory Stress in an In vitro Model Using Caco-2 Cells. Recent Pat Food Nutr Agric 2019; 10:62-69. [PMID: 30088455 DOI: 10.2174/2212798410666180808121328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/02/2018] [Accepted: 08/05/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Inflammatory Bowel Diseases (IBD), are considered a growing global disease, with about ten million people being affected worldwide. Maintenance of intestinal barrier integrity is crucial for preventing IBD onset and exacerbations. Some recent patents regarding oily formulations containing probiotics (WO2010122107A1 and WO2010103374A9) and the use of probiotics for gastrointestinal complaints (US20110110905A1 and US9057112B2) exist, or are pending application. OBJECTIVE In this work, we studied the effect of a fixed combination of registered Lactobacillus reuteri and Lactobacillus acidophilus strains and herbal extracts in an in vitro inflammation experimental model. METHODS Caco-2 cell monolayer was exposed to INF-γ+TNF-α or to LPS; Trans Epithelial Electrical Resistance (TEER) and paracellular permeability were investigated. ZO-1 and occludin Tight Junctions (TJs) were also investigated by mean of immunofluorescence. RESULTS Pre-treatment with the fixed combination of probiotics and herbal extracts prevented the inflammation-induced TEER decrease, paracellular permeability increase and TJs translocation. CONCLUSIONS In summary, the fixed combination of probiotics and herbal extracts investigated in this research was found to be an interesting candidate for targeting the re-establishment of intestinal barrier function in IBD conditions.
Collapse
Affiliation(s)
- Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Daniela Catanzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Vittoria Borgonetti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Maria C Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Paolo Governa
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
- SIFITLab, Italian Society of Phytotherapy, 53100 Siena, Italy
| | | | - Marco Biagi
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
- SIFITLab, Italian Society of Phytotherapy, 53100 Siena, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
- SIFITLab, Italian Society of Phytotherapy, 53100 Siena, Italy
| |
Collapse
|
213
|
Tunisi L, Forte N, Fernández-Rilo AC, Mavaro I, Capasso R, D'Angelo L, Milić N, Cristino L, Di Marzo V, Palomba L. Orexin-A Prevents Lipopolysaccharide-Induced Neuroinflammation at the Level of the Intestinal Barrier. Front Endocrinol (Lausanne) 2019; 10:219. [PMID: 31024456 PMCID: PMC6467935 DOI: 10.3389/fendo.2019.00219] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/18/2019] [Indexed: 12/20/2022] Open
Abstract
In states of intestinal dysbiosis, a perturbation of the normal microbiome composition, the intestinal epithelial barrier (IEB) permeability is increased as a result of the disruption of the epithelial tight junction protein network, in which occludin is mostly affected. The loss of IEB integrity promotes endotoxemia, that is, bacterial lipopolysaccharide (LPS) translocation from the intestinal lumen to the circulatory system. This condition induces an enhancement of pro-inflammatory cytokines, which leads to neuroinflammation through the gut-brain axis. Orexin-A (OX-A), a neuropeptide implicated in many physiological functions and produced mainly in the brain lateral hypothalamic area, is expressed also in several peripheral tissues. Orexin-producing neurons have been found in the myenteric plexus to project to orexin receptor 1 (OX-1R)-expressing enterocytes of the intestinal villi. In the present study we investigated the protective role of OX-A against LPS-induced increase of IEB permeability and microglia activation in both an in vivo and in vitro model of the gut-brain axis. By exploiting biochemical, immunocytochemical, immunohistochemical, and functional approaches, we demonstrate that OX-A preserves the IEB and occludin expression, thus preventing endotoxemia and subsequent neuroinflammation.
Collapse
Affiliation(s)
- Lea Tunisi
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Nicola Forte
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Alba Clara Fernández-Rilo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Isabella Mavaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Raffaele Capasso
- Department of Agricultural Science, University of Naples Federico II, Portici, Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
- Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Nataša Milić
- Department of Pharmacy, University of Novi Sad, Novi Sad, Serbia
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axus in Metabolic Health, Faculty of Medicine and Faculty of Agricultural and Food Sciences, Université Laval, Québec City, QC, Canada
| | - Letizia Palomba
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- *Correspondence: Letizia Palomba
| |
Collapse
|
214
|
Bashir M, Meddings J, Alshaikh A, Jung D, Le K, Amin R, Ratakonda S, Sharma S, Granja I, Satti M, Asplin J, Hassan H. Enhanced gastrointestinal passive paracellular permeability contributes to the obesity-associated hyperoxaluria. Am J Physiol Gastrointest Liver Physiol 2019; 316:G1-G14. [PMID: 30307745 PMCID: PMC6383380 DOI: 10.1152/ajpgi.00266.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Most kidney stones (KS) are composed of calcium oxalate and small increases in urine oxalate enhance the stone risk. Obesity is a risk factor for KS, and urinary oxalate excretion increases with increased body size. We previously established the obese ob/ob ( ob) mice as a model (3.3-fold higher urine oxalate) to define the pathogenesis of obesity-associated hyperoxaluria (OAH). The purpose of this study was to test the hypothesis that the obesity-associated enhanced small intestinal paracellular permeability contributes to OAH by increasing passive paracellular intestinal oxalate absorption. ob Mice have significantly higher jejunal (1.6-fold) and ileal (1.4-fold) paracellular oxalate absorption ex vivo and significantly higher (5-fold) urine [13C]oxalate following oral gavage with [13C]oxalate, indicating increased intestinal oxalate absorption in vivo. The observation of higher oxalate absorption in vivo compared with ex vivo suggests the possibility of increased paracellular permeability along the entire gut. Indeed, ob mice have significantly higher fractions of the administered sucrose (1.7-fold), lactulose (4.4-fold), and sucralose (3.1-fold) excreted in the urine, reflecting increased gastric, small intestinal, and colonic paracellular permeability, respectively. The ob mice have significantly reduced gastrointestinal occludin, zonula occludens-1, and claudins-1 and -3 mRNA and total protein expression. Proinflammatory cytokines and oxidative stress, which are elevated in obesity, significantly enhanced paracellular intestinal oxalate absorption in vitro and ex vivo. We conclude that obese mice have significantly higher intestinal oxalate absorption and enhanced gastrointestinal paracellular permeability in vivo, which would likely contribute to the pathogenesis of OAH, since there is a transepithelial oxalate concentration gradient to drive paracellular intestinal oxalate absorption. NEW & NOTEWORTHY This study shows that the obese ob/ob mice have significantly increased gastrointestinal paracellular oxalate absorption and remarkably enhanced paracellular permeability along the entire gut in vivo, which are likely mediated by the obesity-associated increased systemic and intestinal inflammation and oxidative stress. A transepithelial oxalate concentration gradient driving gastrointestinal paracellular oxalate absorption exists, and therefore, our novel findings likely contribute to the hyperoxaluria observed in the ob/ob mice and hence to the pathogenesis of obesity-associated hyperoxaluria.
Collapse
Affiliation(s)
- Mohamed Bashir
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Jon Meddings
- 2Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Altayeb Alshaikh
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Daniel Jung
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kim Le
- 2Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Ruhul Amin
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | | | - Sapna Sharma
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Ignacio Granja
- 3Litholink Corporation, Laboratory Corporation of America Holdings, Chicago, Illinois
| | - Mustafa Satti
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| | - John Asplin
- 3Litholink Corporation, Laboratory Corporation of America Holdings, Chicago, Illinois
| | - Hatim Hassan
- 1Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
215
|
Activation of the Nuclear Factor-kappa B Signaling Pathway Damages the Epithelial Barrier in the Human Pancreatic Ductal Adenocarcinoma Cell Line HPAF-II. Pancreas 2019; 48:1380-1385. [PMID: 31688605 PMCID: PMC6867665 DOI: 10.1097/mpa.0000000000001441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVES Injury of the pancreatic duct epithelial barrier plays a critical role in the development of acute pancreatitis. The activity of the nuclear factor-kappa B (NF-κB) pathway is involved in the disruption of the pancreatic duct epithelial barrier. This study investigated how NF-κB impacts the dysfunction of the pancreatic duct epithelial barrier. METHODS A human pancreatic ductal adenocarcinoma cell line was treated with tumor necrosis factor-alpha (TNF-α) and pyrrolidine dithiocarbamate. The expression levels of p65 and p-p65 were detected to evaluate NF-κB activity. Tricellulin (TRIC) expression levels were measured to assess the change in tight junction (TJ)-related proteins. The expression and localization of myosin light chain kinase (MLCK) were investigated. The structure of TJs and monolayer permeability were also examined. RESULTS NF-κB was activated by TNF-α and suppressed by pyrrolidine dithiocarbamate. Activation of NF-κB upregulated the expression levels of TRIC and MLCK. Broadened TJs were observed after NF-κB was activated. Lower monolayer permeability was observed when NF-κB was suppressed. CONCLUSIONS Activation of the NF-κB pathway induced by TNF-α leads to increased TRIC and MLCK expression, resulting in broadened TJs and high permeability, which contribute to damage to the pancreatic duct epithelial barrier.
Collapse
|
216
|
Ašmonaitė G, Sundh H, Asker N, Carney Almroth B. Rainbow Trout Maintain Intestinal Transport and Barrier Functions Following Exposure to Polystyrene Microplastics. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:14392-14401. [PMID: 30451503 DOI: 10.1021/acs.est.8b04848] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Ingestion has been proposed as a prominent exposure route for plastic debris in aquatic organisms, including fish. While the consequences of ingestion of large plastic litter are mostly understood, the impacts resulting from ingestion of microplastics (MPs) are largely unknown. We designed a study that aimed to assess impacts of MPs on fish intestinal physiology and examined integrity of extrinsic, physical and immunological barriers. Rainbow trout were exposed to polystyrene (PS) MPs (100-400 μm) via feed for a period of 4 weeks. Fish were fed four types of diets: control, diets containing virgin PS particles, or particles exposed to two different environmental matrices (sewage or harbor effluent). Extrinsic barrier disturbance in intestinal tissue was evaluated via histology. The paracellular permeability toward ions and molecules was examined using Ussing chambers and mRNA expression analysis of tight junction proteins. Active transport was monitored as transepithelial potential difference, short-circuits current and uptake rate of amino acid 3H-lysine. Immune status parameters were measured through mRNA expression level of cytokines, lysozyme activity, and hematological analysis of immune cells. We could not show that PS MPs induced inflammatory responses or acted as physical or chemical hazards upon ingestion. No measurable effects were exerted on fish intestinal permeability, active transport or electrophysiology.
Collapse
Affiliation(s)
- Giedrė Ašmonaitė
- Department of Biological and Environmental Sciences , University of Gothenburg , Medicinaregatan 18 , 413 90 Göteborg , Sweden
| | - Henrik Sundh
- Department of Biological and Environmental Sciences , University of Gothenburg , Medicinaregatan 18 , 413 90 Göteborg , Sweden
| | - Noomi Asker
- Department of Biological and Environmental Sciences , University of Gothenburg , Medicinaregatan 18 , 413 90 Göteborg , Sweden
| | - Bethanie Carney Almroth
- Department of Biological and Environmental Sciences , University of Gothenburg , Medicinaregatan 18 , 413 90 Göteborg , Sweden
| |
Collapse
|
217
|
Abstract
Acute anterior uveitis (AAU) and the spondyloarthritis (SpA) subtypes ankylosing spondylitis, reactive arthritis and psoriatic arthritis are among the inflammatory diseases affected by the biology of the intestinal microbiome. In this Review, the relationship between AAU, SpA and the microbiome is discussed, with a focus on the major SpA risk gene HLA-B*27 and how it is associated with both intestinal tolerance and the loss of ocular immune privilege that can accompany AAU. We provide four potential mechanisms to account for how dysbiosis, barrier function and immune response contribute to the development of ocular inflammation and the pathogenesis of AAU. Finally, potential therapeutic avenues to target the microbiota for the clinical management of AAU and SpA are outlined.
Collapse
Affiliation(s)
- James T Rosenbaum
- Departments of Ophthalmology, Medicine and Cell Biology, Oregon Health and Science University, Portland, OR, USA
- Legacy Devers Eye Institute, Portland, OR, USA
| | - Mark Asquith
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
218
|
Wang H, Zhai N, Chen Y, Fu C, Huang K. OTA induces intestinal epithelial barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca 2+-mediated MLCK activation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:106-112. [PMID: 29966834 DOI: 10.1016/j.envpol.2018.06.062] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/23/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Ochratoxin A (OTA) is a frequent contaminant of feed and food worldwide. The toxicity of OTA on intestinal barrier was investigated in porcine intestinal epithelial cells (IPEC-J2). We observed that OTA induced intestinal barrier dysfunction as indicated by the reduction in transepithelial electrical resistance (TEER) and elevation in paracellular permeability to 4 kDa dextran. The barrier dysfunction was accompanied with tight junction disruption including a down-regulation in ZO-1 expression and redistribution of Occludin and ZO-1. Moreover, OTA exposure increased reactive oxygen species (ROS) generation, elevated the intracellular calcium level ([Ca2+]c) and activated myosin light chain kinase (MLCK). Simultaneously, NAC, a ROS scavenger, blocked OTA-induced ROS generation, [Ca2+]c elevation, barrier dysfunction and tight junction disruption, suggesting that OTA-induced ROS generation may act as a trigger. Next, we found that OTA-induced MLCK activation was inhibited by BAPTA-AM, the cytosolic Ca2+ chelator, demonstrating that OTA-induced MLCK activation is dependent on [Ca2+]c elevation. Furthermore, inhibition of MLCK with ML-7 or inhibition of [Ca2+]c elevation with BAPTA-AM markedly prevented OTA-induced barrier dysfunction and tight junction disruption. Taken together, our results indicated that OTA induces ROS generation, and then elevates the [Ca2+]c and MLCK activity in turn, which finally induces barrier dysfunction and disrupts tight junction in IPEC-J2 cell monolayers.
Collapse
Affiliation(s)
- Hong Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Nianhui Zhai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ying Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Chongyang Fu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
219
|
Figueroa-Lozano S, de Vos P. Relationship Between Oligosaccharides and Glycoconjugates Content in Human Milk and the Development of the Gut Barrier. Compr Rev Food Sci Food Saf 2018; 18:121-139. [DOI: 10.1111/1541-4337.12400] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Susana Figueroa-Lozano
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
220
|
Van Looveren K, Libert C. Should we target TNF receptors in the intestinal epithelium with glucocorticoids during systemic inflammation? Expert Opin Ther Targets 2018; 22:1029-1037. [DOI: 10.1080/14728222.2018.1539078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kelly Van Looveren
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
221
|
Li BR, Wu J, Li HS, Jiang ZH, Zhou XM, Xu CH, Ding N, Zha JM, He WQ. In Vitro and In Vivo Approaches to Determine Intestinal Epithelial Cell Permeability. J Vis Exp 2018. [PMID: 30394372 DOI: 10.3791/57032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The intestinal barrier defends against pathogenic microorganism and microbial toxin. Its function is regulated by tight junction permeability and epithelial cell integrity, and disruption of the intestinal barrier function contributes to progression of gastrointestinal and systemic disease. Two simple methods are described here to measure the permeability of intestinal epithelium. In vitro, Caco-2BBe cells are plated in tissue culture wells as a monolayer and transepithelial electrical resistance (TER) can be measured by an epithelial (volt/ohm) meter. This method is convincing because of its user-friendly operation and repeatability. In vivo, mice are gavaged with 4 kDa fluorescein isothiocyanate (FITC)-dextran, and the FITC-dextran concentrations are measured in collected serum samples from mice to determine the epithelial permeability. Oral gavage provides an accurate dose, and therefore is the preferred method to measure the intestinal permeability in vivo. Taken together, these two methods can measure the permeability of the intestinal epithelium in vitro and in vivo, and hence be used to study the connection between diseases and barrier function.
Collapse
Affiliation(s)
- Ban-Ruo Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Jia Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Hua-Shan Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Zhi-Hui Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Xiu-Min Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Cai-Hua Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Ning Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University
| | - Juan-Min Zha
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University;
| | - Wei-Qi He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University; Department of Oncology, The First Affiliated Hospital of Soochow University;
| |
Collapse
|
222
|
Giménez-Bastida JA, Laparra-Llopis JM, Baczek N, Zielinski H. Buckwheat and buckwheat enriched products exert an anti-inflammatory effect on the myofibroblasts of colon CCD-18Co. Food Funct 2018; 9:3387-3397. [PMID: 29870039 PMCID: PMC6597957 DOI: 10.1039/c8fo00193f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Buckwheat (BW) constitutes a good source of bioactive components that show anti-inflammatory effects in vitro and in vivo. The use of functional foods in the prevention and treatment of inflammatory bowel diseases (IBDs) has aroused increasing interest. This study investigates the effect of in vitro digested BW and BW-enriched products (BW-enriched wheat breads, roasted BW groats -fermented and non-fermented-, and BW sprouts) on colon myofibroblasts, the cells involved in the regulation of inflammatory response in the intestine. The cells were treated with different digested-BW products, alone or together with TNF-α (20 ng mL-1), and the effects on the cell migration, mitochondrial membrane potential and cell cycle, processes altered during intestinal inflammation, were investigated. A significant reduction in TNF-α-induced migration (25.5%, p < 0.05) and attenuation of the TNF-α-altered cell cycle (p < 0.05) was observed in myofibroblasts treated with BW-enriched white wheat bread. These results contribute to extend the beneficial effects derived from BW bioactive compounds, and suggest that BW consumption can exert beneficial effects on IBDs.
Collapse
Affiliation(s)
- J A Giménez-Bastida
- Department of Pharmacology. Vanderbilt University School of Medicine, RRB 514, 23rd Ave. S. at Pierce, Nashville, TN 37232-6602, USA
| | | | | | | |
Collapse
|
223
|
Ponce de León-Rodríguez MDC, Guyot JP, Laurent-Babot C. Intestinal in vitro cell culture models and their potential to study the effect of food components on intestinal inflammation. Crit Rev Food Sci Nutr 2018; 59:3648-3666. [DOI: 10.1080/10408398.2018.1506734] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Jean-Pierre Guyot
- NUTRIPASS—University of Montpellier, IRD, Montpellier SupAgro, Montpellier, France
| | | |
Collapse
|
224
|
Putt KK, Pei R, White HM, Bolling BW. Yogurt inhibits intestinal barrier dysfunction in Caco-2 cells by increasing tight junctions. Food Funct 2018; 8:406-414. [PMID: 28091645 DOI: 10.1039/c6fo01592a] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation disrupts intestinal barrier function and may contribute to the pathology of obesity and other diseases. The goal of this study was to determine the mechanism by which yogurt improves intestinal barrier function. Caco-2 cells were differentiated on Transwell inserts and used as a model of intestinal barrier permeability. Transepithelial electrical resistance (TEER) and flux of 4 kDa fluorescein isothiocyanate-dextran (FD) and lucifer yellow (LY) were used as indicators of monolayer integrity and paracellular permeability. Immunofluorescence microscopy and real time quantitative polymerase chain were used to assess the localization and expression of tight junction proteins known to regulate intestinal permeability. Differentiated cells were treated with a vehicle control (C), inflammatory stimulus (I) (interleukin-1β, tumor necrosis factor-α, interferon-γ, and lipopolysaccharide), or I and 0.03 g mL-1 yogurt (IY). After 48 h, I reduced Caco-2 TEER by 46%, while IY reduced TEER by only 27% (P < 0.0001). FD and LY flux reflected TEER measurements, with IY having significantly lower permeability than I (P < 0.05). Yogurt also improved localization of occludin and zona occludens protein 1 (ZO-1) at tight junctions of differentiated Caco-2 cells. IY increased Caco-2 claudin-1, ZO-1, and occludin mRNA relative to I (P < 0.05). In a simulated digestion, the barrier-improving bioactivity of yogurt was maintained through the gastric phase, but was reduced to the level of I after intestinal digestion (P < 0.05). Therefore, yogurt improved inflammation-disrupted intestinal barrier function in a Caco-2 model by increasing tight junctions, but the beneficial effect on barrier function was reduced at latter stages of digestion.
Collapse
Affiliation(s)
- Kelley K Putt
- Department of Food Science, University of Wisconsin-Madison, 1605 Linden Dr., Madison, WI 53706, USA.
| | - Ruisong Pei
- Department of Food Science, University of Wisconsin-Madison, 1605 Linden Dr., Madison, WI 53706, USA.
| | - Heather M White
- Department of Animal Science, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Bradley W Bolling
- Department of Food Science, University of Wisconsin-Madison, 1605 Linden Dr., Madison, WI 53706, USA.
| |
Collapse
|
225
|
Jiang L, Lv J, Liu J, Hao X, Ren F, Guo H. Donkey milk lysozyme ameliorates dextran sulfate sodium-induced colitis by improving intestinal barrier function and gut microbiota composition. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
226
|
Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med 2018; 50:1-9. [PMID: 30115904 PMCID: PMC6095905 DOI: 10.1038/s12276-018-0126-x] [Citation(s) in RCA: 963] [Impact Index Per Article: 137.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is a specialized organ in which dynamic interactions between host cells and the complex environment occur in addition to food digestion. Together with the chemical barrier of the mucosal layer and the cellular immune system, the epithelial cell layer performs a pivotal role as the first physical barrier against external factors and maintains a symbiotic relationship with commensal bacteria. The tight junction proteins, including occludin, claudins, and zonula occludens, are crucial for the maintenance of epithelial barrier integrity. To allow the transport of essential molecules and restrict harmful substances, the intracellular signaling transduction system and a number of extracellular stimuli such as cytokines, small GTPases, and post-translational modifications dynamically modulate the tight junction protein complexes. An imbalance in these regulations leads to compromised barrier integrity and is linked with pathological conditions. Despite the obscurity of the causal relationship, the loss of barrier integrity is considered to contribute to inflammatory bowel disease, obesity, and metabolic disorders. The elucidation of the role of diseases in barrier integrity and the underlying regulatory mechanisms have improved our understanding of the intestinal barrier to allow the development of novel and potent therapeutic approaches. A better understanding of how the cells that line the inside of the intestines allow nutrients in, while keeping harmful substances and pathogens out could lead to new therapies for inflammatory bowel disease, obesity, and other conditions. A team from South Korea led by Sung Ho Ryu from Pohang University of Science and Technology review the regulatory mechanisms that help maintain the intestinal epithelial barrier. They discuss the role of tight junction proteins in forming a seal between adjacent cells and the various signaling pathways that loosen or tighten these junctions to enable limited transport. Loss of barrier integrity because of genetics, gut microbes, auto-immunity, diet, or other factors is often implicated in disease, and restoring barrier function with drugs or probiotics could help ameliorate many health problems.
Collapse
|
227
|
Lu Y, Li L, Zhang JW, Zhong XQ, Wei JA, Han L. Total polysaccharides of the Sijunzi decoction attenuate tumor necrosis factor-α-induced damage to the barrier function of a Caco-2 cell monolayer via the nuclear factor-κB-myosin light chain kinase-myosin light chain pathway. World J Gastroenterol 2018; 24:2867-2877. [PMID: 30018481 PMCID: PMC6048434 DOI: 10.3748/wjg.v24.i26.2867] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/25/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the protective effects and underlying mechanisms of total polysaccharides of the Sijunzi decoction (TPSJ) on the epithelial barriers in vitro.
METHODS Caco-2 cell monolayers were treated with or without TPSJ in the presence or absence of TNF-α, and paracellular permeability and transepithelial electrical resistance (TEER) were measured to evaluate the epithelial barrier function. Immunofluorescence and western blotting were respectively used to evaluate the distribution and expression of the tight junction proteins claudin 1, claudin 2, zo3, and occludin in Caco-2 cells. Western blotting was also used to evaluate the cellular expression of myosin light chain (MLC), phosphorylated MLC (pMLC), MLC kinase (MLCK), and nuclear factor (NF)-κB p65.
RESULTS TPSJ promoted the proliferation of Caco-2 cells and inhibited TNF-α-induced secretion of pro-inflammatory cytokines. Furthermore, TPSJ significantly ameliorated both the reduction of TEER and the increased paracellular permeability observed in tumor necrosis factor (TNF)-α-damaged Caco-2 monolayers. Furthermore, TPSJ remarkably attenuated TNF-α-induced morphological changes, downregulated the expression of claudin 1, claudin 2, zo3, and occludin, and markedly suppressed TNF-α-mediated upregulation of p-MLC and MLCK expression. Finally, TPSJ inhibited the activation and expression of NF-κB p65.
CONCLUSION Our results demonstrate that TPSJ alleviates the TNF-α-induced impairment of the intestinal epithelial cell barrier function by suppressing NF-κB p65-mediated phosphorylation of MLCK and MLC.
Collapse
Affiliation(s)
- Yue Lu
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Leng Li
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Jin-Wei Zhang
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Xiao-Qin Zhong
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Jian-An Wei
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Ling Han
- The Second Clinical College (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|
228
|
Patra AK, Amasheh S, Aschenbach JR. Modulation of gastrointestinal barrier and nutrient transport function in farm animals by natural plant bioactive compounds – A comprehensive review. Crit Rev Food Sci Nutr 2018; 59:3237-3266. [DOI: 10.1080/10408398.2018.1486284] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Amlan Kumar Patra
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, Berlin, Germany
- Institute of Animal Nutrition, West Bengal University of Animal and Fishery Sciences, 37 K. B. Sarani, Belgachia, Kolkata, India
| | - Salah Amasheh
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, Berlin, Germany
| | - Jörg Rudolf Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, Berlin, Germany
| |
Collapse
|
229
|
Luo L, Zhou Z, Xue J, Wang Y, Zhang J, Cai X, Liu Y, Yang F. Bletilla striata polysaccharide has a protective effect on intestinal epithelial barrier disruption in TAA-induced cirrhotic rats. Exp Ther Med 2018; 16:1715-1722. [PMID: 30186392 PMCID: PMC6122099 DOI: 10.3892/etm.2018.6430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
It has been reported that intestinal epithelial barrier dysfunction serves an important role in the development of liver cirrhosis. However, at present there is no satisfactory treatment for intestinal mucosal lesions and ulcers associated with cirrhosis. The aim of the present study was to investigate the effect of Bletilla striata polysaccharide (BSP) on intestinal epithelial barrier disruption in rats with thioacetamide (TAA)-induced liver cirrhosis. Rats were randomly divided into 5 groups (n=10): BSP low dosage (15 mg/kg), BSP middle dosage (30 mg/kg), BSP high dosage (60 mg/kg), experiment and control groups. All groups except control group were administered with TAA (200 mg/kg/day) to induce liver cirrhosis. Following modeling, rats in the low, middle and high-dose BSP groups received BSP. ELISA kits were used to measure the endotoxin, alanine transaminase (ALT) and aspartate transaminase (AST) levels in the portal vein, while interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression in the ileal tissue was measured. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to detect the expression of zonula occludens (ZO)-1 and occludin mRNA and protein, respectively. Intestinal epithelial tissue pathology was detected using hematoxylin-eosin (HE) staining. Immunohistochemistry was performed to assess the expression of ZO-1 and occludin in intestinal epithelial tissues. Following treatment with BSP, ALT, AST and endotoxin levels in the portal vein, as well as IL-6 and TNF-α expression in ileal tissues, were significantly decreased compared with model group (P<0.05 or 0.01). Furthermore, BSP treatment upregulated the expression of ZO-1 and occludin mRNA and protein compared with the model group (P<0.05 or 0.01) and cytoplasmic staining for these proteins was increased. The degree of intestinal epithelial tissue pathological damage was significantly reduced in the BSP groups. In conclusion, BSP is able to reduce endotoxin levels, inhibit the inflammatory cytokines IL-6 and TNF-α and elevate expression of ZO-1 and occludin at tight junctions. Together, these results suggest a novel protective agent for the restoration of intestinal epithelial barrier disruption.
Collapse
Affiliation(s)
- Lei Luo
- Department of Gastroenterology, The Second People's Hospital of Yichang, Yichang, Hubei 443000, P.R. China
| | - Zhang Zhou
- Department of Anesthesia, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Juan Xue
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei 430015, P.R. China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Juan Zhang
- Department of Pulmonary Diseases, Jingmen City Hospital of Traditional Chinese Medicine, Jingmen, Hubei 448000, P.R. China
| | - Xin Cai
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, Hubei 430060, P.R. China
| | - Yuqing Liu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, Hubei 430060, P.R. China
| | - Fan Yang
- Department of Hepatology, Hubei Provincial Hospital of Chinese Medicine, Wuhan, Hubei 430074, P.R. China
| |
Collapse
|
230
|
Barbaro MR, Fuschi D, Cremon C, Carapelle M, Dino P, Marcellini MM, Dothel G, De Ponti F, Stanghellini V, Barbara G. Escherichia coli Nissle 1917 restores epithelial permeability alterations induced by irritable bowel syndrome mediators. Neurogastroenterol Motil 2018; 30:e13388. [PMID: 29956419 DOI: 10.1111/nmo.13388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal permeability is altered in a subgroup of irritable bowel syndrome (IBS) patients and may contribute to symptom development. The aim of this study was to evaluate the in vitro effect of the probiotic Escherichia coli Nissle 1917 (EcN) on Caco-2 permeability alterations induced by mediators released by IBS mucosal biopsies compared to asymptomatic controls (AC). METHODS Caco-2 cells were used as an in vitro model of intestinal permeability. Seven AC and 28 well-phenotyped IBS (9 IBS-D, 8 IBS-C, and 11 IBS-M) patients were enrolled. Mucosal mediators spontaneously released (SUP) by IBS and AC biopsies were collected. Two concentrations of EcN (108 and 106 ) were applied to Caco-2 with or without SUP or SLIGRL (a protease-activated receptor-2 activating peptide), tumor necrosis factor-α, and interferon-γ. Paracellular permeability was assessed by evaluating the flow of sulfonic-acid conjugated to fluorescein through Caco-2 monolayer. KEY RESULTS EcN 108 significantly reinforced Caco-2 monolayer compared to cells incubated with medium alone. IBS SUP induced a significant increase in paracellular permeability compared to AC SUP, independently of IBS bowel habit. EcN 108 induced a significant recovery of permeability rate compared to IBS SUP. Permeability increase induced by IBS SUP significantly correlated with severity and frequency of abdominal pain and abdominal distension. The co-incubation of EcN and IBS SUP abolished the above significant correlations. CONCLUSIONS AND INFERENCES EcN reinforces the integrity of Caco-2 monolayer and reverts the increase of permeability induced by mediators released by IBS biopsies. Future studies should investigate EcN therapeutic potentials in IBS.
Collapse
Affiliation(s)
- M R Barbaro
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - D Fuschi
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - C Cremon
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - M Carapelle
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - P Dino
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - M M Marcellini
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - G Dothel
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - F De Ponti
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - V Stanghellini
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - G Barbara
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
231
|
Huang Y, Feng Y, Wang Y, Wang P, Wang F, Ren H. Severe Burn-Induced Intestinal Epithelial Barrier Dysfunction Is Associated With Endoplasmic Reticulum Stress and Autophagy in Mice. Front Physiol 2018; 9:441. [PMID: 29740349 PMCID: PMC5925571 DOI: 10.3389/fphys.2018.00441] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 04/06/2018] [Indexed: 12/24/2022] Open
Abstract
The disruption of intestinal barrier plays a vital role in the pathophysiological changes after severe burn injury, however, the underlying mechanisms are poorly understood. Severe burn causes the disruption of intestinal tight junction (TJ) barrier. Previous studies have shown that endoplasmic reticulum (ER) stress and autophagy are closely associated with the impairment of intestinal mucosa. Thus, we hypothesize that ER stress and autophagy are likely involved in burn injury-induced intestinal epithelial barrier dysfunction. Mice received a 30% total body surface area (TBSA) full-thickness burn, and were sacrificed at 0, 1, 2, 6, 12 and 24 h postburn. The results showed that intestinal permeability was increased significantly after burn injury, accompanied by the damage of mucosa and the alteration of TJ proteins. Severe burn induced ER stress, as indicated by increased intraluminal chaperone binding protein (BIP), CCAAT/enhancer-binding protein homologous protein (CHOP) and inositol-requiring enzyme 1(IRE1)/X-box binding protein 1 splicing (XBP1). Autophagy was activated after burn injury, as evidenced by the increase of autophagy related protein 5 (ATG5), Beclin 1 and LC3II/LC3I ratio and the decrease of p62. Besides, the number of autophagosomes was also increased after burn injury. The levels of p-PI3K(Ser191), p-PI3K(Ser262), p-AKT(Ser473), and p-mTOR were decreased postburn, suggesting that autophagy-related PI3K/AKT/mTOR pathway is involved in the intestinal epithelial barrier dysfunction following severe burn. In summary, severe burn injury induces the ER stress and autophagy in intestinal epithelia, leading to the disruption of intestinal barrier.
Collapse
Affiliation(s)
- Yalan Huang
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanhai Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yu Wang
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Pei Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fengjun Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hui Ren
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
232
|
Molecular Pathophysiology of Epithelial Barrier Dysfunction in Inflammatory Bowel Diseases. Proteomes 2018; 6:proteomes6020017. [PMID: 29614738 PMCID: PMC6027334 DOI: 10.3390/proteomes6020017] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/13/2022] Open
Abstract
Over the years, the scientific community has explored myriads of theories in search of the etiology and a cure for inflammatory bowel disease (IBD). The cumulative evidence has pointed to the key role of the intestinal barrier and the breakdown of these mechanisms in IBD. More and more scientists and clinicians are embracing the concept of the impaired intestinal epithelial barrier and its role in the pathogenesis and natural history of IBD. However, we are missing a key tool that bridges these scientific insights to clinical practice. Our goal is to overcome the limitations in understanding the molecular physiology of intestinal barrier function and develop a clinical tool to assess and quantify it. This review article explores the proteins in the intestinal tissue that are pivotal in regulating intestinal permeability. Understanding the molecular pathophysiology of impaired intestinal barrier function in IBD may lead to the development of a biochemical method of assessing intestinal tissue integrity which will have a significant impact on the development of novel therapies targeting the intestinal mucosa.
Collapse
|
233
|
Chen K, Zhou XQ, Jiang WD, Wu P, Liu Y, Jiang J, Kuang SY, Tang L, Tang WN, Zhang YA, Feng L. Impaired intestinal immune barrier and physical barrier function by phosphorus deficiency: Regulation of TOR, NF-κB, MLCK, JNK and Nrf2 signalling in grass carp (Ctenopharyngodon idella) after infection with Aeromonas hydrophila. FISH & SHELLFISH IMMUNOLOGY 2018; 74:175-189. [PMID: 29305994 DOI: 10.1016/j.fsi.2017.12.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 06/07/2023]
Abstract
In aquaculture, the occurrence of enteritis has increased and dietary nutrition is considered as one of the major strategies to solve this problem. In the present study, we assume that dietary phosphorus might enhance intestinal immune barrier and physical barrier function to reduce the occurrence of enteritis in fish. To test this assumption, a total of 540 grass carp (Ctenopharyngodon idella) were investigated by feeding graded levels of available phosphorus (0.95-8.75 g/kg diet) and then infection with Aeromonas hydrophila. The results firstly showed that phosphorus deficiency decreased the ability to combat enteritis, which might be related to the impairment of intestinal immune barrier and physical barrier function. Compared with optimal phosphorus level, phosphorus deficiency decreased fish intestinal antimicrobial substances activities or contents and down-regulated antimicrobial peptides mRNA levels leading to the impairment of intestinal immune response. Phosphorus deficiency down-regulated fish intestinal anti-inflammatory cytokines mRNA levels and up-regulated the mRNA levels of pro-inflammatory cytokines [except IL-1β and IL-12p35 in distal intestine (DI) and IL-12p40] causing aggravated of intestinal inflammatory responses, which might be related to the signalling molecules target of rapamycin and nuclear factor kappa B. In addition, phosphorus deficiency disturbed fish intestinal tight junction function and induced cell apoptosis as well as oxidative damage leading to impaired of fish intestinal physical barrier function, which might be partially associated with the signalling molecules myosin light chain kinase, c-Jun N-terminal protein kinase and NF-E2-related factor 2, respectively. Finally, based on the ability to combat enteritis, dietary available phosphorus requirement for grass carp (254.56-898.23 g) was estimated to be 4.68 g/kg diet.
Collapse
Affiliation(s)
- Kang Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China.
| |
Collapse
|
234
|
Tricellulin is regulated via interleukin-13-receptor α2, affects macromolecule uptake, and is decreased in ulcerative colitis. Mucosal Immunol 2018; 11:345-356. [PMID: 28612843 PMCID: PMC5730503 DOI: 10.1038/mi.2017.52] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/01/2017] [Indexed: 02/04/2023]
Abstract
In the two inflammatory bowel diseases, ulcerative colitis (UC) and Crohn's disease (CD), altered expression of tight junction (TJ) proteins leads to an impaired epithelial barrier including increased uptake of luminal antigens supporting the inflammation. Here, we focused on regulation of tricellulin (Tric), a protein of the tricellular TJ essential for the barrier against macromolecules, and hypothesized a role in paracellular antigen uptake. We report that Tric is downregulated in UC, but not in CD, and that its reduction increases the passage of macromolecules. Using a novel visualization method, passage sites were identified at TJ regions usually sealed by Tric. We show that interleukin-13 (IL-13), beyond its known effect on claudin-2, downregulates Tric expression. These two effects of IL-13 are regulated by different signaling pathways: The IL-13 receptor α1 upregulates claudin-2, whereas IL-13 receptor α2 downregulates Tric. We suggest to target the α2 receptor in future developments of therapeutical IL-13-based biologicals.
Collapse
|
235
|
Van Itallie CM, Tietgens AJ, Aponte A, Gucek M, Cartagena-Rivera AX, Chadwick RS, Anderson JM. MARCKS-related protein regulates cytoskeletal organization at cell-cell and cell-substrate contacts in epithelial cells. J Cell Sci 2018; 131:jcs.210237. [PMID: 29222109 DOI: 10.1242/jcs.210237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/04/2017] [Indexed: 01/14/2023] Open
Abstract
Treatment of epithelial cells with interferon-γ and TNF-α (IFN/TNF) results in increased paracellular permeability. To identify relevant proteins mediating barrier disruption, we performed proximity-dependent biotinylation (BioID) of occludin and found that tagging of MARCKS-related protein (MRP; also known as MARCKSL1) increased ∼20-fold following IFN/TNF administration. GFP-MRP was focused at the lateral cell membrane and its overexpression potentiated the physiological response of the tight junction barrier to cytokines. However, deletion of MRP did not abrogate the cytokine responses, suggesting that MRP is not required in the occludin-dependent IFN/TNF response. Instead, our results reveal a key role for MRP in epithelial cells in control of multiple actin-based structures, likely by regulation of integrin signaling. Changes in focal adhesion organization and basal actin stress fibers in MRP-knockout (KO) cells were reminiscent of those seen in FAK-KO cells. In addition, we found alterations in cell-cell interactions in MRP-KO cells associated with increased junctional tension, suggesting that MRP may play a role in focal adhesion-adherens junction cross talk. Together, our results are consistent with a key role for MRP in cytoskeletal organization of cell contacts in epithelial cells.
Collapse
Affiliation(s)
- Christina M Van Itallie
- Laboratory of Tight Junction Structure and Function, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - Amber Jean Tietgens
- Laboratory of Tight Junction Structure and Function, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - Angel Aponte
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - Marjan Gucek
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - Alexander X Cartagena-Rivera
- Section on Auditory Mechanics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - Richard S Chadwick
- Section on Auditory Mechanics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| | - James M Anderson
- Laboratory of Tight Junction Structure and Function, National Institutes of Health, Building 50, Room 4525, 50 South Drive, Bethesda, MD 20892, USA
| |
Collapse
|
236
|
Moussa L, Usunier B, Demarquay C, Benderitter M, Tamarat R, Sémont A, Mathieu N. Bowel Radiation Injury: Complexity of the Pathophysiology and Promises of Cell and Tissue Engineering. Cell Transplant 2018; 25:1723-1746. [PMID: 27197023 DOI: 10.3727/096368916x691664] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ionizing radiation is effective to treat malignant pelvic cancers, but the toxicity to surrounding healthy tissue remains a substantial limitation. Early and late side effects not only limit the escalation of the radiation dose to the tumor but may also be life-threatening in some patients. Numerous preclinical studies determined specific mechanisms induced after irradiation in different compartments of the intestine. This review outlines the complexity of the pathogenesis, highlighting the roles of the epithelial barrier in the vascular network, and the inflammatory microenvironment, which together lead to chronic fibrosis. Despite the large number of pharmacological molecules available, the studies presented in this review provide encouraging proof of concept regarding the use of mesenchymal stromal cell (MSC) therapy to treat radiation-induced intestinal damage. The therapeutic efficacy of MSCs has been demonstrated in animal models and in patients, but an enormous number of cells and multiple injections are needed due to their poor engraftment capacity. Moreover, it has been observed that although MSCs have pleiotropic effects, some intestinal compartments are less restored after a high dose of irradiation. Future research should seek to optimize the efficacy of the injected cells, particularly with regard to extending their life span in the irradiated tissue. Moreover, improving the host microenvironment, combining MSCs with other specific regenerative cells, or introducing new tissue engineering strategies could be tested as methods to treat the severe side effects of pelvic radiotherapy.
Collapse
Affiliation(s)
- Lara Moussa
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Benoît Usunier
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Christelle Demarquay
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Marc Benderitter
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Radia Tamarat
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Alexandra Sémont
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Noëlle Mathieu
- Institut de Radioprotection et de SÛreté Nucléaire (IRSN), PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| |
Collapse
|
237
|
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: 155] [Impact Index Per Article: 22.1] [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
|
238
|
Alhagamhmad MH. Enteral Nutrition in the Management of Crohn's Disease: Reviewing Mechanisms of Actions and Highlighting Potential Venues for Enhancing the Efficacy. Nutr Clin Pract 2018; 33:483-492. [PMID: 29323428 DOI: 10.1002/ncp.10004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/12/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022] Open
Abstract
Crohn's disease (CD) is a chronic condition that affects the gut and has adverse effects on growth and development. There is a global increase in the incidence and prevalence rates, and several factors are believed to contribute to this rise, including dietary habits. In contrast, the use of enteral nutrition (EN) as an exclusive source of nutrition is increasingly becoming the preferred induction treatment of pediatric CD patients in part to address the nutrition complications. However, EN therapy is considered less effective in adults with CD. A better understanding of the molecular mechanisms of enteral therapy will help improve the clinical management of CD. It is increasingly becoming evident that the therapeutic utility of EN is in part due to the reversal of the microbial changes and the direct immunomodulatory effects. Moreover, there is a potential tendency for enhancing the efficacy of EN therapy by improving the palatability of the given formulas and, more important, by magnifying the anti-inflammatory properties. Recent observations have shown that the immunomodulatory effects of EN are mediated at least in part by blocking nuclear factor-κB. Furthermore, it is likely that several ingredients of EN contribute to this activity, in particular glutamine and arginine amino acids. In addition, manipulating the composition of EN therapy by altering concentrations of the key ingredients is found to have the potential for more efficient therapy. In this review, the underlying mechanisms of EN actions will be discussed further with a focus on the potential methods for enhancing the efficacy.
Collapse
Affiliation(s)
- Moftah H Alhagamhmad
- Faculty of Medicine, University of Benghazi (Al-Arab Medical University), Benghazi, Libya
| |
Collapse
|
239
|
Buckley A, Turner JR. Cell Biology of Tight Junction Barrier Regulation and Mucosal Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029314. [PMID: 28507021 DOI: 10.1101/cshperspect.a029314] [Citation(s) in RCA: 432] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mucosal surfaces are lined by epithelial cells. In the intestine, the epithelium establishes a selectively permeable barrier that supports nutrient absorption and waste secretion while preventing intrusion by luminal materials. Intestinal epithelia therefore play a central role in regulating interactions between the mucosal immune system and luminal contents, which include dietary antigens, a diverse intestinal microbiome, and pathogens. The paracellular space is sealed by the tight junction, which is maintained by a complex network of protein interactions. Tight junction dysfunction has been linked to a variety of local and systemic diseases. Two molecularly and biophysically distinct pathways across the intestinal tight junction are selectively and differentially regulated by inflammatory stimuli. This review discusses the mechanisms underlying these events, their impact on disease, and the potential of using these as paradigms for development of tight junction-targeted therapeutic interventions.
Collapse
Affiliation(s)
- Aaron Buckley
- Departments of Pathology and Medicine (Gastroenterology), Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Jerrold R Turner
- Departments of Pathology and Medicine (Gastroenterology), Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
240
|
Ubah OC, Steven J, Kovaleva M, Ferguson L, Barelle C, Porter AJR, Barelle CJ. Novel, Anti-hTNF-α Variable New Antigen Receptor Formats with Enhanced Neutralizing Potency and Multifunctionality, Generated for Therapeutic Development. Front Immunol 2017; 8:1780. [PMID: 29312310 PMCID: PMC5743654 DOI: 10.3389/fimmu.2017.01780] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022] Open
Abstract
The management of chronic inflammatory diseases, such as inflammatory bowel disease, psoriasis, and rheumatoid arthritis has significantly improved over the last decade with the clinical availability of anti-TNF-α biologics. Despite this undoubted treatment success, a combination of acquired resistance together with an increased risk of systemic complications, means that a significant number of patients either fail to find a suitable targeted therapy or frustratingly discover that an approach that did work is no longer efficacious. Here, we report the isolation and characterization of a new class of super-neutralizing anti-TNF-α biologics formats, the building blocks of which were originally derived as variable new antigen receptor (VNAR) domains from an immunized nurse shark. These parental small, stable VNAR monomers recognize and neutralize tumor necrosis factor (TNF)-α, in cell-based assays, at nanomolar concentrations. However, the simple, single-chain molecular architecture of VNARs allows for easy and multiple reformatting options. Through reformatting, we achieved a 50,000-fold enhancement in in vitro efficacy with super-neutralizing fusion proteins able to block TNF-α induced cytotoxicity in the 2–5 pM range while retaining other functionality through the addition of fusion proteins known to extend serum half-life in vivo. In an in vitro intestinal epithelial barrier dysfunction efficacy model, the lead VNAR domains, restored barrier function and prevented paracellular flux with comparable efficacy to adalimumab (Humira®). In addition, all multivalent VNAR constructs restored trans-epithelial electrical resistance (TEER) to approximately 94% of the untreated control. Reformatted VNAR domains should be considered as a new class of biologic agents for the treatment of hTNF-α driven diseases; either used systemically with appropriate half-life extension or alternatively where site-specific delivery of small and stable neutralizers may provide improvements to current therapy options.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew J R Porter
- Elasmogen Ltd., Aberdeen, Scotland.,Scottish Biologics Facility, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland
| | | |
Collapse
|
241
|
Lysophosphatidic Acid Receptor 1 Is Important for Intestinal Epithelial Barrier Function and Susceptibility to Colitis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:353-366. [PMID: 29128569 DOI: 10.1016/j.ajpath.2017.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/28/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023]
Abstract
Intestinal epithelial cells form a barrier that is critical in protecting the host from the hostile luminal environment. Previously, we showed that lysophosphatidic acid (LPA) receptor 1 regulates proliferation of intestinal epithelial cells, such that the absence of LPA1 mitigates the epithelial wound healing process. This study provides evidence that LPA1 is important for the maintenance of epithelial barrier integrity. The epithelial permeability, determined by fluorescently labeled dextran flux and transepithelial resistance, is increased in the intestine of mice with global deletion of Lpar1, Lpar1-/- (Lpa1-/-). Serum liposaccharide level and bacteria loads in the intestinal mucosa and peripheral organs were elevated in Lpa1-/- mice. Decreased claudin-4, caudin-7, and E-cadherin expression in Lpa1-/- mice further suggested defective apical junction integrity in these mice. Regulation of LPA1 expression in Caco-2 cells modulated epithelial permeability and the expression levels of junctional proteins. The increased epithelial permeability in Lpa1-/- mice correlated with increased susceptibility to an experimental model of colitis. This resulted in more severe inflammation and increased mortality compared with control mice. Treatment of Caco-2 cells with tumor necrosis factor-α and interferon-γ significantly increased paracellular permeability, which was blocked by cotreatment with LPA, but not LPA1 knockdown cells. Similarly, orally given LPA blocked tumor necrosis factor-mediated intestinal barrier defect in mice. LPA1 plays a significant role in maintenance of epithelial barrier in the intestine via regulation of apical junction integrity.
Collapse
|
242
|
Hill DR, Huang S, Nagy MS, Yadagiri VK, Fields C, Mukherjee D, Bons B, Dedhia PH, Chin AM, Tsai YH, Thodla S, Schmidt TM, Walk S, Young VB, Spence JR. Bacterial colonization stimulates a complex physiological response in the immature human intestinal epithelium. eLife 2017; 6:29132. [PMID: 29110754 PMCID: PMC5711377 DOI: 10.7554/elife.29132] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/29/2017] [Indexed: 12/19/2022] Open
Abstract
The human gastrointestinal tract is immature at birth, yet must adapt to dramatic changes such as oral nutrition and microbial colonization. The confluence of these factors can lead to severe inflammatory disease in premature infants; however, investigating complex environment-host interactions is difficult due to limited access to immature human tissue. Here, we demonstrate that the epithelium of human pluripotent stem-cell-derived human intestinal organoids is globally similar to the immature human epithelium and we utilize HIOs to investigate complex host-microbe interactions in this naive epithelium. Our findings demonstrate that the immature epithelium is intrinsically capable of establishing a stable host-microbe symbiosis. Microbial colonization leads to complex contact and hypoxia driven responses resulting in increased antimicrobial peptide production, maturation of the mucus layer, and improved barrier function. These studies lay the groundwork for an improved mechanistic understanding of how colonization influences development of the immature human intestine.
Collapse
Affiliation(s)
- David R Hill
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Sha Huang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Melinda S Nagy
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Veda K Yadagiri
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Courtney Fields
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Dishari Mukherjee
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, United States
| | - Brooke Bons
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Priya H Dedhia
- Department of Surgery, University of Michigan, Ann Arbor, United States
| | - Alana M Chin
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Yu-Hwai Tsai
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Shrikar Thodla
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, United States
| | - Seth Walk
- Department of Microbiology and Immunology, Montana State University, Bozeman, United States
| | - Vincent B Young
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Jason R Spence
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States.,Department of Cell andDevelopmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
243
|
Marlicz W, Yung DE, Skonieczna-Żydecka K, Loniewski I, van Hemert S, Loniewska B, Koulaouzidis A. From clinical uncertainties to precision medicine: the emerging role of the gut barrier and microbiome in small bowel functional diseases. Expert Rev Gastroenterol Hepatol 2017; 11:961-978. [PMID: 28618973 DOI: 10.1080/17474124.2017.1343664] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the last decade, remarkable progress has been made in the understanding of disease pathophysiology. Many new theories expound on the importance of emerging factors such as microbiome influences, genomics/omics, stem cells, innate intestinal immunity or mucosal barrier complexities. This has introduced a further dimension of uncertainty into clinical decision-making, but equally, may shed some light on less well-understood and difficult to manage conditions. Areas covered: Comprehensive review of the literature on gut barrier and microbiome relevant to small bowel pathology. A PubMed/Medline search from 1990 to April 2017 was undertaken and papers from this range were included. Expert commentary: The scenario of clinical uncertainty is well-illustrated by functional gastrointestinal disorders (FGIDs). The movement towards achieving a better understanding of FGIDs is expressed in the Rome IV guidelines. Novel diagnostic and therapeutic protocols focused on the GB and SB microbiome can facilitate diagnosis, management and improve our understanding of the underlying pathological mechanisms in FGIDs.
Collapse
Affiliation(s)
- Wojciech Marlicz
- a Department of Gastroenterology , Pomeranian Medical University , Szczecin , Poland
| | - Diana E Yung
- b Centre for Liver and Digestive Disorders , Royal Infirmary of Edinburgh , Edinburgh , United Kingdom
| | | | - Igor Loniewski
- c Department of Biochemistry and Human Nutrition , Pomeranian Medical University , Szczecin , Poland.,d Sanprobi Sp. z o.o. Sp. K , Szczecin , Poland
| | | | - Beata Loniewska
- f Department of Neonatal Diseases , Pomeranian Medical University , Szczecin , Poland
| | - Anastasios Koulaouzidis
- g Centre for Liver and Digestive Disorders , Royal Infirmary of Edinburgh , Edinburgh , United Kingdom
| |
Collapse
|
244
|
Takigawa M, Iida M, Nagase S, Suzuki H, Watari A, Tada M, Okada Y, Doi T, Fukasawa M, Yagi K, Kunisawa J, Kondoh M. Creation of a Claudin-2 Binder and Its Tight Junction-Modulating Activity in a Human Intestinal Model. J Pharmacol Exp Ther 2017; 363:444-451. [PMID: 28928120 DOI: 10.1124/jpet.117.242214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/31/2017] [Indexed: 01/03/2023] Open
Abstract
Disruption of the gastrointestinal epithelial barrier is a hallmark of chronic inflammatory bowel diseases (IBDs). The transmembrane protein claudin 2 (CLDN2) is a component of epithelial tight junctions (TJs). In the intestines of patients with IBDs, the expression of the pore-forming TJ protein CLDN2 is upregulated. Although CLDN2 is involved in these leaky barriers, whether it can be a target to enhance TJ integrity is unknown because a CLDN2-specific inhibitor has not been developed. Here, we used DNA immunization to generate a monoclonal antibody (mAb) that recognized an extracellular loop of CLDN2. Treatment of epithelial cell monolayers with the mAb increased barrier integrity. In addition, the anti-CLDN2 mAb attenuated the decrease in TJ integrity induced by the proinflammatory cytokine tumor necrosis factor-α (TNF-α), and cotreatment of cells with anti-TNF-α mAb and anti-CLDN2 mAb showed additive attenuating effects. These findings indicate that CLDN2 may be a target for enhancing TJ integrity, and CLDN2 binder may be an enhancer of mucosal barrier integrity and a potential therapeutic option for IBDs.
Collapse
Affiliation(s)
- Mutsumi Takigawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Manami Iida
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Shotaro Nagase
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Hidehiko Suzuki
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Akihiro Watari
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Minoru Tada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Masayoshi Fukasawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Kiyohito Yagi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Jun Kunisawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Mu.T., M.I., S.N., A.W., Y.O., T.D., K.Y., J.K., M.K.); Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institute of Biomedical Innovation, Osaka, Japan (Mu.T., H.S., J.K.); Division of Biological Chemistry and Biologicals, National Institutes of Health Sciences, Tokyo, Japan (Mi.T.); Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan (M.F.); Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan (J.K.); Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (J.K.); and Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan (J.K.)
| |
Collapse
|
245
|
Shashikanth N, Yeruva S, Ong MLDM, Odenwald MA, Pavlyuk R, Turner JR. Epithelial Organization: The Gut and Beyond. Compr Physiol 2017; 7:1497-1518. [DOI: 10.1002/cphy.c170003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
246
|
Abstract
The gastrointestinal mucosa constitutes a critical barrier where millions of microbes and environmental antigens come in close contact with the host immune system. Intestinal barrier defects have been associated with a broad range of diseases and therefore denote a new therapeutic target. Areas covered: This review is based on an extensive literature search in PubMed of how the intestinal barrier contributes to health and as a trigger for disease. It discusses the anatomy of the intestinal barrier and explains the available methods to evaluate its function. Also reviewed is the importance of diet and lifestyle factors on intestinal barrier function, and three prototypes of chronic diseases (inflammatory bowel disease, celiac disease and nonalcoholic fatty liver disease) that have been linked to barrier defects are discussed. Expert commentary: The intestinal barrier has been investigated by various methods, but correlation of results across studies is difficult, representing a major shortcoming in the field. New upcoming techniques and research on the effect of barrier-restoring therapeutics may improve our current understanding of the gut barrier, and provide a step forward towards personalised medicine.
Collapse
|
247
|
Zhang Y, Han Y, Zhao Y, Lv Y, Hu Y, Tan Y, Bi X, Yu B, Kou J. DT-13 Ameliorates TNF-α-Induced Vascular Endothelial Hyperpermeability via Non-Muscle Myosin IIA and the Src/PI3K/Akt Signaling Pathway. Front Immunol 2017; 8:925. [PMID: 28855900 PMCID: PMC5557769 DOI: 10.3389/fimmu.2017.00925] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022] Open
Abstract
DT-13(25(R,S)-ruscogenin-1-O-[β-d-glucopyranosyl-(1→2)][β-d-xylopyranosyl-(1→3)]-β-d-fucopyranoside) has been identified as an important factor in TNF-α-induced vascular inflammation. However, the effect of DT-13 on TNF-α-induced endothelial permeability and the potential molecular mechanisms remain unclear. Hence, this study was undertaken to elucidate the protective effect of DT-13 on TNF-α-induced endothelial permeability and the underlying mechanisms in vivo and in vitro. The in vivo results showed that DT-13 could ameliorate endothelial permeability in mustard oil-induced plasma leakage in the skin and modulate ZO-1 organization. In addition, the in vitro results showed that pretreatment with DT-13 could increase the transendothelial electrical resistance value and decrease the sodium fluorescein permeability coefficient. Moreover, DT-13 altered the mRNA and protein levels of ZO-1 as determined by real-time PCR, Western blotting, and immunofluorescence analyses. DT-13 treatment decreased the phosphorylations of Src, PI3K, and Akt in TNF-α-treated human umbilical vein endothelial cells (HUVECs). Further analyses with PP2 (10 µM, inhibitor of Src) indicated that DT-13 modulated endothelial permeability in TNF-α-induced HUVECs in an Src-dependent manner. LY294002 (10 µM, PI3K inhibitor) also had the same effect on DT-13 but did not affect phosphorylation of Src. Following decreased expression of non-muscle myosin IIA (NMIIA), the effect of DT-13 on the phosphorylations of Src, PI3K, and Akt was abolished. This study provides pharmacological evidence showing that DT-13 significantly ameliorated the TNF-α-induced vascular endothelial hyperpermeability through modulation of the Src/PI3K/Akt pathway and NMIIA, which play an important role in this process.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Yuwei Han
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Yazheng Zhao
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Yanni Lv
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Yang Hu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Yisha Tan
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Xueyuan Bi
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Boyang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| | - Junping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
248
|
Kämpfer AAM, Urbán P, Gioria S, Kanase N, Stone V, Kinsner-Ovaskainen A. Development of an in vitro co-culture model to mimic the human intestine in healthy and diseased state. Toxicol In Vitro 2017; 45:31-43. [PMID: 28807632 PMCID: PMC5744654 DOI: 10.1016/j.tiv.2017.08.011] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022]
Abstract
The intestine forms the largest interface between the environment and the human organism. Its integrity and functioning are crucial for the uptake of nutrients while preventing access of harmful antigens. Inflammatory conditions can significantly change the normal functioning of the intestine. In vitro models that adequately reproduce both healthy and inflamed intestinal tissue could provide a useful tool for studying the mechanisms of intestinal inflammation and investigating new therapeutic drugs. We established a co-culture of Caco-2 and PMA-differentiated THP-1 cells that mimics the intestine in healthy and controlled inflamed states. In homoeostatic conditions without stimulation, Caco-2 and THP-1 cells were co-cultured for 48 h without affecting the barrier integrity and with no increase in the release of cytokines, nitric oxide or lactate dehydrogenase. To simulate the inflamed intestine, the Caco-2 barrier was primed with IFN-γ and THP-1 cells were pre-stimulated with LPS and IFN-γ. In these conditions a significant but temporary reduction in barrier integrity was measured, and large concentrations of pro-inflammatory cytokines and cytotoxicity markers detected. With its ability to feature numerous hallmarks of intestinal inflammation the presented co-culture model of epithelial cells and macrophages offers a unique possibility to study exposure effects in relation to the health status of the intestine. A novel, tunable co-culture model of Caco-2 and THP-1 cells was established. The THP-1 differentiation protocol is crucial for a stable co-culture with Caco-2. Synergistic effects of TNF-α and IFN-γ were key to induce inflammation in vitro. The inflamed co-culture shows barrier disruption, cytokine release and cytotoxicity. Downregulation of inflammation is prevented by pretreatment of cells with cytokines.
Collapse
Affiliation(s)
- Angela A M Kämpfer
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027 Ispra, VA, Italy; Nano-Safety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Patricia Urbán
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027 Ispra, VA, Italy
| | - Sabrina Gioria
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027 Ispra, VA, Italy
| | - Nilesh Kanase
- Nano-Safety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Vicki Stone
- Nano-Safety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Agnieszka Kinsner-Ovaskainen
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027 Ispra, VA, Italy.
| |
Collapse
|
249
|
Lahey KA, Ronaghan NJ, Shang J, Dion SP, Désilets A, Leduc R, MacNaughton WK. Signaling pathways induced by serine proteases to increase intestinal epithelial barrier function. PLoS One 2017; 12:e0180259. [PMID: 28671992 PMCID: PMC5495298 DOI: 10.1371/journal.pone.0180259] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/13/2017] [Indexed: 12/30/2022] Open
Abstract
Changes in barrier function of the gastrointestinal tract are thought to contribute to the inflammatory bowel diseases Crohn's disease and ulcerative colitis. Previous work in our lab demonstrated that apical exposure of intestinal epithelial cell lines to serine proteases results in an increase in transepithelial electrical resistance (TER). However, the underlying mechanisms governing this response are unclear. We aimed to determine the requirement for proteolytic activity, epidermal growth factor receptor (EGFR) activation, and downstream intracellular signaling in initiating and maintaining enhanced barrier function following protease treatment using a canine intestinal epithelial cell line (SCBN). We also examined the role of phosphorylation of myosin regulatory light chain on the serine protease-induced increase in TER through. It was found that proteolytic activity of the serine proteases trypsin and matriptase is required to initiate and maintain the protease-mediated increase in TER. We also show that MMP-independent EGFR activation is essential to the sustained phase of the protease response, and that Src kinases may mediate EGFR transactivation. PI3-K and ERK1/2 signaling were important in reaching a maximal increase in TER following protease stimulation; however, their upstream activators are yet to be determined. CK2 inhibition prevented the increase in TER induced by serine proteases. The bradykinin B(2) receptor was not involved in the change in TER in response to serine proteases, and no change in phosphorylation of MLC was observed after trypsin or matriptase treatment. Taken together, our data show a requirement for ongoing proteolytic activity, EGFR transactivation, as well as downstream PI3-K, ERK1/2, and CK2 signaling in protease-mediated barrier enhancement of intestinal epithelial cells. The pathways mediating enhanced barrier function by proteases may be novel therapeutic targets for intestinal disorders characterized by disrupted epithelial barrier function.
Collapse
Affiliation(s)
- Kelcie A. Lahey
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Natalie J. Ronaghan
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Judie Shang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Sébastien P. Dion
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Leduc
- Département de Pharmacologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Wallace K. MacNaughton
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
250
|
Complementary Roles of Nod2 in Hematopoietic and Nonhematopoietic Cells in Preventing Gut Barrier Dysfunction Dependent on MLCK Activity. Inflamm Bowel Dis 2017; 23:1109-1119. [PMID: 28520587 DOI: 10.1097/mib.0000000000001135] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Crohn's disease (CD) pathogenesis is multifactorial involving genetic and environmental factors. Loss of function mutations in the nucleotide oligomerization domain 2 (NOD2) gene are the main genetic risk factor for CD. Like patients with CD, Nod2 mice are characterized by an enhanced Th1 immune response and a defective mucosal barrier function evidenced by increased intestinal permeability. We previously showed that the latter is related to hematopoietic Nod2 deficiency. Our aim was to explore the mechanisms by which Nod2 expressed in the hematopoietic and in the nonhematopoietic compartments interplay to control epithelial paracellular permeability. METHODS Depletion of CD4 T cells in Nod2 mice and treatments with inhibitors were conducted in chimeric mice transplanted with bone marrow cells from Nod2-deficient donors into Nod2-sufficient recipients or vice versa. Caco-2 cells overexpressing a NOD2 gene which did or did not include a CD-associated polymorphism were treated with inhibitors or siRNAs and cocultured with hematopoietic cells from Peyer's patches. RESULTS In vivo and in vitro Nod2 in hematopoietic cells regulates epithelial paracellular permeability through cytokine production influencing myosin light chain kinase (MLCK) activity. Indeed, tumor necrosis factor-α and interferon-γ secretion by CD4 T cells upregulated expression and activity of epithelial MLCK leading to increased epithelial tight junction opening. When stimulated by muramyl dipeptide, Nod2 in the nonhematopoietic compartment normalized the permeability and T-cell cytokine secretion and regulated MLCK activity. This MLCK regulation is mediated by TAK1 and RICK-dependent mechanisms. CONCLUSIONS Our study demonstrates how hematopoietic and nonhematopoietic Nod2 regulate intestinal barrier function, improving our knowledge on the mechanisms involved in CD pathogenesis.
Collapse
|