• Title/Summary/Keyword: intestinal epithelial permeability

Search Result 20, Processing Time 0.024 seconds

In Vitro Profiling of Bacterial Influence and Herbal Applications of Lonicerae Flos on the Permeability of Intestinal Epithelial Cells (장내 상피세포 점막 투과성에 대한 유산균 및 금은화의 효과)

  • Lee, Sin-Ji;Lee, Myeong-Jong;Jung, Ji-Eun;Kim, Ho-Jun;Bose, Shambhunath
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.41 no.7
    • /
    • pp.881-887
    • /
    • 2012
  • This study was designed to examine influence of bacteria and herbal applications of Lonicerae Flos on the permeability of intestinal epithelial cells. The HCT-116 human intestinal cell was used as an in vivo model of "leaky gut". Dextran sodium sulphate (DSS) was used to induce an increase in the permeability of epithelial cell tight junctions. Probiotics including Lactobacillus acidophilus, Lactobacillus plantarum, Salomonella spp. and Staphylococcus aureus were used to evaluate bacterial influence on the intestinal epithelial cells. Herbal extract Lonicerae Flos was also assessed to find out what kind of effects it has when used alone or fermented with Lactobacillus plantarum. The permeability of intestinal cell lines was assessed using transepithelial electrical resistance (TEER) and horseradish peroxide (HRP) assessments. This data provides evidence for the probiotic benefits of intestinal permeability. The data also provides promising evidence of herbal effects on reducing intestinal permeability, especially when fermented with probiotics.

Autophagy down-regulates NLRP3-dependent inflammatory response of intestinal epithelial cells under nutrient deprivation

  • Yun, Yewon;Baek, Ahruem;Kim, Dong-Eun
    • BMB Reports
    • /
    • v.54 no.5
    • /
    • pp.260-265
    • /
    • 2021
  • Dysregulation of inflammation induced by noninfectious stress conditions, such as nutrient deprivation, causes tissue damage and intestinal permeability, resulting in the development of inflammatory bowel diseases. We studied the effect of autophagy on cytokine secretion related to intestinal permeability under nutrient deprivation. Autophagy removes NLRP3 inflammasomes via ubiquitin-mediated degradation under starvation. When autophagy was inhibited, starvation-induced NLRP3 inflammasomes and their product, IL-1β, were significantly enhanced. A prolonged nutrient deprivation resulted in an increased epithelial mesenchymal transition (EMT), leading to intestinal permeability. Under nutrient deprivation, IL-17E/25, which is secreted by IL-1β, demolished the intestinal epithelial barrier. Our results suggest that an upregulation of autophagy maintains the intestinal barrier by suppressing the activation of NLRP3 inflammasomes and the release of their products, including pro-inflammatory cytokines IL-1β and IL-17E/25, under nutrient deprivation.

Contributions of HO-1-Dependent MAPK to Regulating Intestinal Barrier Disruption

  • Zhang, Zhenling;Zhang, Qiuping;Li, Fang;Xin, Yi;Duan, Zhijun
    • Biomolecules & Therapeutics
    • /
    • v.29 no.2
    • /
    • pp.175-183
    • /
    • 2021
  • The mitogen-activated protein kinase (MAPK) pathway controls intestinal epithelial barrier permeability by regulating tight junctions (TJs) and epithelial cells damage. Heme oxygenase-1 (HO-1) and carbon monoxide (CO) protect the intestinal epithelial barrier function, but the molecular mechanism is not yet clarified. MAPK activation and barrier permeability were studied using monolayers of Caco-2 cells treated with tissue necrosis factor α (TNF-α) transfected with FUGW-HO-1 or pLKO.1-sh-HO-1 plasmid. Intestinal mucosal barrier permeability and MAPK activation were also investigated using carbon tetrachloride (CCl4) administration with CoPP (a HO-1 inducer), ZnPP (a HO-1 inhibitor), CO releasing molecule 2 (CORM-2), or inactived-CORM-2-treated wild-type mice and mice with HO-1 deficiency in intestinal epithelial cells. TNF-α increased epithelial TJ disruption and cleaved caspase-3 expression, induced ERK, p38, and JNK phosphorylation. In addition, HO-1 blocked TNF-α-induced increase in epithelial TJs disruption, cleaved caspase-3 expression, as well as ERK, p38, and JNK phosphorylation in an HO-1-dependent manner. CoPP and CORM-2 directly ameliorated intestinal mucosal injury, attenuated TJ disruption and cleaved caspase-3 expression, and inhibited epithelial ERK, p38, and JNK phosphorylation after chronic CCl4 injection. Conversely, ZnPP completely reversed these effects. Furthermore, mice with intestinal epithelial HO-1 deficient exhibited a robust increase in mucosal TJs disruption, cleaved caspase-3 expression, and MAPKs activation as compared to the control group mice. These data demonstrated that HO-1-dependent MAPK signaling inhibition preserves the intestinal mucosal barrier integrity by abrogating TJ dysregulation and epithelial cell damage. The differential targeting of gut HO-1-MAPK axis leads to improved intestinal disease therapy.

Lactobacillus casei LC01 Regulates Intestinal Epithelial Permeability through miR-144 Targeting of OCLN and ZO1

  • Hou, Qiuke;Huang, Yongquan;Wang, Yan;Liao, Liu;Zhu, Zhaoyang;Zhang, Wenjie;Liu, Yongshang;Li, Peiwu;Chen, Xinlin;Liu, Fengbin
    • Journal of Microbiology and Biotechnology
    • /
    • v.30 no.10
    • /
    • pp.1480-1487
    • /
    • 2020
  • Our previous report determined that miR-144 is a key regulator of intestinal epithelial permeability in irritable bowel syndrome with diarrhea (IBS-D) rats. Recent evidence has shown that lactobacilli play an important role in the relief of IBS-D symptoms. However, few studies have addressed the mechanisms by which microRNAs and lactobacilli exert their beneficial effects on intestinal epithelial permeability. Hence, to elucidate whether miRNAs and lactobacilli play roles in intestinal epithelial barrier regulation, we compared miRNA expression levels in intestinal epithelial cells (IECs) under Lactobacillus casei (L. casei LC01) treatment. IECs and L. casei LC01 were co-cultured and then subjected to microRNA microarray assay. qRT-PCR, western blot and ELISA were used to detect the expression of occludin (OCLN) and zonula occludens 1 (ZO1/TJP1). The interaction between miRNAs and L. casei LC01 acting in IECs was investigated through transfection of RNA oligoribonucleotides and pcDNA 3.1 plasmid. The results are as follows: 1) L. casei LC01 decreased the expression of miR-144 and FD4 and promoted OCLN and ZO1 expression in IECs; 2) L. casei LC01 enhanced the barrier function of IECs via downregulation of miR-144 and upregulation of OCLN and ZO1; 3) Under L. casei LC01 treatment, OCLN and ZO1 overexpression could partially eliminate the promoting effect of miR-144 on intestinal permeability in IECs. Our results demonstrate that L. casei LC01 regulates intestinal permeability of IECs through miR-144 targeting of OCLN and ZO1. L. casei LC01 can be a possible therapeutic target for managing dysfunction of the intestinal epithelial barrier.

Inhibition of Interleukin-1α-induced Intestinal Epithelial Tight Junction Permeability by Curcumin Treatment in Caco-2 Cells in Caco-2 Cells (Caco-2 세포에서 커큐민 처리에 의한 IL-1α로 유도된 소장 상피세포의 tight junction 투과성 저해)

  • Kim, Choon Young
    • Journal of Life Science
    • /
    • v.26 no.9
    • /
    • pp.1082-1087
    • /
    • 2016
  • The intestinal tight junction (TJ) plays an important role as a paracellular barrier. Impaired TJ permeability and enhanced proinflammatory cytokine production are crucial pathophysiological mechanisms in inflammatory bowel diseases (IBDs). Although proinflammatory cytokines, tumor necrosis factor-alpha and interluekin-1 beta, which are markedly increased in IBD patients, have been reported to increase intestinal TJ permeability, the role of interleukin-1 alpha (IL-1α) in the TJ has not been studied. Phytochemicals could prevent proinflammatory cytokine-caused TJ alteration. Curcumin (CCM), a biologically active component of turmeric, has a strong anti-inflammatory activity. The purpose of this study was to elucidate the effect of IL-1α on intestinal epithelial TJ permeability and the role of CCM in IL-1α′s action on TJ in an in vitro intestinal epithelial system, Caco-2 monolayers. The TJ integrity of Caco-2 monolayers was estimated by measuring the flux of FITC-labeled dextran and transepithelial electrical resistance (TEER). Apical IL-1α (100 ng/ml) treatment elevated TJ permeability and suppressed TEER of Caco-2 monolayers. Pretreatment with CCM (20 μM) for 30 min significantly inhibited IL-1α-induced TJ alterations, such as increased TJ permeability and decreased in TEER values. These results demonstrated that IL-1α-induced increases in Caco-2 TJ permeability and CCM blocked the action of IL-1α in the TJ.

Effect of Unfermented and Fermented Atractylodes macrocephalae on Gut Permeability and Lipopolysaccharide-Induced Inflammation (백출 및 발효백출의 장점막 투과성 개선 효과 및 항염증효과)

  • Han, Kyungsun;Kim, Kicheol;Wang, Jinghua;Kim, Hojun
    • Journal of Korean Medicine for Obesity Research
    • /
    • v.13 no.1
    • /
    • pp.24-32
    • /
    • 2013
  • Objectives: The aim of this study is to investigate anti-imflammatory and protective effect for intestinal epithelial cells with Atractylodes macrocephae (AM), a traditional Korean Herbal medicine and fermented Atractylodes macrocephae (FAM) with Lactobacillus plantarum. Methods: HCT-116 and Raw 264.7 cells were used in this study. Using NO assay, we measured lipopolysaccharide (LPS)-induced anti-inflammatory effect. We measured permeability of intestinal epithelial cells with transepithelial electrical resistance and horseradish peroxide flux assay. Water soluble tetrazolium salt assay was used to see cell proliferation. All the results were presented in mean and standard deviation. We used Student's t-test for analyzing significance of results. Results: In Raw 264.7 cells NO production decreased 22.4% with pre-treatment of AM and FAM, especially with FAM in high concentration. In HCT-116 cells LPS-induced intestinal permeability had a protective effect with both AM and FAM, which was also tend to be proportional to the concentration. Cell viability increased up to 135.52% after treatment of high concentration of FAM in HCT-116, while there was no significant change in Raw 264.7 cells with herb treatments. Conclusions: These results show evidence that AM, especially fermented ones, significantly reduced intestinal membrane permeability. They also had a protective effect as well as an anti-inflammation effect for HCT-116 and Raw 264.7 cells. This suggest that FAM may be a therapeutic agent for Leaky gut syndrome by reducing intestinal permeability.

Berberine Prevents Intestinal Mucosal Barrier Damage During Early Phase of Sepsis in Rat through the Toll-Like Receptors Signaling Pathway

  • Li, Guo-Xun;Wang, Xi-Mo;Jiang, Tao;Gong, Jian-Feng;Niu, Ling-Ying;Li, Ning
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.19 no.1
    • /
    • pp.1-7
    • /
    • 2015
  • Our previous study has shown berberine prevents damage to the intestinal mucosal barrier during early phase of sepsis in rat through mechanisms independent of the NOD-like receptors signaling pathway. In this study, we explored the regulatory effects of berberine on Toll-like receptors during the intestinal mucosal damaging process in rats. Male Sprague-Dawlay (SD) rats were treated with berberine for 5 d before undergoing cecal ligation and puncture (CLP) to induce polymicrobial sepsis. The expression of Toll-like receptor 2 (TLR 2), TLR 4, TLR 9, the activity of nuclear factor-kappa B ($NF-{\kappa}B$), the levels of selected cytokines and chemokines, percentage of cell death in intestinal epithelial cells, and mucosal permeability were investigated at 0, 2, 6, 12 and 24 h after CLP. Results showed that the tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$) and interleukin-6 (IL-6) level were significantly lower in berberine-treated rats compared to the control animals. Conversely, the expression level of tight junction proteins, percentage of cell death in intestinal epithelial cells and the mucosal permeability were significantly higher in berberine-treated rats. The mRNA expression of TLR 2, TLR 4, and TLR 9 were significantly affected by berberine treatment. Our results indicate that pretreatment with berberine attenuates tissue injury and protects the intestinal mucosal barrier in early phase of sepsis and this may possibly have been mediated through the TLRs pathway.

Bacillus subtilis Protects Porcine Intestinal Barrier from Deoxynivalenol via Improved Zonula Occludens-1 Expression

  • Gu, Min Jeong;Song, Sun Kwang;Park, Sung Moo;Lee, In Kyu;Yun, Cheol-Heui
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.27 no.4
    • /
    • pp.580-586
    • /
    • 2014
  • Intestinal epithelial cells (IECs) forming the barrier for the first-line of protection are interconnected by tight junction (TJ) proteins. TJ alteration results in impaired barrier function, which causes potentially excessive inflammation leading to intestinal disorders. It has been suggested that toll-like receptor (TLR) 2 ligands and some bacteria enhance epithelial barrier function in humans and mice. However, no such study has yet to be claimed in swine. The aim of the present study was to examine whether Bacillus subtilis could improve barrier integrity and protection against deoxynivalenol (DON)-induced barrier disruption in porcine intestinal epithelial cell line (IPEC-J2). We found that B. subtilis decreased permeability of TJ and improved the expression of zonula occludens (ZO)-1 and occludin during the process of forming TJ. In addition, ZO-1 expression of IPEC-J2 cells treated with B. subtilis was up-regulated against DON-induced damage. In conclusion, B. subtilis may have potential to enhance epithelial barrier function and to prevent the cells from DON-induced barrier dysfunction.

Effects of functional nutrients on chicken intestinal epithelial cells induced with oxidative stress

  • Hyun Woo Kim;Seung Yun Lee;Sun Jin Hur;Dong Yong Kil;Jong Hyuk Kim
    • Journal of Animal Science and Technology
    • /
    • v.65 no.5
    • /
    • pp.1040-1052
    • /
    • 2023
  • The objective of this study was to investigate the protective effects of functional nutrients including various functional amino acids, vitamins, and minerals on chicken intestinal epithelial cells (cIECs) treated with oxidative stress. The cIECs were isolated from specific pathogen free eggs. Cells were exposed to 0 mM supplement (control), 20 mM threonine (Thr), 0.4 mM tryptophan (Trp), 1 mM glycine (Gly), 10 μM vitamin C (VC), 40 μM vitamin E (VE), 5 μM vitamin A (VA), 34 μM chromium (Cr), 0.42 μM selenium (Se), and 50 μM zinc (Zn) for 24 h with 6 replicates for each treatment. After 24 h, cells were further incubated with fresh culture medium (positive control, PC) or 1 mM H2O2 with different supplements (negative control, NC and each treatment). Oxidative stress was measured by cell proliferation, whereas tight junction barrier function was analyzed by fluorescein isothiocyanate (FITC)-dextran permeability and transepithelial electrical resistance (TEER). Results indicated that cell viability and TEER values were less (p < 0.05) in NC treatments with oxidative stress than in PC treatments. In addition, FITC-dextran values were greater (p < 0.05) in NC treatments with oxidative stress than in PC treatments. The supplementations of Thr, Trp, Gly, VC, and VE in cells treated with H2O2 showed greater (p < 0.05) cell viability than the supplementation of VA, Cr, Se, and Zn. The supplementations of Trp, Gly, VC, and Se in cells treated with H2O2 showed the least (p < 0.05) cellular permeability. In addition, the supplementation of Thr, VE, VA, Cr, and Zn in cells treated with H2O2 decreased (p < 0.05) cellular permeability. At 48 h, the supplementations of Thr, Trp, and Gly in cells treated with H2O2 showed the greatest (p < 0.05) TEER values among all treatments, and the supplementations of VC and VE in cells treated with H2O2 showed greater (p < 0.05) TEER values than the supplementations of VA, Cr, Se, and Zn in cells treated with H2O2. In conclusion, Thr, Trp, Gly, and VC supplements were effective in improving cell viability and intestinal barrier function of cIECs exposed to oxidative stress.

Curcumin protects against the intestinal ischemia-reperfusion injury: involvement of the tight junction protein ZO-1 and TNF-α related mechanism

  • Tian, Shuying;Guo, Ruixue;Wei, Sichen;Kong, Yu;Wei, Xinliang;Wang, Weiwei;Shi, Xiaomeng;Jiang, Hongyu
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.20 no.2
    • /
    • pp.147-152
    • /
    • 2016
  • Present study aimed to investigate the effect of curcumin-pretreatment on intestinal I/R injury and on intestinal mucosa barrier. Thirty Wistar rats were randomly divided into: sham, I/R, and curcumin groups (n=10). Animals in curcumin group were pretreated with curcumin by gastric gavage (200 mg/kg) for 2 days before I/R. Small intestine tissues were prepared for Haematoxylin & Eosin (H&E) staining. Serum diamine oxidase (DAO) and tumor necrosis factor (TNF)-${\alpha}$ levels were measured. Expression of intestinal TNF-${\alpha}$ and tight junction protein (ZO-1) proteins was detected by Western blot and/or immunohistochemistry. Serum DAO level and serum and intestinal TNF-${\alpha}$ leves were significantly increased after I/R, and the values were markedly reduced by curcumin pretreatment although still higher than that of sham group (p<0.05 or p<0.001). H&E staining showed the significant injury to intestinal mucosa following I/R, and curcumin pretreatment significantly improved the histological structure of intestinal mucosa. I/R insult also induced significantly down-regulated expression of ZO-1, and the effect was dramatically attenuated by curcumin-pretreatment. Curcumin may protect the intestine from I/R injury through restoration of the epithelial structure, promotion of the recovery of intestinal permeability, as well as enhancement of ZO-1 protein expression, and this effect may be partly attributed to the TNF-${\alpha}$ related pathway.