1
|
Nakano H, Setoguchi S, Kawano K, Miyagawa H, Sakao K, Hou DX. Effects of Amazake Produced with Different Aspergillus on Gut Barrier and Microbiota. Foods 2023; 12:2568. [PMID: 37444313 DOI: 10.3390/foods12132568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/18/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract. To explore the preventive effects of dietary foods on IBD, we evaluated the effects of the traditional Japanese fermented beverage "Amazake" on gut barrier function in this study. Black koji Amazake (BA) derived from Aspergillus luchuensis MEM-C strain and yellow koji Amazake (YA) derived from Aspergillus oryzae were made in this study, and their nutrients were analyzed. Mice with mild gut barrier dysfunction induced by Western diet were administered with 10% of each Amazake for two months. Mice gut microbiota were analyzed by 16S rRNA gene sequencing. BA contained a higher amount of isomaltooligosaccharides, citric acid, and ferulic acid than YA. The animal data revealed that BA significantly induced the expressions of antioxidant factors and enzymes such as NF-E2-related factor 2 (Nfr2), heme oxygenase 1 (HO1), and superoxide dismutase-2 (SOD-2). The gut barrier protein, occludin, and fecal immunoglobulin A (IgA) were also significantly enhanced by BA. Furthermore, the levels of serum endotoxin and hepatic monocyte chemotactic protein-1 (MCP-1) were decreased in both the BA and YA groups. In gut microbiota, Lachnospiraceae was increased by BA while Akkermansia muciniphilia was increased by YA. Black koji Amazake contained a higher amount of isomaltooligosaccharides, citric acid, and ferulic acid than yellow koji Amazake and contributed to protecting gut barrier function to reduce endotoxin intrusion and inflammation.
Collapse
Affiliation(s)
- Hironobu Nakano
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Sho Setoguchi
- Kirishima Shuzo Co., Ltd., 4-28-1 Shimokawahigashi, Miyazaki 885-8588, Japan
| | - Kuniaki Kawano
- Kirishima Shuzo Co., Ltd., 4-28-1 Shimokawahigashi, Miyazaki 885-8588, Japan
| | - Hiroshi Miyagawa
- Kirishima Shuzo Co., Ltd., 4-28-1 Shimokawahigashi, Miyazaki 885-8588, Japan
| | - Kozue Sakao
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
- Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - De-Xing Hou
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
- Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
2
|
Shutova MS, Borowczyk J, Russo B, Sellami S, Drukala J, Wolnicki M, Brembilla NC, Kaya G, Ivanov AI, Boehncke WH. Inflammation modulates intercellular adhesion and mechanotransduction in human epidermis via ROCK2. iScience 2023; 26:106195. [PMID: 36890793 PMCID: PMC9986521 DOI: 10.1016/j.isci.2023.106195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Aberrant mechanotransduction and compromised epithelial barrier function are associated with numerous human pathologies including inflammatory skin disorders. However, the cytoskeletal mechanisms regulating inflammatory responses in the epidermis are not well understood. Here we addressed this question by inducing a psoriatic phenotype in human keratinocytes and reconstructed human epidermis using a cytokine stimulation model. We show that the inflammation upregulates the Rho-myosin II pathway and destabilizes adherens junctions (AJs) promoting YAP nuclear entry. The integrity of cell-cell adhesion but not the myosin II contractility per se is the determinative factor for the YAP regulation in epidermal keratinocytes. The inflammation-induced disruption of AJs, increased paracellular permeability, and YAP nuclear translocation are regulated by ROCK2, independently from myosin II activation. Using a specific inhibitor KD025, we show that ROCK2 executes its effects via cytoskeletal and transcription-dependent mechanisms to shape the inflammatory response in the epidermis.
Collapse
Affiliation(s)
- Maria S. Shutova
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
- University Hospitals of Geneva, Division of Dermatology and Venereology, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Julia Borowczyk
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Barbara Russo
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
- University Hospitals of Geneva, Division of Dermatology and Venereology, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sihem Sellami
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Justyna Drukala
- Jagiellonian University, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Cracow, Poland
| | - Michal Wolnicki
- Department of Pediatric Urology, Jagiellonian University Medical College, Cracow, Poland
| | - Nicolo C. Brembilla
- University Hospitals of Geneva, Division of Dermatology and Venereology, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gurkan Kaya
- University Hospitals of Geneva, Division of Dermatology and Venereology, Geneva, Switzerland
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Wolf-Henning Boehncke
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
- University Hospitals of Geneva, Division of Dermatology and Venereology, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Chitosan Oligosaccharide Attenuates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction through Suppressing the Inflammatory Response and Oxidative Stress in Mice. Antioxidants (Basel) 2022; 11:antiox11071384. [PMID: 35883875 PMCID: PMC9312058 DOI: 10.3390/antiox11071384] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
This study was conducted to investigate the protective effect of chitosan oligosaccharide (COS) against lipopolysaccharide (LPS)-induced intestinal injury. The results demonstrated that COS improved the mucosal morphology of the jejunum and colon in LPS-challenged mice. COS alleviated the LPS-induced down-regulation of tight junction protein expressions and reduction of goblet cells number and mucin expression. The mRNA expressions of anti-microbial peptides secreted by the intestinal cells were also up-regulated by COS. Additionally, COS decreased pro-inflammatory cytokine production and neutrophil recruitment in the jejunum and colon of LPS-treated mice. COS ameliorated intestinal oxidative stress through up-regulating the mRNA expressions of nuclear factor E2-related factor 2 and downstream antioxidant enzymes genes. Correlation analysis indicated that the beneficial effects of COS on intestinal barrier function were associated with its anti-inflammatory activities and antioxidant capacity. Our study provides evidence for the application of COS to the prevention of intestinal barrier dysfunction caused by the stress of a LPS challenge.
Collapse
|
4
|
Shutova MS, Boehncke WH. Mechanotransduction in Skin Inflammation. Cells 2022; 11:2026. [PMID: 35805110 PMCID: PMC9265324 DOI: 10.3390/cells11132026] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
In the process of mechanotransduction, the cells in the body perceive and interpret mechanical stimuli to maintain tissue homeostasis and respond to the environmental changes. Increasing evidence points towards dysregulated mechanotransduction as a pathologically relevant factor in human diseases, including inflammatory conditions. Skin is the organ that constantly undergoes considerable mechanical stresses, and the ability of mechanical factors to provoke inflammatory processes in the skin has long been known, with the Koebner phenomenon being an example. However, the molecular mechanisms and key factors linking mechanotransduction and cutaneous inflammation remain understudied. In this review, we outline the key players in the tissue's mechanical homeostasis, the available data, and the gaps in our current understanding of their aberrant regulation in chronic cutaneous inflammation. We mainly focus on psoriasis as one of the most studied skin inflammatory diseases; we also discuss mechanotransduction in the context of skin fibrosis as a result of chronic inflammation. Even though the role of mechanotransduction in inflammation of the simple epithelia of internal organs is being actively studied, we conclude that the mechanoregulation in the stratified epidermis of the skin requires more attention in future translational research.
Collapse
Affiliation(s)
- Maria S. Shutova
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| |
Collapse
|
5
|
Yang HY, Liang ZH, Xie JL, Wu Q, Qin YY, Zhang SY, Tang GD. Gelsolin impairs barrier function in pancreatic ductal epithelial cells by actin filament depolymerization in hypertriglyceridemia‑induced pancreatitis in vitro. Exp Ther Med 2022; 23:290. [PMID: 35317441 PMCID: PMC8908475 DOI: 10.3892/etm.2022.11219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022] Open
Abstract
Gelsolin (GSN) is a calcium-regulated actin-binding protein that can sever actin filaments. Notably, actin dynamics affect the structure and function of epithelial barriers. The present study investigated the role of GSN in the barrier function of pancreatic ductal epithelial cells (PDECs) in hypertriglyceridemia-induced pancreatitis (HTGP). The human PDEC cell line HPDE6-C7 underwent GSN knockdown and was treated with caerulein (CAE) + triglycerides (TG). Intracellular calcium levels and the actin filament network were analyzed under a fluorescence microscope. The expression levels of GSN, E-cadherin, nectin-2, ZO-1 and occludin were evaluated by reverse transcription-quantitative polymerase chain reaction and western blotting. Ultrastructural changes in tight junctions were observed by transmission electron microscopy. Furthermore, the permeability of PDECs was analyzed by fluorescein isothiocyanate-dextran fluorescence. The results revealed that CAE + TG increased intracellular calcium levels, actin filament depolymerization and GSN expression, and increased PDEC permeability by decreasing the expression levels of E-cadherin, nectin-2, ZO-1 and occludin compared with the control. Moreover, changes in these markers, with the exception of intracellular calcium levels, were reversed by silencing GSN. In conclusion, GSN may disrupt barrier function in PDECs by causing actin filament depolymerization in HTGP in vitro.
Collapse
Affiliation(s)
- Hui-Ying Yang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhi-Hai Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jin-Lian Xie
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qing Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Ying-Ying Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shi-Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guo-Du Tang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
6
|
The ACE2 Receptor for Coronavirus Entry Is Localized at Apical Cell—Cell Junctions of Epithelial Cells. Cells 2022; 11:cells11040627. [PMID: 35203278 PMCID: PMC8870730 DOI: 10.3390/cells11040627] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Transmembrane proteins of adherens and tight junctions are known targets for viruses and bacterial toxins. The coronavirus receptor ACE2 has been localized at the apical surface of epithelial cells, but it is not clear whether ACE2 is localized at apical Cell—Cell junctions and whether it associates with junctional proteins. Here we explored the expression and localization of ACE2 and its association with transmembrane and tight junction proteins in epithelial tissues and cultured cells by data mining, immunoblotting, immunofluorescence microscopy, and co-immunoprecipitation experiments. ACE2 mRNA is abundant in epithelial tissues, where its expression correlates with the expression of the tight junction proteins cingulin and occludin. In cultured epithelial cells ACE2 mRNA is upregulated upon differentiation and ACE2 protein is widely expressed and co-immunoprecipitates with the transmembrane proteins ADAM17 and CD9. We show by immunofluorescence microscopy that ACE2 colocalizes with ADAM17 and CD9 and the tight junction protein cingulin at apical junctions of intestinal (Caco-2), mammary (Eph4) and kidney (mCCD) epithelial cells. These observations identify ACE2, ADAM17 and CD9 as new epithelial junctional transmembrane proteins and suggest that the cytokine-enhanced endocytic internalization of junction-associated protein complexes comprising ACE2 may promote coronavirus entry.
Collapse
|
7
|
The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins (Basel) 2020; 12:toxins12100619. [PMID: 32998222 PMCID: PMC7600953 DOI: 10.3390/toxins12100619] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Mycotoxins are fungal metabolites that occur in human foods and animal feeds, potentially threatening human and animal health. The intestine is considered as the first barrier against these external contaminants, and it consists of interconnected physical, chemical, immunological, and microbial barriers. In this context, based on in vitro, ex vivo, and in vivo models, we summarize the literature for compromised intestinal barrier issues caused by various mycotoxins, and we reviewed events related to disrupted intestinal integrity (physical barrier), thinned mucus layer (chemical barrier), imbalanced inflammatory factors (immunological barrier), and dysfunctional bacterial homeostasis (microbial barrier). We also provide important information on deoxynivalenol, a leading mycotoxin implicated in intestinal dysfunction, and other adverse intestinal effects induced by other mycotoxins, including aflatoxins and ochratoxin A. In addition, intestinal perturbations caused by mycotoxins may also contribute to the development of mycotoxicosis, including human chronic intestinal inflammatory diseases. Therefore, we provide a clear understanding of compromised intestinal barrier induced by mycotoxins, with a view to potentially develop innovative strategies to prevent and treat mycotoxicosis. In addition, because of increased combinatorial interactions between mycotoxins, we explore the interactive effects of multiple mycotoxins in this review.
Collapse
|
8
|
Cao X, Sun L, Lechuga S, Naydenov NG, Feygin A, Ivanov AI. A Novel Pharmacological Approach to Enhance the Integrity and Accelerate Restitution of the Intestinal Epithelial Barrier. Inflamm Bowel Dis 2020; 26:1340-1352. [PMID: 32266946 PMCID: PMC7441106 DOI: 10.1093/ibd/izaa063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Disruption of the gut barrier is an essential mechanism of inflammatory bowel diseases (IBDs) contributing to the development of mucosal inflammation. A hallmark of barrier disruption is the disassembly of epithelial adherens junctions (AJs) driven by decreased expression of a major AJ protein, E-cadherin. A group of isoxazole compounds, such as E-cadherin-upregulator (ECU) and ML327, were previously shown to stimulate E-cadherin expression in poorly differentiated human cancer cells. This study was designed to examine whether these isoxazole compounds can enhance and protect model intestinal epithelial barriers in vitro. METHODS The study was conducted using T84, SK-CO15, and HT-29 human colonic epithelial cell monolayers. Disruption of the epithelial barrier was induced by pro-inflammatory cytokines, tumor necrosis factor-α, and interferon-γ. Barrier integrity and epithelial junction assembly was examined using different permeability assays, immunofluorescence labeling, and confocal microscopy. Epithelial restitution was analyzed using a scratch wound healing assay. RESULTS E-cadherin-upregulator and ML327 treatment of intestinal epithelial cell monolayers resulted in several barrier-protective effects, including reduced steady-state epithelial permeability, inhibition of cytokine-induced barrier disruption and junction disassembly, and acceleration of epithelial wound healing. Surprisingly, these effects were not due to upregulation of E-cadherin expression but were mediated by multiple mechanisms including inhibition of junction protein endocytosis, attenuation of cytokine-induced apoptosis, and activation of promigratory Src and AKT signaling. CONCLUSIONS Our data highlight ECU and ML327 as promising compounds for developing new therapeutic strategies to protect the integrity and accelerate the restitution of the intestinal epithelial barrier in IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Xuelei Cao
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH
| | - Lei Sun
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH
| | - Nayden G Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH
| | - Alex Feygin
- School of Nursing, Virginia Commonwealth University, Richmond, VA
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH,Address correspondence to: Andrei I. Ivanov, PhD, Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC22, Cleveland, OH 44195, USA. E-mail:
| |
Collapse
|
9
|
Swain SD, Grifka-Walk HN, Gripentrog J, Lehmann M, Deuling B, Jenkins B, Liss H, Blaseg N, Bimczok D, Kominsky DJ. Slug and Snail have differential effects in directing colonic epithelial wound healing and partially mediate the restitutive effects of butyrate. Am J Physiol Gastrointest Liver Physiol 2019; 317:G531-G544. [PMID: 31393789 PMCID: PMC6842986 DOI: 10.1152/ajpgi.00071.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Restitution of wounds in colonic epithelium is essential in the maintenance of health. Microbial products, such as the short-chain fatty acid butyrate, can have positive effects on wound healing. We used an in vitro model of T84 colonic epithelial cells to determine if the Snail genes Slug (SNAI2) and Snail (SNAI1), implemented in keratinocyte monolayer healing, are involved in butyrate-enhanced colonic epithelial wound healing. Using shRNA-mediated Slug/Snail knockdown, we found that knockdown of Slug (Slug-KD), but not Snail (Snail-KD), impairs wound healing in scratch assays with and without butyrate. Slug and Snail had differential effects on T84 monolayer barrier integrity, measured by transepithelial resistance, as Snail-KD impaired the barrier (with or without butyrate), whereas Slug-KD enhanced the barrier, again with or without butyrate. Targeted transcriptional analysis demonstrated differential expression of several tight junction genes, as well as focal adhesion genes. This included altered regulation of Annexin A2 and ITGB1 in Slug-KD, which was reflected in confocal microscopy, showing increased accumulation of B1-integrin protein in Slug-KD cells, which was previously shown to impair wound healing. Transcriptional analysis also indicated altered expression of genes associated with epithelial terminal differentiation, such that Slug-KD cells skewed toward overexpression of secretory cell pathway-associated genes. This included trefoil factors TFF1 and TFF3, which were expressed at lower than control levels in Snail-KD cells. Since TFFs can enhance the barrier in epithelial cells, this points to a potential mechanism of differential modulation by Snail genes. Although Snail genes are crucial in epithelial wound restitution, butyrate responses are mediated by other pathways as well.NEW & NOTEWORTHY Although butyrate can promote colonic mucosal healing, not all of its downstream pathways are understood. We show that the Snail genes Snail and Slug are mediators of butyrate responses. Furthermore, these genes, and Slug in particular, are necessary for efficient restitution of wounds and barriers in T84 epithelial cells even in the absence of butyrate. These effects are achieved in part through effects on regulation of β1 integrin and cellular differentiation state.
Collapse
Affiliation(s)
- Steve D. Swain
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | | | - Jeannie Gripentrog
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Margaret Lehmann
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Benjamin Deuling
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Brittany Jenkins
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Hailey Liss
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Nathan Blaseg
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Diane Bimczok
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Douglas J. Kominsky
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| |
Collapse
|
10
|
Nunes C, Freitas V, Almeida L, Laranjinha J. Red wine extract preserves tight junctions in intestinal epithelial cells under inflammatory conditions: implications for intestinal inflammation. Food Funct 2019; 10:1364-1374. [PMID: 30735221 DOI: 10.1039/c8fo02469c] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The altered expression and subcellular distribution of tight junction (TJ) proteins, leading to a dysfunctional intestinal barrier, is a key mechanistic feature of inflammatory bowel disease (IBD). Therefore, increasing the integrity of the intestinal barrier by manipulating the TJ may constitute an innovative and effective therapeutic strategy in IBD. In this context, recent studies showed that dietary polyphenols are able to protect the intestinal TJ barrier integrity. Here, using a cellular model of intestinal inflammation, consisting of cytokine-stimulated HT-29 colon epithelial cells, we show that a polyphenolic extract obtained from Portuguese red wine (RWE) decreased the paracellular permeability across the cell monolayer compared with the control cells, even in the presence of pro-inflammatory cytokines. The beneficial effect of RWE was exerted at three complementary levels: (1) by promoting a significant increase of the mRNA of key barrier-forming TJ proteins, including occludin, claudin-5 and zonnula occludens (ZO)-1 above the levels observed in the control cells; (2) by preventing the decrease in the expression of these proteins under inflammatory conditions and (3) by averting the increase in claudin-2 mRNA, a channel-forming TJ protein induced by pro-inflammatory cytokines. Taken together, these results strongly suggest that polyphenols presented and consumed in red wine as a mixture can reinforce and protect the intestinal barrier against inflammatory stimulus by affecting the TJ protein expression and, thus, without the need for purifying individual compounds, might represent a readily available therapeutic intervention against IBD and intestinal inflammation.
Collapse
Affiliation(s)
- Carla Nunes
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | | | | | | |
Collapse
|
11
|
Li F, Pascal LE, Stolz DB, Wang K, Zhou Y, Chen W, Xu Y, Chen Y, Dhir R, Parwani AV, Nelson JB, DeFranco DB, Yoshimura N, Balasubramani GK, Gingrich JR, Maranchie JK, Jacobs BL, Davies BJ, Hrebinko RL, Bigley JD, McBride D, Guo P, He D, Wang Z. E-cadherin is downregulated in benign prostatic hyperplasia and required for tight junction formation and permeability barrier in the prostatic epithelial cell monolayer. Prostate 2019; 79:1226-1237. [PMID: 31212363 PMCID: PMC6599563 DOI: 10.1002/pros.23806] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND We previously reported the presence of prostate-specific antigen (PSA) in the stromal compartment of benign prostatic hyperplasia (BPH). Since PSA is expressed exclusively by prostatic luminal epithelial cells, PSA in the BPH stroma suggests increased tissue permeability and the compromise of epithelial barrier integrity. E-cadherin, an important adherens junction component and tight junction regulator, is known to exhibit downregulation in BPH. These observations suggest that the prostate epithelial barrier is disrupted in BPH and E-cadherin downregulation may increase epithelial barrier permeability. METHODS The ultra-structure of cellular junctions in BPH specimens was observed using transmission electron microscopy (TEM) and E-cadherin immunostaining analysis was performed on BPH and normal adjacent specimens from BPH patients. In vitro cell line studies using benign prostatic epithelial cell lines were performed to determine the impact of small interfering RNA knockdown of E-cadherin on transepithelial electrical resistance and diffusion of fluorescein isothiocyanate (FITC)-dextran in transwell assays. RESULTS The number of kiss points in tight junctions was reduced in BPH epithelial cells as compared with the normal adjacent prostate. Immunostaining confirmed E-cadherin downregulation and revealed a discontinuous E-cadherin staining pattern in BPH specimens. E-cadherin knockdown increased monolayer permeability and disrupted tight junction formation without affecting cell density. CONCLUSIONS Our results indicate that tight junctions are compromised in BPH and loss of E-cadherin is potentially an important underlying mechanism, suggesting targeting E-cadherin loss could be a potential approach to prevent or treat BPH.
Collapse
Affiliation(s)
- Feng Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Laura E Pascal
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yibin Zhou
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Chen
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yadong Xu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Urology, The Second Affiliated Hospital of Centre West University, Changsha, Hunan, China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rajiv Dhir
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joel B Nelson
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Donald B DeFranco
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Goundappa K Balasubramani
- Department of Epidemiology, Epidemiology Data Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jeffrey R Gingrich
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jodi K Maranchie
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Bruce L Jacobs
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Benjamin J Davies
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ronald L Hrebinko
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joel D Bigley
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dawn McBride
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhou Wang
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Zhao G, Zhuo YZ, Cui LH, Li CX, Chen SY, Li D, Liu JH, Li DH, Cui NQ, Zhang SK. Modified Da-chai-hu Decoction regulates the expression of occludin and NF-κB to alleviate organ injury in severe acute pancreatitis rats. Chin J Nat Med 2019; 17:355-362. [PMID: 31171270 DOI: 10.1016/s1875-5364(19)30041-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Modified Da-chai-hu Decoction (MDD), a traditional Chinese medicinal formulation, which was empirically generated from Da-chai-hu decoction, has been utilized to treat severe acute pancreatitis (SAP) for decades. The aim of the present study was to explore its potential organprotective mechanism in SAP. In the present study, rat SAP model was induced by retrograde injection of 3.5% sodium taurocholate into the biliopancreatic duct, MDD (23.35 g/kg body weight, twelve times the clinical dose) were orally given at 2 h before and 10 h after injection. At 12 h after model induction, blood was taken from vena cava for analysis of amylase, diamine oxidase (DAO), pulmonary surfactant protein-A (SP-A), and C-reactive protein (CRP). Histopathological change of pancreas, ileum and lung was assayed by H&E staining, myeloperoxidase (MPO) activity were determinated using colorimetric assay, and the expressions of occludin and nuclear factor-κB (NF-κB) were detected by real-time RT-PCR and western blot, respectively. In addition, the tissue concentrations of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and monocyte chemoattractant protein-1 (MCP-1) were measured by enzyme-linked immunosorbent assay (ELISA). The results showed that in SAP rats, MDD significantly alleviated histopathological damage, depressed the MPO activity and the concentrations of TNF-α, IL-1β, and MCP-1 of pancreas, ileum and lung, and reduced the serum levels of amylase [(3283.4 ± 585.5) U·L-1vs (5626.4 ± 795.1)U·L-1], DAO [(1100.1 ± 334.3) U·L-1vs (1666.4 ± 525.3) U·L-1] and CRP [(7.6 ± 1.2) μg·mL-1vs (17.8 ± 3.8) μg·mL-1]. However, the serum SP-A concentration [(106.1 ± 16.6) pg·mL-1vs (90.1 ± 14.9) pg·mL-1] was elevated when treated SAP rats with MDD. Furthermore, MDD increased the occludin expression and reduced the NF-κB expression in pancreas, ileum and lung of SAP rats. Our findings suggested that MDD administration was an effective therapeutic approach for SAP treatment. It could up-regulate occludin expression to protect intercellular tight junction and down-regulate NF-κB expression to inhibit inflammatory reaction of pancreas, ileum and lung.
Collapse
Affiliation(s)
- Guang Zhao
- Hepatobiliary and Pancreatic Surgery, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China; Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China
| | - Yu-Zhen Zhuo
- Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China; Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Li-Hua Cui
- Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China; Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Cai-Xia Li
- Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China; Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Sha-Yan Chen
- Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China; Department of Clinical Laboratory, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Dan Li
- Hepatobiliary and Pancreatic Surgery, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China; Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China
| | - Jun-Hong Liu
- Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Di-Hua Li
- Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China
| | - Nai-Qiang Cui
- Hepatobiliary and Pancreatic Surgery, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China; Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China
| | - Shu-Kun Zhang
- Nankai Clinical College, Tianjin Medical University, Tianjin 300107, China; Institute of Integrated Traditional Chinese and Western medicine, Tianjin Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western medicine, Tianjin 300100, China.
| |
Collapse
|
13
|
Popović N, Djokić J, Brdarić E, Dinić M, Terzić-Vidojević A, Golić N, Veljović K. The Influence of Heat-Killed Enterococcus faecium BGPAS1-3 on the Tight Junction Protein Expression and Immune Function in Differentiated Caco-2 Cells Infected With Listeria monocytogenes ATCC 19111. Front Microbiol 2019; 10:412. [PMID: 30891021 PMCID: PMC6411766 DOI: 10.3389/fmicb.2019.00412] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Listeria monocytogenes, the common foodborne pathogenic bacteria species, compromises the intestinal epithelial barrier, leading to development of the listeriosis, a severe disease especially among immunocompromised individuals. L. monocytogenes infection usually requires antibiotic treatment, however, excessive use of antibiotics promotes emergence of antibiotic resistance and the destruction of gut microbiota. Probiotics, including lactic acid bacteria (LAB), have been repeatedly proven as an alternative approach for the treatment of various infections. We have analyzed the potential of Enterococcus faecium BGPAS1-3, a dairy isolate exhibiting strong direct antilisterial effect, to modulate the response of differentiated Caco-2 intestinal epithelial cells to L. monocytogenes ATCC 19111 infection. We showed that the molecule with antilisterial effect is a bacterial cell-wall protein that is highly resistant to the high-temperature treatment. When we tested the antilisterial potential of heat-killed BGPAS1-3, we found that it could prevent tight junction disruption in differentiated Caco-2 monolayer infected with L. monocytogenes ATCC 19111, induce antilisterial host response mechanisms, and stimulate the production of protective TGF-β in intestinal epithelial cells. We also showed that the modulation of MyD88 dependent TLR2 and TLR4 pathways by BGPAS1-3 are involved in host response against L. monocytogenes ATCC 19111. Since heat-killed BGPAS1-3 possess strong antilisterial effects, such postbiotic could be used as a controllable and safe therapeutic.
Collapse
Affiliation(s)
| | - Jelena Djokić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | |
Collapse
|
14
|
Lechuga S, Ivanov AI. Disruption of the epithelial barrier during intestinal inflammation: Quest for new molecules and mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1183-1194. [PMID: 28322932 DOI: 10.1016/j.bbamcr.2017.03.007] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium forms a key protective barrier that separates internal organs from the harmful environment of the gut lumen. Increased permeability of the gut barrier is a common manifestation of different inflammatory disorders contributing to the severity of disease. Barrier permeability is controlled by epithelial adherens junctions and tight junctions. Junctional assembly and integrity depend on fundamental homeostatic processes such as cell differentiation, rearrangements of the cytoskeleton, and vesicle trafficking. Alterations of intestinal epithelial homeostasis during mucosal inflammation may impair structure and remodeling of apical junctions, resulting in increased permeability of the gut barrier. In this review, we summarize recent advances in our understanding of how altered epithelial homeostasis affects the structure and function of adherens junctions and tight junctions in the inflamed gut. Specifically, we focus on the transcription reprogramming of the cell, alterations in the actin cytoskeleton, and junctional endocytosis and exocytosis. We pay special attention to knockout mouse model studies and discuss the relevance of these mechanisms to human gastrointestinal disorders.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA; Virginia Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
15
|
Maidji E, Somsouk M, Rivera JM, Hunt PW, Stoddart CA. Replication of CMV in the gut of HIV-infected individuals and epithelial barrier dysfunction. PLoS Pathog 2017; 13:e1006202. [PMID: 28241080 PMCID: PMC5328284 DOI: 10.1371/journal.ppat.1006202] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/26/2017] [Indexed: 01/25/2023] Open
Abstract
Although invasive cytomegalovirus (CMV) disease is uncommon in the era of antiretroviral therapy (ART), asymptomatic CMV coinfection is nearly ubiquitous in HIV infected individuals. While microbial translocation and gut epithelial barrier dysfunction may promote persistent immune activation in treated HIV infection, potentially contributing to morbidity and mortality, it has been unclear whether CMV replication in individuals with no symptoms of CMV disease might play a role in this process. We hypothesized that persistent CMV replication in the intestinal epithelium of HIV/CMV-coinfected individuals impairs gut epithelial barrier function. Using a combination of state-of-the-art in situ hybridization technology (RNAscope) and immunohistochemistry, we detected CMV DNA and proteins and evidence of intestinal damage in rectosigmoid samples from CMV-positive individuals with both untreated and ART-suppressed HIV infection. Two different model systems, primary human intestinal cells differentiated in vitro to form polarized monolayers and a humanized mouse model of human gut, together demonstrated that intestinal epithelial cells are fully permissive to CMV replication. Independent of HIV, CMV disrupted tight junctions of polarized intestinal cells, significantly reducing transepithelial electrical resistance, a measure of monolayer integrity, and enhancing transepithelial permeability. The effect of CMV infection on the intestinal epithelium is mediated, at least in part, by the CMV-induced proinflammatory cytokine IL-6. Furthermore, letermovir, a novel anti-CMV drug, dampened the effects of CMV on the epithelium. Together, our data strongly suggest that CMV can disrupt epithelial junctions, leading to bacterial translocation and chronic inflammation in the gut and that CMV could serve as a target for therapeutic intervention to prevent or treat gut epithelial barrier dysfunction during HIV infection.
Collapse
Affiliation(s)
- Ekaterina Maidji
- Division of Experimental Medicine, Department of Medicine, Zuckerberg San Francisco General, University of California, San Francisco, San Francisco, California, United States of America
| | - Ma Somsouk
- Division of Gastroenterology, Department of Medicine, Zuckerberg San Francisco General, University of California, San Francisco, San Francisco, California, United States of America
| | - Jose M. Rivera
- Division of Experimental Medicine, Department of Medicine, Zuckerberg San Francisco General, University of California, San Francisco, San Francisco, California, United States of America
| | - Peter W. Hunt
- Division of Experimental Medicine, Department of Medicine, Zuckerberg San Francisco General, University of California, San Francisco, San Francisco, California, United States of America
| | - Cheryl A. Stoddart
- Division of Experimental Medicine, Department of Medicine, Zuckerberg San Francisco General, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
16
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 354] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
17
|
Protein Kinase D2 Protects against Acute Colitis Induced by Dextran Sulfate Sodium in Mice. Sci Rep 2016; 6:34079. [PMID: 27659202 PMCID: PMC5034322 DOI: 10.1038/srep34079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease is characterized by dysregulation of the mucosal immune system resulting from impaired intestinal epithelial barrier function. Protein kinase D2 has been implicated in the regulation of immune responses. The present study was to define PKD2 might affect murine colitis. Colitis was induced in wild-type mice (PKD2WT/WT) and PKD2 catalytic activity deficient mice (PKD2SSAA/SSAA) with dextran sulfate sodium. PKD2SSAA-knockin mice displayed catalytic activity deficiency and increased susceptibility to DSS-induced colitis with enhanced weight loss, colonic inflammation compared with PKD2WT/WT mice. Furthermore, crucial inflammatory cytokines mRNA levels in PKD2SSAA-knockin mice were higher than controls accompanied with down-regulation of ZO-1, MUC2 and intestinal barrier dysfunction. However, there were no differences in the proliferation or apoptosis of intestinal epithelial cells in PKD2SSAA-knockin mice compared with wild-type controls. In addition, PKD2 expression was repressed in patients with IBD compared with healthy controls. These studies suggested that activation of PKD2 in the colonic epithelium microenvironment may contribute to protect against DSS-induced colitis through regulation of intestinal mucosal immunity and barrier function.
Collapse
|
18
|
Kumar S, Chaudhary AK, Kumar R, O'Malley J, Dubrovska A, Wang X, Yadav N, Goodrich DW, Chandra D. Combination therapy induces unfolded protein response and cytoskeletal rearrangement leading to mitochondrial apoptosis in prostate cancer. Mol Oncol 2016; 10:949-65. [PMID: 27106131 DOI: 10.1016/j.molonc.2016.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/13/2016] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
Development of therapeutic resistance is responsible for most prostate cancer (PCa) related mortality. Resistance has been attributed to an acquired or selected cancer stem cell phenotype. Here we report the histone deacetylase inhibitor apicidin (APC) or ER stressor thapsigargin (TG) potentiate paclitaxel (TXL)-induced apoptosis in PCa cells and limit accumulation of cancer stem cells. TXL-induced responses were modulated in the presence of TG with increased accumulation of cells at G1-phase, rearrangement of the cytoskeleton, and changes in cytokine release. Cytoskeletal rearrangement was associated with modulation of the cytoplasmic and mitochondrial unfolded protein response leading to mitochondrial dysfunction and release of proapoptotic proteins from mitochondria. TXL in combination with APC or TG enhanced caspase activation. Importantly, TXL in combination with TG induced caspase activation and apoptosis in X-ray resistant LNCaP cells. Increased release of transforming growth factor-beta (TGF-β) was observed while phosphorylated β-catenin level was suppressed with TXL combination treatments. This was accompanied by a decrease in the CD44(+)CD133(+) cancer stem cell-like population, suggesting treatment affects cancer stem cell properties. Taken together, combination treatment with TXL and either APC or TG induces efficient apoptosis in both proliferating and cancer stem cells, suggesting this therapeutic combination may overcome drug resistance and recurrence in PCa.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse, Dresden, Germany; German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Xinjiang Wang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
19
|
Abe A, Takano K, Kojima T, Nomura K, Kakuki T, Kaneko Y, Yamamoto M, Takahashi H, Himi T. Interferon-gamma increased epithelial barrier function via upregulating claudin-7 expression in human submandibular gland duct epithelium. J Mol Histol 2016; 47:353-63. [PMID: 26956365 DOI: 10.1007/s10735-016-9667-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/03/2016] [Indexed: 02/07/2023]
Abstract
Tight junctions (TJs) are necessary for salivary gland function and may serve as indicators of salivary gland epithelial dysfunction. IgG4-related disease (IgG4-RD) is a newly recognized fibro-inflammatory condition which disrupts the TJ associated epithelial barrier. The salivary glands are one of the most frequently involved organs in IgG4-RD, however, changes of the TJ associated epithelial barrier in salivary gland duct epithelium is poorly understood. Here, we investigated the regulation and function of TJs in human submandibular gland ductal epithelial cells (HSDECs) in normal and IgG4-RD. We examined submandibular gland (SMG) tissue from eight control individuals and 22 patients with IgG4-RD and established an HSDEC culture system. Immunohistochemistry, immunocytochemistry, western blotting, and measurement of transepithelial electrical resistance (TER) were performed. Claudin-4, claudin-7, occludin, and JAM-A were expressed at the apical side of the duct epithelium in submandibular gland (SMG) tissue and at the cell borders in HSDECs of normal and IgG4-RD. The expression and distribution of TJs in SMG tissue were not different in control individuals and patients with IgG4-RD in vivo and in vitro. Although interferon-gamma (IFNγ) generally disrupts the integrity and function of TJs, as manifested by decreased epithelial barrier function, IFNγ markedly increased the epithelial barrier function of HSDECs via upregulation of claudin-7 expression in HSDECs from patients with IgG4-RD. This is the first report showing an IFNγ-dependent increase in epithelial barrier function in the salivary gland duct epithelium. Our results provide insights into the functional significance of TJs in salivary gland duct epithelium in physiological and pathological conditions, including IgG4-RD.
Collapse
Affiliation(s)
- Ayumi Abe
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan.
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuaki Nomura
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Takuya Kakuki
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yakuto Kaneko
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Motohisa Yamamoto
- Department of the Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of the Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, S1W16, Chuo-ku, Sapporo, 060-8543, Japan
| |
Collapse
|
20
|
Marinković E, Lukić I, Kosanović D, Inić-Kanada A, Gavrović-Jankulović M, Stojanović M. Recombinantly produced banana lectin isoform promotes balanced pro-inflammatory response in the colon. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
21
|
Blevins CH, Sharma AN, Johnson ML, Geno D, Gupta M, Bharucha AE, Katzka DA, Iyer PG. Influence of reflux and central obesity on intercellular space diameter of esophageal squamous epithelium. United European Gastroenterol J 2015; 4:177-83. [PMID: 27087944 DOI: 10.1177/2050640615598426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/08/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND While central obesity increases gastroesophageal reflux (GER) by mechanically disrupting the anti-reflux barrier, limited data exist on pathways by which central obesity may potentiate esophageal injury by non-mechanical means. Obesity has been associated with an impaired epithelial intestinal barrier. OBJECTIVE We aimed to assess the influence of central obesity and reflux on the squamous esophageal epithelial intercellular space diameter (ICSD). METHODS The ICSD was measured using electron microscopy in esophageal biopsies from individuals who underwent ambulatory pH monitoring and endoscopy. Anthropometric measurements were obtained on all participants. Participants were classified into four groups: with and without central obesity and reflux. RESULTS Sixteen individuals were studied with four in each study group. The mean ICSD was almost three-fold greater (p < 0.001) in the group with central obesity without reflux, compared to controls without central obesity and reflux. It was also comparable to the ICSD in groups with acid reflux only and those with both reflux and central obesity. CONCLUSIONS There is evidence of esophageal squamous ICSD increase in individuals with central obesity who do not have evidence of acid and nonacid reflux on ambulatory pH monitoring. This may reflect a mechanism by which central obesity potentiates reflux-induced esophageal injury and inflammation.
Collapse
Affiliation(s)
- Christopher H Blevins
- Department of Internal Medicine, Mayo Clinic, Rochester, USA; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Anamay N Sharma
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Michele L Johnson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Deborah Geno
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Milli Gupta
- Division of Gastroenterology, University of Calgary, Alberta, Canada
| | - Adil E Bharucha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - David A Katzka
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| |
Collapse
|
22
|
Heat-killed yogurt-containing lactic acid bacteria prevent cytokine-induced barrier disruption in human intestinal Caco-2 cells. ANN MICROBIOL 2015. [DOI: 10.1007/s13213-015-1093-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
23
|
Lechuga S, Baranwal S, Ivanov AI. Actin-interacting protein 1 controls assembly and permeability of intestinal epithelial apical junctions. Am J Physiol Gastrointest Liver Physiol 2015; 308:G745-56. [PMID: 25792565 PMCID: PMC4421013 DOI: 10.1152/ajpgi.00446.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/11/2015] [Indexed: 01/31/2023]
Abstract
Adherens junctions (AJs) and tight junctions (TJs) are crucial regulators of the integrity and restitution of the intestinal epithelial barrier. The structure and function of epithelial junctions depend on their association with the cortical actin cytoskeleton that, in polarized epithelial cells, is represented by a prominent perijunctional actomyosin belt. The assembly and stability of the perijunctional cytoskeleton is controlled by constant turnover (disassembly and reassembly) of actin filaments. Actin-interacting protein (Aip) 1 is an emerging regulator of the actin cytoskeleton, playing a critical role in filament disassembly. In this study, we examined the roles of Aip1 in regulating the structure and remodeling of AJs and TJs in human intestinal epithelium. Aip1 was enriched at apical junctions in polarized human intestinal epithelial cells and normal mouse colonic mucosa. Knockdown of Aip1 by RNA interference increased the paracellular permeability of epithelial cell monolayers, decreased recruitment of AJ/TJ proteins to steady-state intercellular contacts, and attenuated junctional reassembly in a calcium-switch model. The observed defects of AJ/TJ structure and functions were accompanied by abnormal organization and dynamics of the perijunctional F-actin cytoskeleton. Moreover, loss of Aip1 impaired the apico-basal polarity of intestinal epithelial cell monolayers and inhibited formation of polarized epithelial cysts in 3-D Matrigel. Our findings demonstrate a previously unanticipated role of Aip1 in regulating the structure and remodeling of intestinal epithelial junctions and early steps of epithelial morphogenesis.
Collapse
Affiliation(s)
- Susana Lechuga
- 1Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia;
| | - Somesh Baranwal
- 1Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia;
| | - Andrei I. Ivanov
- 1Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia; ,2Virginia Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia; ,3VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
24
|
F-actin binding protein, anillin, regulates integrity of intercellular junctions in human epithelial cells. Cell Mol Life Sci 2015; 72:3185-3200. [PMID: 25809162 DOI: 10.1007/s00018-015-1890-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 02/26/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023]
Abstract
Tight junctions (TJ) and adherens junctions (AJ) are key morphological features of differentiated epithelial cells that regulate the integrity and permeability of tissue barriers. Structure and remodeling of epithelial junctions depends on their association with the underlying actomyosin cytoskeleton. Anillin is a unique scaffolding protein interacting with different cytoskeletal components, including actin filaments and myosin motors. Its role in the regulation of mammalian epithelial junctions remains unexplored. Downregulation of anillin expression in human prostate, colonic, and lung epithelial cells triggered AJ and TJ disassembly without altering the expression of junctional proteins. This junctional disassembly was accompanied by dramatic disorganization of the perijunctional actomyosin belt; while the general architecture of the actin cytoskeleton, and activation status of non-muscle myosin II, remained unchanged. Furthermore, loss of anillin disrupted the adducin-spectrin membrane skeleton at the areas of cell-cell contact, selectively decreased γ-adducin expression, and induced cytoplasmic aggregation of αII-spectrin. Anillin knockdown activated c-Jun N-terminal kinase (JNK), and JNK inhibition restored AJ and TJ integrity and cytoskeletal organization in anillin-depleted cells. These findings suggest a novel role for anillin in regulating intercellular adhesion in model human epithelia by mechanisms involving the suppression of JNK activity and controlling the assembly of the perijunctional cytoskeleton.
Collapse
|
25
|
Ivanov AI. Tissue Barriers: Introducing an exciting new journal. Temperature (Austin) 2014; 1:151-3. [PMID: 27626042 PMCID: PMC5008708 DOI: 10.4161/23328940.2014.978716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 10/16/2014] [Accepted: 10/16/2014] [Indexed: 11/19/2022] Open
Abstract
This Editorial is written to introduce Tissue Barriers, a new Taylor & Francis journal, to the readers of Temperature. It describes the role of temperature in the regulation of different tissue barriers under normal and disease conditions. It also highlights the most interesting articles published in the first volume of Tissue Barriers.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Human and Molecular Genetics; Virginia Institute of Molecular Medicine; VCU Massey Cancer Center; Virginia Commonwealth University ; Richmond, VA USA
| |
Collapse
|
26
|
Le Dréan G, Segain JP. Connecting metabolism to intestinal barrier function: The role of leptin. Tissue Barriers 2014; 2:e970940. [PMID: 25610758 DOI: 10.4161/21688362.2014.970940] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/25/2014] [Indexed: 12/16/2022] Open
Abstract
Structure and function of the intestinal epithelial barrier (IEB) are dependent upon the integrity of junctional protein structures sealing the apical surface between epithelial cells. Tight junctions (TJ) and the surrounding apical F-actin cytoskeleton are involved in the regulation of paracellular permeability. The regulation of actin cytoskeleton organization by RhoA/Rho-kinase (ROCK) pathway plays an important role in TJ assembly and function. There is mounting evidence that the adipocyte-derived hormone leptin exerts pleiotropic effects on the intestinal epithelium including nutrient absorption, epithelial growth, inflammation and injury. Leptin activates multiple cell signaling pathways in intestinal epithelial cells (IEC) that can explain these pleiotropic effects. However, these pathways are also involved in the primary role of leptin that is the regulation of energy and glucose metabolism homeostasis. In this commentary, we examine how the interplay between leptin signaling pathways that regulate cell metabolism could impact upon IEB function.
Collapse
Key Words
- AMPK
- AMPK, AMP-activated protein kinase
- IEB, intestinal epithelial barrier
- IEC, intestinal epithelial cells
- JAK, Janus kinase
- JAK/STAT
- LepR-b, leptin receptor
- MEF, mouse embryonic fibroblast
- MLC, myosin light chain
- ROCK, Rho-kinase
- RhoA/ROCK
- STAT, signal transducer and activator of transcription
- TJ, tight junctions
- VAT, visceral adipose tissue
- barrier repair
- intestinal epithelial barrier
- leptin
- metabolism
- tight-junction
Collapse
Affiliation(s)
- Gwenola Le Dréan
- Université de Nantes; Institut des Maladies de l'Appareil Digestif (IMAD); Centre de Recherche en Nutrition Humaine du Grand Ouest (CRNH) ; Nantes, France ; CHU Hôtel-Dieu, Place Alexis Ricordeau ; Nantes, France
| | - Jean-Pierre Segain
- Université de Nantes; Institut des Maladies de l'Appareil Digestif (IMAD); Centre de Recherche en Nutrition Humaine du Grand Ouest (CRNH) ; Nantes, France ; CHU Hôtel-Dieu, Place Alexis Ricordeau ; Nantes, France
| |
Collapse
|
27
|
|