Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
2.
Curr Nutr Food Sci ; 9(2): 99-107, 2013 May 01.
Article in English | MEDLINE | ID: mdl-24353483

ABSTRACT

Probiotics are beneficial bacteria present in various dietary components and many of these colonize in the human and animal intestine. In the gut probiotics help the host by assisting in maintenance of normal mucosal homeostasis. Probiotics not only help maintain normal function of the gut mucosa, but also protect mucosa from injurious factors such as toxins, allergens and pathogens. The beneficial effect of probiotics is mediated by multiple mechanisms, including cytoprotection, cell proliferation, cell migration, resistance to apoptosis, synthesis of proteins and gene expression. One of the important cytoprotective effects of probiotics in the intestinal mucosa is to strengthen the epithelial tight junctions and preservation of mucosal barrier function. Probiotics not only enhance barrier function by inducing synthesis and assembly of tight junction proteins, but also preventing disruption of tight junctions by injurious factors. Bioactive factors released by probiotics trigger activation of various cell signaling pathways that lead to strengthening of tight junctions and the barrier function. This article reviews and summarizes the current understanding of various probiotics that are involved in the protection of gut barrier function, highlights the cellular and molecular mechanisms involved in the protective effect and addresses the clinical implications of probiotic supplementation.

3.
Tissue Barriers ; 1(4): e25718, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24665411

ABSTRACT

Bile ducts play a crucial role in the formation and secretion of bile as well as excretion of circulating xenobiotic substances. In addition to its secretory and excretory functions, bile duct epithelium plays an important role in the formation of a barrier to the diffusion of toxic substances from bile into the hepatic interstitial tissue. Disruption of barrier function and toxic injury to liver cells appear to be involved in the pathogenesis of a variety of liver diseases such as primary sclerosing cholangitis, primary biliary cirrhosis and cholangiocarcinoma. Although the investigations into understanding the structure and regulation of tight junctions in gut, renal and endothelial tissues have expanded rapidly, very little is known about the structure and regulation of tight junctions in the bile duct epithelium. In this article we summarize the current understanding of physiology and pathophysiology of bile duct epithelium, the structure and regulation of tight junctions in canaliculi and bile duct epithelia and different mechanisms involved in the regulation of disruption and protection of bile duct epithelial tight junctions. This article will make a case for the need of future investigations toward our understanding of molecular organization and regulation of canalicular and bile duct epithelial tight junctions.

4.
Am J Physiol Gastrointest Liver Physiol ; 303(12): G1356-64, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23064762

ABSTRACT

Acetaldehyde is accumulated at high concentrations in the colonic lumen following ethanol administration. Previous studies demonstrated that acetaldehyde disrupts intestinal epithelial tight junctions and increases paracellular permeability. In the present study, we investigated the role of PP2A in the acetaldehyde-induced disruption of intestinal epithelial tight junctions. Caco-2 cell monolayers were exposed to 200-600 µM acetaldehyde for varying times, and the epithelial barrier function was evaluated by measuring transepithelial electrical resistance and inulin permeability. Acetaldehyde treatment resulted in a time-dependent increase in inulin permeability and redistribution of occludin and ZO-1 from the intercellular junctions. Treatment of cells with fostriecin (a PP2A-selective inhibitor) or knockdown of PP2A by siRNA blocked acetaldehyde-induced increase in inulin permeability and redistribution of occludin and ZO-1. The effects of fostriecin and acetaldehyde were confirmed in mouse intestine ex vivo. Acetaldehyde-induced tight junction disruption and barrier dysfunction were also attenuated by a PP2A-specific inhibitory peptide, TPDYFL. Coimmunoprecipitation studies showed that acetaldehyde increased the interaction of PP2A with occludin and induced dephosphorylation of occludin on threonine residues. Fostriecin and TPDYFL significantly reduced acetaldehyde-induced threonine dephosphorylation of occludin. Acetaldehyde failed to change the level of the methylated form of PP2A-C subunit. However, genistein (a tyrosine kinase inhibitor) blocked acetaldehyde-induced association of PP2A with occludin and threonine dephosphorylation of occludin. These results demonstrate that acetaldehyde-induced disruption of tight junctions is mediated by PP2A translocation to tight junctions and dephosphorylation of occludin on threonine residues.


Subject(s)
Acetaldehyde/administration & dosage , Intestinal Mucosa/metabolism , Occludin/metabolism , Protein Phosphatase 2/metabolism , Signal Transduction/physiology , Tight Junctions/physiology , Animals , Caco-2 Cells , Dose-Response Relationship, Drug , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Mice , Signal Transduction/drug effects , Tight Junctions/drug effects
5.
Article in English | MEDLINE | ID: mdl-21474650

ABSTRACT

The role of mitogen-activated protein kinases (MAPK) in the mechanism of EGF-mediated prevention of acetaldehyde-induced tight junction disruption was evaluated in Caco-2 cell monolayers. Pretreatment of cell monolayers with EGF attenuated acetaldehyde-induced decrease in resistance and increase in inulin permeability and redistribution of occludin, zona occludens-1 (ZO-1), E-cadherin, and ß-catenin from the intercellular junctions. EGF rapidly increased the levels of phospho-ERK1/2, phospho-p38 MAPK, and phospho-JNK1. Pretreatment of cell monolayers with U-0126 (inhibitor of ERK activation), but not SB-202190 and SP-600125 (p38 MAPK and JNK inhibitors), significantly attenuated EGF-mediated prevention of acetaldehyde-induced changes in resistance, inulin permeability, and redistribution of occludin and ZO-1. U-0126, but not SB-202190 and SP-600125, also attenuated EGF-mediated prevention of acetaldehyde effect on the midregion F-actin ring. However, EGF-mediated preservation of junctional distribution of E-cadherin and ß-catenin was unaffected by all three inhibitors. Expression of wild-type or constitutively active MEK1 attenuated acetaldehyde-induced redistribution of occludin and ZO-1, whereas dominant-negative MEK1 prevented EGF-mediated preservation of occludin and ZO-1 in acetaldehyde-treated cells. MEK1 expression did not alter E-cadherin distribution in acetaldehyde-treated cells in the presence or absence of EGF. Furthermore, EGF attenuated acetaldehyde-induced tyrosine-phosphorylation of occludin, ZO-1, claudin-3, and E-cadherin. U-0126, but not SB-202190 and SP-600125, prevented EGF effect on tyrosine-phosphorylation of occludin and ZO-1, but not claudin-3, E-cadherin, or ß-catenin. These results indicate that EGF-mediated protection of tight junctions from acetaldehyde requires the activity of ERK1/2, but not p38 MAPK or JNK1/2, and that EGF-mediated protection of adherens junctions is independent of MAPK activities.


Subject(s)
Acetaldehyde/pharmacology , Adherens Junctions/drug effects , Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Tight Junctions/drug effects , Actins/metabolism , Anthracenes/pharmacology , Butadienes/pharmacology , Caco-2 Cells , Cadherins/metabolism , Claudin-3 , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Inulin/physiology , Membrane Proteins/metabolism , Nitriles/pharmacology , Occludin , Permeability/drug effects , Phosphoproteins/metabolism , Pyridines/pharmacology , Zonula Occludens-1 Protein , beta Catenin/metabolism
6.
Am J Physiol Gastrointest Liver Physiol ; 299(3): G572-84, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20595622

ABSTRACT

Gastrointestinal epithelium faces osmotic stress, both at physiological and pathophysiological conditions. JNK activation is an immediate cellular response to osmotic stress. We investigated the effect of osmotic stress on intestinal epithelial barrier function and delineated the role of JNK2 in osmotic stress-induced tight junction (TJ) regulation in Caco-2 cell monolayers and ileum of Jnk(-/-) and Jnk2(-/-) mice. The role of JNK activation in osmotic stress-induced TJ disruption was evaluated using JNK-specific inhibitor and antisense oligonucleotides. Furthermore, the effect of cold restraint stress in vivo on TJ integrity was determined in rats. Osmotic stress disrupted TJs and barrier function in Caco-2 cell monolayers without affecting cell viability. Osmotic stress activated JNK1 and JNK2 and the inhibition of JNK by SP600125 attenuated osmotic stress-induced TJ disruption. TJ disruption and barrier dysfunction by osmotic stress was associated with JNK-dependent remodeling of actin cytoskeleton. Knockdown of JNK2 accelerated TJ assembly and attenuated osmotic stress-induced TJ disruption in Caco-2 cell monolayers. In mouse ileum in vitro, osmotic stress increased paracellular permeability, which was attenuated by SP600125. Osmotic stress disrupted actin cytoskeleton and TJs and increased paracellular permeability in the ileum of wild-type and JNK1(-/-) mice, but not in JNK2(-/-) mouse ileum. Cold restraint stress activated JNK in rat ileum and caused JNK-dependent remodeling of actin cytoskeleton and redistribution of occludin and zona occluden-1 from the intercellular junctions. These results reveal the role of JNK2 in the mechanism of osmotic stress-induced TJ disruption in the intestinal epithelium.


Subject(s)
Epithelial Cells/physiology , Intestinal Mucosa/cytology , Mitogen-Activated Protein Kinase 9/metabolism , Tight Junctions/physiology , Animals , Caco-2 Cells , Calcium/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/drug effects , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Osmotic Pressure , Protein Transport , Rats , Rats, Sprague-Dawley , Stress, Physiological
7.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G186-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20378826

ABSTRACT

Recent studies showed that c-Src and phosphatidylinositol 3 (PI3) kinase mediate the oxidative stress-induced disruption of tight junctions in Caco-2 cell monolayers. The present study evaluated the roles of PI3 kinase and Src kinase in the oxidative stress-induced activation of focal adhesion kinase (FAK) and acceleration of cell migration. Oxidative stress, induced by xanthine and xanthine oxidase system, rapidly increased phosphorylation of FAK on Y397, Y925, and Y577 in the detergent-insoluble and soluble fractions and increased its tyrosine kinase activity. The PI3 kinase inhibitors, wortmannin and LY294002, and the Src kinase inhibitor, 4-amino-5[chlorophyll]-7-[t-butyl]pyrazolo[3-4-d]pyrimidine, attenuated tyrosine phosphorylation of FAK. Oxidative stress induced phosphorylation of c-Src on Y418 by a PI3 kinase-dependent mechanism, whereas oxidative stress-induced activation of PI3 kinase was independent of Src kinase activity. Hydrogen peroxide accelerated Caco-2 cell migration in a concentration-dependent manner. Promotion of cell migration by hydrogen peroxide was attenuated by LY294002 and PP2. Reduced expression of FAK by siRNA attenuated hydrogen peroxide-induced acceleration of cell migration. The expression of constitutively active c-Src(Y527F) enhanced cell migration, whereas the expression of dominant negative c-Src(K296R/Y528F) attenuated hydrogen peroxide-induced stimulation of cell migration. Oxidative stress-induced activation of c-Src and FAK was associated with a rapid increase in the tyrosine phosphorylation and the levels of paxillin and p130(CAS) in actin-rich, detergent-insoluble fractions. This study shows that oxidative stress activates FAK and accelerates cell migration in an intestinal epithelium by a PI3 kinase- and Src kinase-dependent mechanism.


Subject(s)
Cell Movement/drug effects , Focal Adhesion Kinase 1/metabolism , Hydrogen Peroxide/pharmacology , Intestinal Mucosa/drug effects , Oxidants/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/drug effects , Animals , Caco-2 Cells , Chickens , Crk-Associated Substrate Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Focal Adhesion Kinase 1/genetics , Humans , Intestinal Mucosa/enzymology , Mice , Mutation , Oxidative Stress/drug effects , Paxillin/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/genetics , RNA Interference , Time Factors , Tyrosine , Vinculin/metabolism , Xanthine/metabolism , Xanthine Oxidase/metabolism
8.
Methods Mol Biol ; 447: 171-83, 2008.
Article in English | MEDLINE | ID: mdl-18369919

ABSTRACT

A significant body of evidence indicates that endotoxemia plays a crucial role in the pathogenesis of alcoholic liver disease. There are several possible factors that may be involved in inducing alcoholic endotoxemia, but increased intestinal permeability to enteric endotoxins appears to be the major contributing factor. In the normal gut, the epithelial barrier function prevents diffusion of toxins across the epithelium. However, the barrier is disrupted in patients with alcoholic liver disease. We showed that acetaldehyde disrupts intestinal epithelial tight junctions and increases paracellular permeability to endotoxins in Caco-2 cell monolayer, the extensively studied model of the differentiated intestinal epithelium. The mechanisms involved in acetaldehyde-induced increase in intestinal permeability to endotoxins can be elucidated in this model of the intestinal epithelium.


Subject(s)
Acetaldehyde/toxicity , Endotoxins/metabolism , Epithelial Cells/drug effects , Ethanol/toxicity , Intestinal Mucosa/drug effects , Tight Junctions/drug effects , Acetaldehyde/metabolism , Caco-2 Cells , Cell Culture Techniques , Cell Survival/drug effects , Electric Impedance , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Ethanol/metabolism , Fluorescent Antibody Technique , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Inulin/metabolism , L-Lactate Dehydrogenase/metabolism , Liver Diseases, Alcoholic/metabolism , Microscopy, Confocal , Permeability , Tight Junctions/metabolism , Time Factors
9.
Am J Physiol Gastrointest Liver Physiol ; 294(4): G1060-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18292183

ABSTRACT

Probiotics promote intestinal epithelial integrity and reduce infection and diarrhea. We evaluated the effect of Lactobacillus rhamnosus GG-produced soluble proteins (p40 and p75) on the hydrogen peroxide-induced disruption of tight junctions and barrier function in Caco-2 cell monolayers. Pretreatment of cell monolayers with p40 or p75 attenuated the hydrogen peroxide-induced decrease in transepithelial resistance and increase in inulin permeability in a time- and dose-dependent manner. p40 and p75 also prevented hydrogen peroxide-induced redistribution of occludin, ZO-1, E-cadherin, and beta-catenin from the intercellular junctions and their dissociation from the detergent-insoluble fractions. Both p40 and p75 induced a rapid increase in the membrane translocation of PKCbetaI and PKCepsilon. The attenuation of hydrogen peroxide-induced inulin permeability and redistribution of tight junction proteins by p40 and p75 was abrogated by Ro-32-0432, a PKC inhibitor. p40 and p75 also rapidly increased the levels of phospho-ERK1/2 in the detergent-insoluble fractions. U0126 (a MAP kinase inhibitor) attenuated the p40- and p75-mediated reduction of hydrogen peroxide-induced tight junction disruption and inulin permeability. These studies demonstrate that probiotic-secretory proteins protect the intestinal epithelial tight junctions and the barrier function from hydrogen peroxide-induced insult by a PKC- and MAP kinase-dependent mechanism.


Subject(s)
Bacterial Proteins/pharmacology , Hydrogen Peroxide/toxicity , Intestinal Mucosa/drug effects , Lacticaseibacillus rhamnosus/chemistry , Mitogen-Activated Protein Kinases/metabolism , Probiotics/pharmacology , Protein Kinase C/metabolism , Tight Junctions/drug effects , Bacterial Proteins/isolation & purification , Butadienes/pharmacology , Caco-2 Cells , Cadherins/metabolism , Dose-Response Relationship, Drug , Electric Impedance , HT29 Cells , Humans , Indoles/pharmacology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Inulin/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitriles/pharmacology , Occludin , Permeability , Phosphoproteins/metabolism , Probiotics/isolation & purification , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , Pyrroles/pharmacology , Tight Junctions/enzymology , Tight Junctions/metabolism , Time Factors , Zonula Occludens-1 Protein , beta Catenin/metabolism
10.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G308-18, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17446308

ABSTRACT

Bile duct epithelium forms a barrier to the backflow of bile into the liver parenchyma. However, the structure and regulation of the tight junctions in bile duct epithelium is not well understood. In the present study, we evaluated the effect of lipopolysaccharide on tight junction integrity and barrier function in normal rat cholangiocyte monolayers. Lipopolysaccharide disrupts barrier function and increases paracellular permeability in a time- and dose-dependent manner. Lipopolysaccharide induced a redistribution of tight junction proteins, occludin, claudin-1, claudin-4, and zonula occludens (ZO)-1 from the intercellular junctions and reduced the level of ZO-1. Tyrosine kinase inhibitors (genistein and PP2) prevented lipopolysaccharide-induced increase in permeability and subcellular redistribution of ZO-1. Reduced expression of c-Src, TLR4, or LBP by specific small interfering RNA attenuated lipopolysaccharide-induced permeability and redistribution of ZO-1. ML-7, a myosin light chain kinase inhibitor, attenuated LPS-induced permeability. Lipopolysaccharide treatment rapidly increased the phosphorylation of occludin and ZO-1 on tyrosine residues, which was prevented by genistein and PP2. Occludin and ZO-1 were found to be highly phosphorylated on threonine residues in intact cell monolayers. Threonine-phosphorylation of occludin was rapidly reduced by lipopolysaccharide administration. Lipopolysaccharide-induced dephosphorylation of occludin on Thr residues was prevented by genistein and PP2. In conclusion, lipopolysaccharide disrupts the tight junction of a bile duct epithelial monolayer by a c-Src-, TLR4-, LBP-, and myosin light chain kinase-dependent mechanism.


Subject(s)
Acute-Phase Proteins/physiology , Bile Ducts/cytology , Bile Ducts/physiology , Carrier Proteins/physiology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/physiology , Myosin-Light-Chain Kinase/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Tight Junctions/drug effects , Toll-Like Receptor 4/physiology , Animals , Cholangitis, Sclerosing/physiopathology , Epithelium/drug effects , Epithelium/physiology , Genistein/pharmacology , Membrane Proteins/metabolism , Occludin , Permeability/drug effects , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Rats , Zonula Occludens-1 Protein
11.
Am J Physiol Gastrointest Liver Physiol ; 289(2): G367-75, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15718285

ABSTRACT

Acetaldehyde, a toxic metabolite of ethanol oxidation, is suggested to play a role in the increased risk for gastrointestinal cancers in alcoholics. In the present study, the effect of acetaldehyde on tyrosine phosphorylation, immunofluorescence localization, and detergent-insoluble fractions of the tight junction and the adherens junction proteins was determined in the human colonic mucosa. The role of EGF and L-glutamine in prevention of acetaldehyde-induced effects was also evaluated. Acetaldehyde reduced the protein tyrosine phosphatase activity, thereby increasing the tyrosine phosphorylation of occludin, E-cadherin, and beta-catenin. The levels of occludin, zonula occludens-1, E-cadherin, and beta-catenin in detergent-insoluble fractions were reduced by acetaldehyde, while it increased their levels in detergent-soluble fractions. Pretreatment with EGF or L-glutamine prevented acetaldehyde-induced protein tyrosine phosphorylation, redistribution from intercellular junctions, and reduction in the levels of detergent-insoluble fractions of occludin, zonula occludens-1, E-cadherin, and beta-catenin. These results demonstrate that acetaldehyde induces tyrosine phosphorylation and disrupts tight junction and adherens junction in human colonic mucosa, which can be prevented by EGF and glutamine.


Subject(s)
Acetaldehyde/pharmacology , Adherens Junctions/drug effects , Epidermal Growth Factor/pharmacology , Glutamine/pharmacology , Intestinal Mucosa/drug effects , Tight Junctions/drug effects , Colon/cytology , Colon/drug effects , Detergents , Drug Interactions , Female , Humans , In Vitro Techniques , Intestinal Mucosa/cytology , Male , Phosphorylation/drug effects , Solubility , Tyrosine/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 287(3): G510-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331350

ABSTRACT

Role of L-glutamine in the protection of intestinal epithelium from acetaldehyde-induced disruption of barrier function was evaluated in Caco-2 cell monolayer. L-Glutamine reduced the acetaldehyde-induced decrease in transepithelilal electrical resistance and increase in permeability to inulin and lipopolysaccharide in a time- and dose-dependent manner; d-glutamine, L-aspargine, L-arginine, L-lysine, or L-alanine produced no significant protection. The glutaminase inhibitor 6-diazo-5-oxo-L-norleucine failed to affect the L-glutamine-mediated protection of barrier function. L-Glutamine reduced the acetaldehyde-induced redistribution of occludin, zonula occludens-1 (ZO-1), E-cadherin, and beta-catenin from the intercellular junctions. Acetaldehyde dissociates occludin, ZO-1, E-cadherin, and beta-catenin from the actin cytoskeleton, and this effect was reduced by L-glutamine. L-Glutamine induced a rapid increase in the tyrosine phosphorylation of EGF receptor, and the protective effect of L-glutamine was prevented by AG1478, the EGF-receptor tyrosine kinase inhibitor. These results indicate that L-glutamine prevents acetaldehyde-induced disruption of the tight junction and increase in the paracellular permeability in Caco-2 cell monolayer by an EGF receptor-dependent mechanism.


Subject(s)
Acetaldehyde/antagonists & inhibitors , Acetaldehyde/pharmacology , Cell Membrane Permeability/drug effects , Glutamine/pharmacology , Actins/metabolism , Adherens Junctions/drug effects , Caco-2 Cells , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Endotoxins/pharmacology , ErbB Receptors/antagonists & inhibitors , Humans , Lipopolysaccharides/pharmacology , Mannitol/pharmacology , Membrane Proteins/metabolism , Occludin , Phosphorylation , Quinazolines , Tight Junctions/drug effects , Tyrosine/metabolism , Tyrphostins/pharmacology
13.
Am J Physiol Gastrointest Liver Physiol ; 286(6): G881-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15132946

ABSTRACT

A significant body of evidence indicates that endotoxemia and endotoxin-mediated hepatocellular damage play a crucial role in the pathogenesis of alcoholic liver disease. A close correlation between endotoxemia and the severity of alcohol-induced liver injury is supported by a number of clinical and experimental studies. Elevated intestinal permeability appears to be the major factor involved in the mechanism of alcoholic endotoxemia and the pathogenesis of alcoholic liver disease. Ethanol and its metabolic derivatives, acetaldehyde in particular, alter intracellular signal-transduction pathways leading to the disruption of epithelial tight junctions and an increase in paracellular permeability to macromolecules. Studies addressing the mechanisms of such epithelial disruption and the protective factors that prevent ethanol and acetaldehyde-mediated disruption of epithelial tight junctions are critically important in the investigations toward the search of preventive and therapeutic strategies for alcoholic liver disease.


Subject(s)
Endotoxemia/metabolism , Gastroenterology/trends , Intestinal Absorption , Liver Diseases, Alcoholic/metabolism , Animals , Humans , Permeability
14.
Alcohol Clin Exp Res ; 28(5): 797-804, 2004 May.
Article in English | MEDLINE | ID: mdl-15166657

ABSTRACT

BACKGROUND: Intestinal permeability and endotoxemia play a crucial role in the pathogenesis of alcoholic liver disease. Previous studies showed that acetaldehyde disrupts intestinal epithelial barrier function and increases paracellular permeability by a tyrosine kinase-dependent mechanism. In the present study, the role of epidermal growth factor (EGF) in protection of epithelial barrier function from acetaldehyde was evaluated in Caco-2 intestinal epithelial cell monolayer. METHODS: Caco-2 cells on Transwell inserts were exposed to acetaldehyde in the absence or presence of EGF, and the paracellular permeability was evaluated by measuring transepithelial electrical resistance and unidirectional flux of inulin. Integrity of epithelial tight junctions and adherens junctions was analyzed by confocal immunofluorescence microscopy and immunoblot analysis of occludin, zonula occludens (ZO)-1, E-cadherin, and beta-catenin in the actin cytoskeleton. Reorganization of actin cytoskeletal architecture was examined by confocal microscopy. RESULTS: Acetaldehyde increased paracellular permeability to inulin and lipopolysaccharide, and EGF significantly reduced these effects of acetaldehyde in a time- and dose-dependent manner. EGF prevented acetaldehyde-induced reorganization of occludin, ZO-1, E-cadherin, and beta-catenin from the cellular junctions to the intracellular compartments. Acetaldehyde treatment induced a reorganization of actin cytoskeletal network and reduced the levels of occludin, ZO-1, E-cadherin, and beta-catenin associated with the actin cytoskeleton. EGF effectively prevented acetaldehyde-induced reorganization of actin cytoskeleton and the interaction of occludin, ZO-1, E-cadherin, and beta-catenin with the actin cytoskeleton. CONCLUSION: These results indicate that EGF attenuates acetaldehyde-induced disruption of tight junctions and adherens junctions and prevents acetaldehyde-induced reorganization of actin cytoskeleton and its interaction with occludin, ZO-1, E-cadherin, and beta-catenin.


Subject(s)
Acetaldehyde/antagonists & inhibitors , Acetaldehyde/pharmacology , Cell Membrane Permeability/physiology , Epidermal Growth Factor/pharmacology , Intestinal Absorption/physiology , Caco-2 Cells , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Humans , Intestinal Absorption/drug effects
15.
Biochem Biophys Res Commun ; 302(2): 324-9, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12604349

ABSTRACT

Occludin, the transmembrane integral protein of the tight junction, plays a crucial role in the molecular organization and function of tight junction. While the homotypic interaction of extracellular loops of occludin appears to determine the barrier function of tight junction, the intracellular C-terminal tail, C-occludin, interacts with other tight junction proteins such as ZO-1, ZO-2, and ZO-3 and with the actin filaments of cytoskeleton. In the present study we phosphorylated GST-fused C-occludin on tyrosine residues, in TKX1 Epicurian coli or by active c-Src in vitro. c-Src binds to occludin and phosphorylates it on tyrosine residues. The effect of tyrosine phosphorylation of C-occludin on its ability to bind ZO-1, ZO-2, ZO-3, and F-actin was evaluated. Results show that the amounts of ZO-1, ZO-2, and ZO-3 bound to tyrosine phosphorylated C-occludin were several fold less than the amounts bound to non-phosphorylated C-occludin. However, the amount of tyrosine phosphorylated C-occludin bound to F-actin was not significantly different from the amount of non-phosphorylated C-occludin bound to F-actin. These results demonstrate that tyrosine phosphorylation of occludin reduces its ability to bind ZO-1, ZO-2, and ZO-3, but not F-actin. Results also suggest that c-Src-mediated disruption of tight junction may involve tyrosine phosphorylation of occludin.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Tyrosine/metabolism , Actins/metabolism , Binding Sites , Caco-2 Cells , Humans , Occludin , Phosphorylation , Zonula Occludens Proteins , Zonula Occludens-1 Protein , Zonula Occludens-2 Protein
16.
Biochem J ; 368(Pt 1): 183-90, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12188924

ABSTRACT

We have studied the role of nuclear factor of activated T-cells (NFAT) transcription factors in the induction of vascular smooth muscle cell (VSMC) growth by platelet-derived growth factor-BB (PDGF-BB) and thrombin, the receptor tyrosine kinase (RTK) and G-protein-coupled receptor (GPCR) agonists, respectively. NFATc1 but not NFATc2 or NFATc3 was translocated from the cytoplasm to the nucleus upon treatment of VSMCs with PDGF-BB or thrombin. Translocation of NFATc1 was followed by an increase in NFAT-DNA binding activity and NFAT-dependent reporter gene expression. Cyclosporin A (CsA), a potent and specific inhibitor of calcineurin, a calcium/calmodulin-dependent serine phosphatase involved in the dephosphorylation and activation of NFATs, blocked NFAT-DNA binding activity and NFAT-dependent reporter gene expression induced by PDGF-BB and thrombin. CsA also completely inhibited PDGF-BB- and thrombin-induced VSMC growth, as measured by DNA synthesis and cell number. In addition, forced expression of the NFAT-competing peptide VIVIT for calcineurin binding significantly attenuated the DNA synthesis induced by PDGF-BB and thrombin in VSMCs. Together, these findings for the first time demonstrate a role for NFATs in RTK and GPCR agonist-induced growth in VSMCs.


Subject(s)
DNA-Binding Proteins/physiology , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins , Platelet-Derived Growth Factor/pharmacology , Thrombin/pharmacology , Transcription Factors/physiology , Animals , Becaplermin , Cell Division/drug effects , Cell Division/physiology , DNA/biosynthesis , DNA/drug effects , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/agonists
17.
Biochem Biophys Res Commun ; 293(1): 610-6, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-12054646

ABSTRACT

The regulation of protein phosphatase 2A (PP2A) and protein threonine phosphorylation by H(2)O(2) was determined in Caco-2 cell monolayer. Incubation with H(2)O(2) (20 microM) resulted in threonine phosphorylation of a cluster of proteins at the molecular mass range of 170-250 kDa. PKC activity and plasma membrane localization of several isoforms of PKC were not affected by H(2)O(2). However, H(2)O(2) reduced 80-85% of okadaic acid-sensitive protein phosphatase activity. Immunocomplex protein phosphatase assay demonstrated that H(2)O(2) reduced the activity of PP2A, but not that of PP2C or PP1. Oxidized glutathione inhibited PP2A activity in plasma membranes prepared from Caco-2 cells and the phosphatase activity of an isolated PP2A. PP2A activity was also inhibited by N-ethylmaleimide, iodoacetamide, and p-chloromercuribenzoate. Inhibition of PP2A by oxidized glutathione was reversed by reduced glutathione. Glutathione also restored the PP2A activity in plasma membranes isolated from H(2)O(2)-treated Caco-2 cell monolayer. These results indicate that PP2A activity can be regulated by glutathionylation, and that H(2)O(2) inhibits PP2A in Caco-2 cells, which may involve glutathionylation of PP2A.


Subject(s)
Glutathione/pharmacology , Hydrogen Peroxide/pharmacology , Phosphoprotein Phosphatases/metabolism , Cell Membrane/enzymology , Glutathione Disulfide/pharmacology , Humans , Isoenzymes/metabolism , Kinetics , Phosphorylation , Phosphothreonine/metabolism , Protein Kinase C/metabolism , Protein Phosphatase 2 , Tumor Cells, Cultured
18.
Auton Neurosci ; 88(1-2): 99-108, 2001 Apr 12.
Article in English | MEDLINE | ID: mdl-11474552

ABSTRACT

In this study, we investigated measures of nonlinear dynamics and chaos of heart rate time series in 30 normal control subjects and 36 age-matched patients with panic disorder in supine and standing postures. We obtained minimum embedding dimension (MED), largest Lyapunov exponent (LLE) and measures of nonlinearity (NL) of heart rate time series. MED quantifies system's complexity, LLE predictability and NL, deviation from linear processes. There was a significant increase in complexity (p < 0.00001), an increase in predictability (decreased chaos) (p < 0.00001) and an increase in nonlinearity (Snet GS) (p = 0.00001), especially in supine posture in patients with panic disorder. Increased NL score in supine posture may be due to a relative increase in cardiac sympathetic activity and an overall decrease in LLE may indicate an impaired cardiac autonomic flexibility in these patients due possibly to a decrease in cardiac vagal activity. These findings may further explain the reported higher incidence of cardiovascular mortality in patients with anxiety disorders.


Subject(s)
Heart Rate , Models, Cardiovascular , Nonlinear Dynamics , Panic Disorder/physiopathology , Adult , Female , Humans , Male , Reference Values , Supine Position
19.
Am J Physiol Gastrointest Liver Physiol ; 280(6): G1280-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352822

ABSTRACT

Acetaldehyde-induced cytotoxicity is an important factor in pathogenesis of alcohol-related diseases; however, the mechanism of this toxicity is unknown. We recently showed that acetaldehyde increases epithelial paracellular permeability. We asked whether protein tyrosine phosphorylation via modulation of tyrosine kinases and/or PTPases is a mechanism involved in acetaldehyde-induced disruption of the tight junctions in the Caco-2 cell monolayer. Immunofluorescence localization of occludin and ZO-1 showed disruption of the tight junctions in acetaldehyde-treated cell monolayer. Administration of genistein prevented acetaldehyde-induced permeability. Acetaldehyde increased tyrosine phosphorylation of three clusters of proteins with molecular masses of 30-50, 60-90, and 110-150 kDa; three of these proteins were ZO-1, E-cadherin, and beta-catenin. Acetaldehyde reduced PTPase activity in plasma membrane and soluble fractions, whereas tyrosine kinase activity remained unaffected. Treatment with acetaldehyde resulted in a 97% loss of protein tyrosine phosphatase (PTP)1B activity and a partial reduction of PTP1C and PTP1D activities. These results strongly suggest that acetaldehyde inhibits PTPases to increase protein tyrosine phosphorylation, which may result in disruption of the tight junctions.


Subject(s)
Acetaldehyde/pharmacology , Epithelium/drug effects , Tight Junctions/drug effects , Trans-Activators , Tyrosine/metabolism , Caco-2 Cells , Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , Epithelium/metabolism , Genistein/pharmacology , Humans , Membrane Proteins/metabolism , Permeability , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein , beta Catenin
20.
Hum Mol Genet ; 10(9): 919-26, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309365

ABSTRACT

Parkinson's disease (PD) is a common progressive neurodegenerative disorder caused by the loss of dopaminergic neurons in the substantia nigra. Although mutations in alpha-synuclein have been identified in autosomal dominant PD, the mechanism by which dopaminergic neural cell death occurs remains unknown. Proteins encoded by two other genes in which mutations cause familial PD, parkin and UCH-L1, are involved in regulation of the ubiquitin-proteasome pathway, suggesting that dysregulation of the ubiquitin-proteasome pathway is involved in the mechanism by which these mutations cause PD. We established inducible PC12 cell lines in which wild-type or mutant alpha-synuclein can be de-repressed by removing doxycycline. Differentiated PC12 cell lines expressing mutant alpha-synuclein showed decreased activity of proteasomes without direct toxicity. Cells expressing mutant alpha-synuclein showed increased sensitivity to apoptotic cell death when treated with sub-toxic concentrations of an exogenous proteasome inhibitor. Apoptosis was accompanied by mitochondrial depolarization and elevation of caspase-3 and -9, and was blocked by cyclosporin A. These data suggest that expression of mutant alpha-synuclein results in sensitivity to impairment of proteasome activity, leading to mitochondrial abnormalities and neuronal cell death.


Subject(s)
Acetylcysteine/analogs & derivatives , Apoptosis , Cysteine Endopeptidases/metabolism , Mitochondria/physiology , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/physiology , PC12 Cells/physiology , Acetylcysteine/pharmacology , Animals , Blotting, Western , Caspase 3 , Caspase 9 , Caspases/metabolism , Cyclosporine/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Gene Expression Regulation , Immunoenzyme Techniques , Membrane Potentials , Mutation , Nerve Tissue Proteins/genetics , PC12 Cells/drug effects , Parkinson Disease/pathology , Proteasome Endopeptidase Complex , Rats , Synucleins , Tetracycline/pharmacology , Transfection , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...