Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 2030, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765731

ABSTRACT

Intestinal epithelial barrier is affected by multiple factors, such as tumour necrosis factor-α (TNF-α). Plasma concentration of TNF-α is higher in patients with Crohn's disease (CD) than healthy controls (HC) and correlates positively with disease activity. This study aimed to determine the effect of plasma from active, inactive CD patients on intestinal barrier function and to investigate the underlying mechanism. Plasma samples were collected from CD patients and HC. 3D Caco-2 cysts were treated with plasma or TNF-α, with or without pre-incubation of adalimumab (a monoclonal antibody that antagonizes TNF-α) or JNK inhibitor SP600125. The results demonstrated that exposure of the cysts to plasma from CD patients resulted in enhanced paracellular permeability in a disease activity-dependent manner. Compared to HC, active CD plasma decreased ZO-1 and OCCLUDIN expression on mRNA and protein levels, and led to an increased JNK phosphorylation. Pre-incubation with adalimumab or SP600125 ameliorated TJ disruption and barrier dysfunction induced by plasma from CD patients. These results indicate that plasma from CD patients is able to induce epithelial barrier disruption, in part through TNF-α induced TJs modulation. The data also demonstrate an involvement of MAPK pathway, in particular the JNK isoform, in CD patient plasma-induced barrier dysfunction.


Subject(s)
Intestinal Mucosa/drug effects , Permeability/drug effects , Plasma/metabolism , Adalimumab/pharmacology , Adult , Anthracenes/pharmacology , Caco-2 Cells , Cell Culture Techniques/methods , Cell Membrane Permeability/drug effects , Crohn Disease/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Middle Aged , Occludin/metabolism , Phosphorylation , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Zonula Occludens-1 Protein/metabolism
2.
PLoS One ; 10(5): e0123498, 2015.
Article in English | MEDLINE | ID: mdl-25978614

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is a disorder with multifactorial pathophysiology. Intestinal barrier may be altered, especially in diarrhea-predominant IBS (IBS-D). Several mediators may contribute to increased intestinal permeability in IBS. AIM: We aimed to assess effects of tryptase and LPS on in vitro permeability using a 3-dimensional cell model after basolateral cell exposure. Furthermore, we assessed the extent to which these mediators in IBS plasma play a role in intestinal barrier function. MATERIALS AND METHODS: Caco-2 cells were grown in extracellular matrix to develop into polarized spheroids and were exposed to tryptase (10 - 50 mU), LPS (1 - 50 ng/mL) and two-fold diluted plasma samples of 7 patients with IBS-D, 7 with constipation-predominant IBS (IBS-C) and 7 healthy controls (HC). Barrier function was assessed by the flux of FITC-dextran (FD4) using live cell imaging. Furthermore, plasma tryptase and LPS were determined. RESULTS: Tryptase (20 and 50 mU) and LPS (6.25 - 50 ng/mL) significantly increased Caco-2 permeability versus control (all P< 0.05). Plasma of IBS-D only showed significantly elevated median tryptase concentrations (7.1 [3.9 - 11.0] vs. 4.2 [2.2 - 7.0] vs. 4.2 [2.5 - 5.9] µg/mL; P<0.05) and LPS concentrations (3.65 [3.00 - 6.10] vs. 3.10 [2.60-3.80] vs. 2.65 [2.40 - 3.40] EU/ml; P< 0.05) vs. IBS-C and HC. Also, plasma of IBS-D increased Caco-2 permeability versus HC (0.14450 ± 0.00472 vs. 0.00021 ± 0.00003; P < 0.001), which was attenuated by selective inhibition of tryptase and LPS (P< 0.05). CONCLUSION: Basolateral exposure of spheroids to plasma of IBS-D patients resulted in a significantly increased FD4 permeation, which was partially abolished by selective inhibition of tryptase and LPS. These findings point to a role of systemic tryptase and LPS in the epithelial barrier alterations observed in patients with IBS-D.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/drug effects , Irritable Bowel Syndrome/metabolism , Tryptases/pharmacology , Caco-2 Cells , Extracellular Matrix , Humans , Lipopolysaccharides/pharmacology
3.
PLoS One ; 9(9): e107421, 2014.
Article in English | MEDLINE | ID: mdl-25226407

ABSTRACT

BACKGROUND: Ethanol-induced gut barrier disruption is associated with several gastrointestinal and liver disorders. AIM: Since human data on effects of moderate ethanol consumption on intestinal barrier integrity and involved mechanisms are limited, the objectives of this study were to investigate effects of a single moderate ethanol dose on small and large intestinal permeability and to explore the role of mitogen activated protein kinase (MAPK) pathway as a primary signaling mechanism. METHODS: Intestinal permeability was assessed in 12 healthy volunteers after intraduodenal administration of either placebo or 20 g ethanol in a randomised cross-over trial. Localization of the tight junction (TJ) and gene expression, phosphorylation of the MAPK isoforms p38, ERK and JNK as indicative of activation were analyzed in duodenal biopsies. The role of MAPK was further examined in vitro using Caco-2 monolayers. RESULTS: Ethanol increased small and large intestinal permeability, paralleled by redistribution of ZO-1 and occludin, down-regulation of ZO-1 and up-regulation of myosin light chain kinase (MLCK) mRNA expression, and increased MAPK isoforms phosphorylation. In Caco-2 monolayers, ethanol increased permeability, induced redistribution of the junctional proteins and F-actin, and MAPK and MLCK activation, as indicated by phosphorylation of MAPK isoforms and myosin light chain (MLC), respectively, which could be reversed by pretreatment with either MAPK inhibitors or the anti-oxidant L-cysteine. CONCLUSIONS: Administration of moderate ethanol dosage can increase both small and colon permeability. Furthermore, the data indicate a pivotal role for MAPK and its crosstalk with MLCK in ethanol-induced intestinal barrier disruption. TRIAL REGISTRATION: ClinicalTrials.gov NCT00928733.


Subject(s)
Ethanol/adverse effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinases , Signal Transduction/drug effects , Actins/metabolism , Adolescent , Adult , Cell Line , Enzyme Activation , Ethanol/administration & dosage , Ethanol/blood , Fatty Acids/metabolism , Healthy Volunteers , Humans , Liver/metabolism , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Permeability , Phosphorylation , Protein Isoforms , Protein Transport , Tight Junction Proteins/metabolism , Young Adult
4.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G286-94, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24904079

ABSTRACT

There is compelling evidence indicating that ethanol and its oxidative metabolite acetaldehyde can disrupt intestinal barrier function. Apart from the tight junctions, mucins secreted by goblet cells provide an effective barrier. Ethanol has been shown to induce goblet cell injury associated with alterations in mucin glycosylation. However, effects of its most injurious metabolite acetaldehyde remain largely unknown. This study aimed to assess short-term effects of acetaldehyde (0, 25, 50, 75, 100 µM) on functional characteristics of intestinal goblet-like cells (LS174T). Oxidative stress, mitochondrial function, ATP, and intramitochondrial calcium (Ca(2+)) were assessed by dichlorofluorescein, methyltetrazolium, and bioluminescence, MitoTracker green and rhod-2 double-labeling. Membrane integrity and apoptosis were evaluated by measuring lactate dehydrogenase (LDH), caspase 3/7, and cleavage of cytokeratin 18 (CK18). Expression of mucin 2 (MUC2) was determined by cell-based ELISA. Acetaldehyde significantly increased reactive oxygen species generation and decreased mitochondrial function compared with negative controls (P < 0.05). In addition, acetaldehyde dose-dependently decreased ATP levels and induced intramitochondrial Ca(2+) accumulation compared with negative controls (P < 0.05). Furthermore, acetaldehyde induced LDH release and increased caspase3/7 activity and percentage of cells expressing cleaved CK18 and increased MUC2 protein expression compared with negative controls (P < 0.0001). ATP depletion and LDH release could be largely prevented by the antioxidant N-acetylcysteine, suggesting a pivotal role for oxidative stress. Our data demonstrate that acetaldehyde has distinct oxidant-dependent metabolic and cytotoxic effects on LS174T cells that can lead to induction of cellular apoptosis. These effects may contribute to acetaldehyde-induced intestinal barrier dysfunction and subsequently to liver injury.


Subject(s)
Acetaldehyde/toxicity , Apoptosis/drug effects , Colon/drug effects , Energy Metabolism/drug effects , Goblet Cells/drug effects , Oxidants/toxicity , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Antioxidants/pharmacology , Calcium/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Survival/drug effects , Colon/metabolism , Colon/pathology , Dose-Response Relationship, Drug , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Keratin-18/metabolism , L-Lactate Dehydrogenase/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mucin-2/metabolism , Reactive Oxygen Species/metabolism , Time Factors
5.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G677-85, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24557761

ABSTRACT

Evidence indicates that ethanol-induced intestinal barrier dysfunction and subsequent endotoxemia plays a key role in the pathogenesis of alcoholic liver disease. Recently, it has been demonstrated that ethanol induces RhoA kinase activation in intestinal epithelium, thereby disrupting barrier integrity. In this study, the role of a rise in intracellular calcium concentration ([Ca(2+)]i) in ethanol-induced Rho-associated coiled coil-forming kinase (Rho/ROCK) activation and barrier disruption was investigated in Caco-2 cell monolayers. Treatment of Caco-2 monolayers with 40 mmol/l ethanol induced [Ca(2+)]i release as indicated by increased relative fluorescent units of Fluo-3 from 0.06 ± 0.02 to 2.27 ± 1.96 (P < 0.0001). Pretreatment with 1,2-bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM) completely inhibited the release, whereas the inositol 1,4,5-triphosphate receptor (IP3R)-antagonist, Xestospongin C, partially inhibited the ethanol-induced [Ca(2+)]i release (from 2.27 ± 1.96 to 0.03 ± 0.01; P < 0.0001 and from 2.27 ± 1.96 to 1.19 ± 1.80; P < 0.001, respectively). The rise in [Ca(2+)]i was paralleled with increased intestinal permeability, which could be attenuated by either BAPTA-AM or Xestospongin C. Furthermore, ethanol induced Rho/ROCK activation, as indicated by increased phosphorylation of myosin-binding subunit, which could be prevented either by BAPTA, Xestospongin C, or the specific Rho/ROCK inhibitor Y27632. Finally, inhibition of Rho/ROCK kinase by Y27632 ameliorated the ethanol-induced redistribution of zonula occluden-1, adherens junction proteins including E-cadherin and ß-catenin, and also disorganization of F-actin. These findings suggest that ethanol-induced [Ca(2+)]i release, mediated by stimulating IP3R-gated Ca(2+) channel, activates Rho/ROCK in Caco-2 cells, thereby contributing to ethanol-induced intestinal barrier dysfunction.


Subject(s)
Egtazic Acid/analogs & derivatives , Ethanol/pharmacology , Intestinal Mucosa , Liver Diseases, Alcoholic , rho-Associated Kinases/metabolism , Caco-2 Cells , Calcium/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/physiopathology , Permeability/drug effects , Phosphorylation , Solvents/pharmacology , Tight Junctions/metabolism
6.
Alcohol Clin Exp Res ; 38(2): 344-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24033729

ABSTRACT

BACKGROUND: Acetaldehyde (AcH) is mutagenic and can reach high concentrations in colonic lumen after ethanol consumption and is associated with intestinal barrier dysfunction and an increased risk of progressive cancers, including colorectal carcinoma. Snail, the transcription factor of epithelial-mesenchymal transition, is known to down-regulate expression of tight junction (TJ) and adherens junction (AJ) proteins, resulting in loss of epithelial integrity, cancer progression, and metastases. As AcH is mutagenic, the role of Snail in the AcH-induced disruption of intestinal epithelial TJs deserves further investigation. Our aim was to investigate the role of oxidative stress and Snail activation in AcH-induced barrier disruption in Caco-2 monolayers. METHODS: The monolayers were exposed from the apical side to AcH ± L-cysteine. Reactive oxygen species (ROS) generation and Snail activation were assessed by ELISA and immunofluorescence. Paracellular permeability, localization, and expression of ZO-1, occludin, E-cadherin, and ß-catenin were examined using transepithelial electrical resistance (TEER), fluorescein isothiocyanate-labeled dextran 4 kDa (FITC-D4), immunofluorescence, and ELISA, respectively. Involvement of Snail was further addressed by inhibiting Snail using small interfering RNA (siRNA). RESULTS: Exposure to 25 µM AcH increased ROS generation and ROS-dependently induced Snail phosphorylation. In addition, AcH increased paracellular permeability (decrease in TEER and increase in FITC-D4 permeation) in association with redistribution and decrease of TJ and AJ protein levels, which could be attenuated by L-cysteine. Knockdown of Snail by siRNA attenuated the AcH-induced redistribution and decrease in the TJ and AJ proteins, in association with improvement of the barrier function. CONCLUSIONS: Our data demonstrate that oxidative stress-mediated Snail phosphorylation is likely a novel mechanism contributing to the deleterious effects of AcH on the TJ and AJ, and intestinal barrier function.


Subject(s)
Acetaldehyde/toxicity , Epithelial-Mesenchymal Transition/drug effects , Intestinal Mucosa/drug effects , Transcription Factors/physiology , Acetaldehyde/pharmacology , Adherens Junctions/drug effects , Biotransformation/drug effects , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cysteine/pharmacology , Humans , Phosphorylation , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Snail Family Transcription Factors , Tight Junctions/drug effects , Transcription Factors/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 306(2): G147-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24264047

ABSTRACT

Intestinal barrier dysfunction, facilitating translocation of bacteria and bacterial products, plays an important role in the pathophysiology of liver cirrhosis and its complications. Increased intestinal permeability has been found in patients with liver cirrhosis, but data on small and large intestine permeability and tight junctions (TJs) in patients with compensated cirrhosis are scarce. We aimed to investigate both small and large intestine permeability in patients with stable compensated cirrhosis compared with healthy controls and evaluated the expression of TJ proteins in mucosal biopsies at duodenal and sigmoid level. Intestinal permeability was assessed in 26 patients with compensated cirrhosis and 27 matched controls using a multisugar test. Duodenal and sigmoid biopsies were available from a subgroup for analyses of gene transcription and expression of key TJ proteins by qRT-PCR and ELISA, respectively. Median 0-5-h urinary sucrose excretion and lactulose/rhamnose ratio were comparable between patients with compensated cirrhosis and controls, whereas 5-24-h urinary sucralose/erythritol ratio was increased in these patients. Downregulation of gene transcription was found for claudin-3 in duodenal biopsies and claudin-4 in sigmoid biopsies, and at the protein level occludin expression was significantly increased in both duodenal and sigmoid biopsies. This study shows that gastroduodenal and small intestine permeability are not altered, whereas large intestine permeability is increased in patients with stable compensated cirrhosis. Only limited alterations were found regarding the expression of TJ proteins in both the small and large intestine.


Subject(s)
Intestine, Large/metabolism , Liver Cirrhosis/metabolism , Adolescent , Adult , Aged , Case-Control Studies , Claudin-4/metabolism , Colon, Sigmoid/metabolism , Duodenum/metabolism , Endoscopy , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intestine, Small/metabolism , Male , Middle Aged , Occludin/metabolism , Permeability , Prospective Studies , Real-Time Polymerase Chain Reaction , Tight Junction Proteins/metabolism , Young Adult
8.
J Nutr ; 143(12): 1872-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24132573

ABSTRACT

Short-chain fatty acids (SCFAs) have been shown to promote intestinal barrier function, but their protective effects against ethanol-induced intestinal injury and underlying mechanisms remain essentially unknown. The aim of the study was to analyze the influence of SCFAs on ethanol-induced barrier dysfunction and to examine the role of AMP-activated protein kinase (AMPK) as a possible mechanism using Caco-2 monolayers. The monolayers were treated apically with butyrate (2, 10, or 20 mmol/L), propionate (4, 20, or 40 mmol/L), or acetate (8, 40, or 80 mmol/L) for 1 h before ethanol (40 mmol/L) for 3 h. Barrier function was analyzed by measurement of transepithelial resistance and permeation of fluorescein isothiocyanate-labeled dextran. Distribution of the tight junction (TJ) proteins zona occludens-1, occludin, and filamentous-actin (F-actin) was examined by immunofluorescence. Metabolic stress was determined by measuring oxidative stress, mitochondrial function, and ATP using dichlorofluorescein diacetate, dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide, and bioluminescence assay, respectively. AMPK was knocked down by small interfering RNA (siRNA), and its activity was assessed by a cell-based ELISA. Exposure to ethanol significantly impaired barrier function compared with controls (P < 0.0001), disrupted TJ and F-actin cytoskeleton integrity, and induced metabolic stress. However, pretreatment with 2 mmol/L butyrate, 4 mmol/L propionate, and 8 mmol/L acetate significantly alleviated the ethanol-induced barrier dysfunction, TJ and F-actin disruption, and metabolic stress compared with ethanol-exposed monolayers (P < 0.0001). The promoting effects on barrier function were abolished by inhibiting AMPK using either compound C or siRNA. These observations indicate that SCFAs exhibit protective effects against ethanol-induced barrier disruption via AMPK activation, suggesting a potential for SCFAs as prophylactic and/or therapeutic factors against ethanol-induced gut leakiness.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ethanol/pharmacology , Fatty Acids/pharmacology , Intestinal Mucosa/drug effects , Caco-2 Cells , Enzyme Activation , Fatty Acids/administration & dosage , Humans , Intestinal Mucosa/enzymology
9.
Nutr Rev ; 71(7): 483-99, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23815146

ABSTRACT

Ethanol is widely consumed and is associated with an increasing global health burden. Several reviews have addressed the effects of ethanol and its oxidative metabolite, acetaldehyde, on the gastrointestinal (GI) tract, focusing on carcinogenic effects or alcoholic liver disease. However, both the oxidative and the nonoxidative metabolites of ethanol can affect the epithelial barrier of the small and large intestines, thereby contributing to GI and liver diseases. This review outlines the possible mechanisms of ethanol metabolism as well as the effects of ethanol and its metabolites on the intestinal barrier. Limited studies in humans and supporting in vitro data have indicated that ethanol as well as mainly acetaldehyde can increase small intestinal permeability. Limited evidence also points to increased colon permeability following exposure to ethanol or acetaldehyde. In vitro studies have provided several mechanisms for disruption of the epithelial barrier, including activation of different cell-signaling pathways, oxidative stress, and remodeling of the cytoskeleton. Modulation via intestinal microbiota, however, should also be considered. In conclusion, ethanol and its metabolites may act additively or even synergistically in vivo. Therefore, in vivo studies investigating the effects of ethanol and its byproducts on permeability of the small and large intestines are warranted.


Subject(s)
Ethanol/metabolism , Gastrointestinal Diseases/etiology , Intestinal Mucosa/drug effects , Liver Diseases, Alcoholic/etiology , Ethanol/pharmacokinetics , Ethanol/toxicity , Gastrointestinal Diseases/epidemiology , Humans , Intestinal Mucosa/metabolism , Liver Diseases, Alcoholic/epidemiology , Permeability
10.
Liver Int ; 33(10): 1457-69, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23879434

ABSTRACT

Recent evidence suggests that translocation of bacteria and bacterial products, such as endotoxin from the intestinal lumen into the systemic circulation is a contributing factor in the pathogenesis of chronic liver diseases and the development of complications in cirrhosis. In addition to alterations in the intestinal microbiota and immune system, dysfunction of the intestinal epithelial barrier may be an important factor facilitating bacterial translocation. This review aims to provide an overview of the current evidence of intestinal epithelial barrier dysfunction in human chronic liver diseases and cirrhosis, and to discuss possible contributing factors and mechanisms. Data suggest the presence of intestinal epithelial barrier dysfunction in patients with chronic liver diseases, but are more convincing in patients with cirrhosis, especially in those with complications. The barrier dysfunction can result from both direct and indirect effects of aetiological factors, such as alcohol and obesity, which can cause chronic liver diseases and ultimately cirrhosis. On the other hand characteristics of cirrhosis itself, including portal hypertension, alterations in the intestinal microbiota, inflammation and oxidative stress can affect barrier function of both small and large intestine and may contribute to the development of complications. In conclusion, there are indications for intestinal epithelial barrier dysfunction in patients with chronic liver diseases and especially in patients with cirrhosis, which can be caused by various factors affecting both the small and large intestine.


Subject(s)
Bacterial Translocation/physiology , Hypertension, Portal/pathology , Intestinal Mucosa/physiopathology , Liver Cirrhosis/physiopathology , Humans , Hypertension, Portal/etiology , Liver Cirrhosis/complications , Oxidative Stress/physiology , Permeability
11.
PLoS One ; 8(3): e58561, 2013.
Article in English | MEDLINE | ID: mdl-23526996

ABSTRACT

BACKGROUND & AIMS: Evidence is accumulating that ethanol and its oxidative metabolite, acetaldehyde, can disrupt intestinal epithelial integrity, an important factor contributing to ethanol-induced liver injury. However, ethanol can also be metabolized non-oxidatively generating phosphatidylethanol and fatty acid ethyl esters (FAEEs). This study aims to investigate the effects of FAEEs on barrier function, and to explore the role of oxidative stress as possible mechanism. METHODS: Epithelial permeability was assessed by paracellular flux of fluorescein isothiocyanate-conjugated dextran using live cell imaging. Cell integrity was evaluated by lactate dehydrogenase release. Localization and protein levels of ZO-1 and occludin were analyzed by immunofluorescence and cell-based ELISA, respectively. Intracellular oxidative stress and cellular ATP levels were measured by dichlorofluorescein and luciferase driven bioluminescence, respectively. RESULTS: In vitro, ethyl oleate and ethyl palmitate dose dependently increased permeability associated with disruption and decreased ZO-1 and occludin protein levels, respectively, and increased intracellular oxidative stress without compromising cell viability. These effects could partially be attenuated by pretreatment with the antioxidant, resveratrol, pointing to the role of oxidative stress in the FAEEs-induced intestinal barrier dysfunction. CONCLUSIONS: These findings show that FAEEs can induce intestinal barrier dysfunction by disrupting the tight junctions, most likely via reactive oxygen species-dependent mechanism.


Subject(s)
Intestinal Mucosa/drug effects , Intestinal Mucosa/physiopathology , Oleic Acids/toxicity , Palmitic Acids/toxicity , Adenosine Triphosphate/metabolism , Caco-2 Cells , Cell Culture Techniques , Cell Membrane Permeability/drug effects , Ethanol/metabolism , Ethanol/toxicity , Humans , Models, Biological , Occludin/metabolism , Oleic Acids/metabolism , Oxidative Stress , Palmitic Acids/metabolism , Reactive Oxygen Species/metabolism , Resveratrol , Stilbenes/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism
12.
PLoS One ; 7(4): e35008, 2012.
Article in English | MEDLINE | ID: mdl-22563376

ABSTRACT

BACKGROUND: Intestinal barrier dysfunction and translocation of endotoxins are involved in the pathogenesis of alcoholic liver disease. Exposure to ethanol and its metabolite, acetaldehyde at relatively high concentrations have been shown to disrupt intestinal epithelial tight junctions in the conventional two dimensional cell culture models. The present study investigated quantitatively and qualitatively the effects of ethanol at concentrations detected in the blood after moderate ethanol consumption, of its metabolite acetaldehyde and of the combination of both compounds on intestinal barrier function in a three-dimensional cell culture model. METHODS AND FINDINGS: Caco-2 cells were grown in a basement membrane matrix (Matrigel™) to induce spheroid formation and were then exposed to the compounds at the basolateral side. Morphological differentiation of the spheroids was assessed by immunocytochemistry and transmission electron microscopy. The barrier function was assessed by the flux of FITC-labeled dextran from the basal side into the spheroids' luminal compartment using confocal microscopy. Caco-2 cells grown on Matrigel assembled into fully differentiated and polarized spheroids with a central lumen, closely resembling enterocytes in vivo and provide an excellent model to study epithelial barrier functionality. Exposure to ethanol (10-40 mM) or acetaldehyde (25-200 µM) for 3 h, dose-dependently and additively increased the paracellular permeability and induced redistribution of ZO-1 and occludin without affecting cell viability or tight junction-encoding gene expression. Furthermore, ethanol and acetaldehyde induced lysine residue and microtubules hyperacetylation. CONCLUSIONS: These results indicate that ethanol at concentrations found in the blood after moderate drinking and acetaldehyde, alone and in combination, can increase the intestinal epithelial permeability. The data also point to the involvement of protein hyperacetylation in ethanol- and acetaldehyde-induced loss of tight junctions integrity.


Subject(s)
Acetaldehyde/pharmacology , Epithelial Cells/drug effects , Ethanol/pharmacology , Tight Junctions/drug effects , Acetylation/drug effects , Caco-2 Cells , Cell Culture Techniques , Cell Membrane Permeability/drug effects , Fluorescent Dyes/chemistry , Humans , Intestines/cytology , Membrane Proteins/metabolism , Occludin , Phosphoproteins/metabolism , Solvents/pharmacology , Spheroids, Cellular/pathology , Tight Junctions/metabolism , Tubulin/metabolism , Zonula Occludens-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...