Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters








Database
Language
Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 298(2): G167-76, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19926820

ABSTRACT

The glutamic acid derivative thalidomide is a transcriptional inhibitor of TNF-alpha but is also known to affect human blood vessels, which may underlie its teratogenicity. Thalidomide has been used in the treatment of refractory Crohn's disease (CD), but the therapeutic mechanism is not defined. We examined the effect of thalidomide on primary cultures of human intestinal microvascular endothelial cells (HIMEC), the relevant endothelial cell population in inflammatory bowel disease (IBD), to determine its effect on endothelial activation, leukocyte interaction, and VEGF-induced angiogenesis. HIMEC cultures were pretreated with thalidomide before activation with either TNF-alpha/LPS or VEGF. A low-shear-stress flow adhesion assay with either U-937 or whole blood was used to assess HIMEC activation following TNF-alpha/LPS, and a Wright's stain identified adherent leukocytes. Expression of cell adhesion molecules (E-selectin, intercellular adhesion molecule-1, vascular cell adhesion molecule-1) was assessed using radioimmunoassay. Effects of thalidomide on NF-kappaB activation, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) expression in TNF-alpha/LPS-activated HIMEC were determined by RT-PCR and Western blotting. Thalidomide blocked adhesion of both U-937 and whole blood leukocytes by 50% in HIMEC, inhibiting binding of all classes of leukocytes. Thalidomide also blocked NF-kappaB and cell adhesion molecule expression in HIMEC. In marked contrast, thalidomide did not affect either iNOS or COX-2 expression, two key molecules that play a role in the downregulation of HIMEC activation. VEGF-induced HIMEC transmigration, growth, proliferation, tube formation, and Akt phosphorylation were significantly inhibited by thalidomide. In summary, thalidomide exerted a potent effect on HIMEC growth and activation, suggesting that it may also function via an endothelial mechanism in the treatment of CD.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Crohn Disease/drug therapy , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Thalidomide/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Crohn Disease/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , E-Selectin/metabolism , Endothelial Cells/cytology , Humans , Intercellular Adhesion Molecule-1/metabolism , Intestines/blood supply , Leukocytes/cytology , Lipopolysaccharides/pharmacology , Microvessels/cytology , NF-kappa B/metabolism , Neovascularization, Pathologic/pathology , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/pharmacology
2.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G259-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19520742

ABSTRACT

Endothelial activation and surface expression of cell adhesion molecules (CAMs) is critical for binding and recruitment of circulating leukocytes in tissues during the inflammatory response. Endothelial CAM expression plays a critical role in the intestinal microvasculature in inflammatory bowel disease (IBD), as blockade of leukocyte alpha4-integrin binding by gut endothelial CAM ligands has therapeutic benefit in IBD. Mechanisms underlying expression of vascular cell adhesion molecule (VCAM)-1, a ligand for alpha4-integrin in primary cultures of human intestinal microvascular endothelial cells (HIMEC) has not been defined. We investigated the effect of curcumin, phosphatidylinositol 3-kinase (PI 3-kinase)/protein kinase B (Akt), and mitogen-activated protein kinase (MAPK) inhibitors on VCAM-1 expression and function in HIMEC. CAM expression was assessed and HIMEC-leukocyte adhesion was visualized under static and flow conditions. Western blotting and in vitro kinase assays were used to assess Akt and MAPK activation. Nuclear factor-kappaB (NF-kappaB) activation and nuclear translocation of its p65 subunit were determined. Tumor necrosis factor (TNF)-alpha/lipopolysaccharide (LPS)-induced VCAM-1 expression in HIMEC was suppressed by Akt small-interfering RNA, curcumin, and inhibitors of NF-kappaB (SN-50), p38 MAPK (SB-203580) and PI 3-kinase/Akt (LY-294002). VCAM-1 induction was partially suppressed by p44/42 MAPK (PD-098059) but unaffected by c-Jun NH2-terminal kinase (SP-600125) inhibition. Curcumin inhibited Akt/MAPK/NF-kappaB activity and prevented nuclear translocation of the p65 NF-kappaB subunit following TNF-alpha/LPS. At physiological shear stress, curcumin attenuated leukocyte adhesion to TNF-alpha/LPS-activated HIMEC monolayers. In conclusion, curcumin inhibited the expression of VCAM-1 in HIMECs through blockade of Akt, p38 MAPK, and NF-kappaB. Curcumin may represent a novel therapeutic agent targeting endothelial activation in IBD.


Subject(s)
Curcumin/pharmacology , Endothelial Cells/drug effects , Gastrointestinal Agents/pharmacology , Intestines/blood supply , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus , Cell Adhesion/drug effects , Cell Adhesion Molecules , Cells, Cultured , Endothelial Cells/enzymology , Humans , Immunoglobulins/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Microvessels/drug effects , Microvessels/enzymology , Mucoproteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Signal Transduction/drug effects , Time Factors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G388-98, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074641

ABSTRACT

Human esophageal epithelial cells play a key role in esophageal inflammation in response to acidic pH during gastroesophageal reflux disease (GERD), increasing secretion of IL-6 and IL-8. The mechanisms underlying IL-6 and IL-8 expression and secretion in esophageal epithelial cells after acid stimulation are not well characterized. We investigated the role of PKC, MAPK, and NF-kappaB signaling pathways and transcriptional regulation of IL-6 and IL-8 expression in HET-1A cells exposed to acid. Exposure of HET-1A cells to pH 4.5 induced NF-kappaB activity and enhanced IL-6 and IL-8 secretion and mRNA and protein expression. Acid stimulation of HET-1A cells also resulted in activation of MAPKs and PKC (alpha and epsilon). Curcumin, as well as inhibitors of NF-kappaB (SN-50), PKC (chelerythrine), and p44/42 MAPK (PD-098059) abolished the acid-induced expression of IL-6 and IL-8. The JNK inhibitor SP-600125 blocked expression/secretion of IL-6 but only partially attenuated IL-8 expression. The p38 MAPK inhibitor SB-203580 did not inhibit IL-6 expression but exerted a stronger inhibitory effect on IL-8 expression. Together, these data demonstrate that 1) acid is a potent inducer of IL-6 and IL-8 production in HET-1A cells; 2) MAPK and PKC signaling play a key regulatory role in acid-mediated IL-6 and IL-8 expression via NF-kappaB activation; and 3) the anti-inflammatory plant compound curcumin inhibits esophageal activation in response to acid. Thus IL-6 and IL-8 expression by acid may contribute to the pathobiology of mucosal injury in GERD, and inhibition of the NF-kappaB/proinflammatory cytokine pathways may emerge as important therapeutic targets for treatment of esophageal inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Curcumin/pharmacology , Epithelial Cells/drug effects , Esophagus/drug effects , Interleukin-6/metabolism , Interleukin-8/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Anthracenes/pharmacology , Benzophenanthridines/pharmacology , Cell Line , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/immunology , Esophagus/enzymology , Esophagus/immunology , Flavonoids/pharmacology , Humans , Hydrogen-Ion Concentration , Imidazoles/pharmacology , Interleukin-6/genetics , Interleukin-8/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mucous Membrane/drug effects , Mucous Membrane/enzymology , Mucous Membrane/immunology , NF-kappa B/antagonists & inhibitors , Peptides/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Telomerase/metabolism , Time Factors , Transcription, Genetic/drug effects , Up-Regulation
4.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G581-90, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18635600

ABSTRACT

Tissue remodeling and mesenchymal cell accumulation accompanies chronic inflammatory disorders involving joints, lung, vasculature, and bowel. Chronic inflammation may alter DNA-mismatch repair (MMR) systems in mesenchymal cells, but is not defined in Crohn's disease (CD) and its associated intestinal remodeling and stricture formation. We determined whether DNA-MMR alteration plays a role in the pathogenesis of CD tissue remodeling. Control and CD bowel tissues were used to generate primary cultures of muscularis mucosa myofibroblasts, which were assessed directly or following stimulation with TNF-alpha/LPS or H2O2. MutS homolog (MSH)2, MSH3, and MSH6 expression in tissues and myofibroblasts was determined. Immunohistochemical staining revealed an increased expression of MSH2 in CD muscularis mucosa and submucosal tissues compared with controls or uninvolved CD tissue, and MSH2 expression was increased in CD myofibroblasts compared with control cells. TNF-alpha/LPS and H2O2 further enhanced MSH2 expression in both control and CD cells, which were decreased by simvastatin. There were no significant changes in MSH3 and MSH6 expression. Proliferating cell nuclear antigen and Ki67 staining of CD tissue revealed increased proliferation in the muscularis mucosa and submucosa of chronically inflamed tissues, and enhanced proliferation was seen in CD myofibroblasts compared with controls. Simvastatin reversed the effects of inflammatory stress on the DNA-MMR and inhibited proliferation of control and CD myofibroblasts. Gene silencing with MSH2 siRNA selectively decreased CD myofibroblast proliferation. These data demonstrate a potential role for MSH2 in the pathogenesis of nonneoplastic mesenchymal cell accumulation and intestinal remodeling in CD chronic inflammation.


Subject(s)
Cell Proliferation , Crohn Disease/enzymology , Fibroblasts/enzymology , Intestinal Obstruction/etiology , Intestines/enzymology , MutS Homolog 2 Protein/metabolism , Adult , Cell Proliferation/drug effects , Cells, Cultured , Crohn Disease/complications , Crohn Disease/genetics , Crohn Disease/pathology , DNA-Binding Proteins/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fluorescent Antibody Technique , Humans , Hydrogen Peroxide/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunohistochemistry , Intestinal Obstruction/enzymology , Intestinal Obstruction/genetics , Intestinal Obstruction/pathology , Intestines/drug effects , Intestines/pathology , Lipopolysaccharides/pharmacology , Male , Microsatellite Instability , Middle Aged , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Simvastatin/pharmacology , Thymidine/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
5.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1323-36, 2007 May.
Article in English | MEDLINE | ID: mdl-17218473

ABSTRACT

Nitric oxide (.NO) generation from conversion of l-arginine to citrulline by nitric oxide synthase isoforms plays a critical role in vascular homeostasis. Loss of .NO is linked to vascular pathophysiology and is decreased in chronically inflamed gut blood vessels in inflammatory bowel disease (IBD; Crohn's disease and ulcerative colitis). Mechanisms underlying decreased .NO production in IBD gut microvessels are not fully characterized. Loss of .NO generation may result from increased arginase (AR) activity, which enzymatically competes with nitric oxide synthase for the common substrate l-arginine. We characterized AR expression in IBD microvessels and endothelial cells and its contribution to decreased .NO production. AR expression was assessed in resected gut tissues and human intestinal microvascular endothelial cells (HIMEC). AR expression significantly increased in both ulcerative colitis and Crohn's disease microvessels and submucosal tissues compared with normal. TNF-alpha/lipopolysaccharide increased AR activity, mRNA and protein expression in HIMEC in a time-dependent fashion. RhoA/ROCK pathway, a negative regulator of .NO generation in endothelial cells, was examined. The RhoA inhibitor C3 exoenzyme and the ROCK inhibitor Y-27632 both attenuated TNF-alpha/lipopolysaccharide-induced MAPK activation and blocked AR expression in HIMEC. A significantly higher AR activity and increased RhoA activity were observed in IBD submucosal tissues surrounding microvessels compared with normal control gut tissue. Functionally, inhibition of AR activity decreased leukocyte binding to HIMEC in an adhesion assay. Loss of .NO production in IBD microvessels is linked to enhanced levels of AR in intestinal endothelial cells exposed to chronic inflammation in vivo.


Subject(s)
Arginase/metabolism , Endothelium/physiopathology , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/enzymology , ADP Ribose Transferases/pharmacology , Amides/pharmacology , Arginine/pharmacology , Botulinum Toxins/pharmacology , Cell Adhesion Molecules/physiology , Crohn Disease/physiopathology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intracellular Signaling Peptides and Proteins/physiology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/physiology , Nitric Oxide/biosynthesis , Protein Serine-Threonine Kinases/physiology , Pyridines/pharmacology , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Valine/pharmacology , rho-Associated Kinases , rhoA GTP-Binding Protein/physiology
6.
Am J Physiol Cell Physiol ; 291(5): C931-45, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16790501

ABSTRACT

The heat shock response maintains cellular homeostasis following sublethal injury. Heat shock proteins (Hsps) are induced by thermal, oxyradical, and inflammatory stress, and they chaperone denatured intracellular proteins. Hsps also chaperone signal transduction proteins, modulating signaling cascades during repeated stress. Gastroesophageal reflux disease (GERD) affects 7% of the US population, and it is linked to prolonged esophageal acid exposure. GERD is characterized by enhanced and selective leukocyte recruitment from esophageal microvasculature, implying activation of microvascular endothelium. We investigated whether phosphatidylinositol 3-kinase (PI3K)/Akt and MAPK regulate Hsp induction in primary cultures of human esophageal microvascular endothelial cells (HEMEC) in response to acid exposure (pH 4.5). Inhibitors of signaling pathways were used to define the contribution of PI3K/Akt and MAPKs in the heat shock response and following acid exposure. Acid significantly enhanced phosphorylation of Akt and MAPKs in HEMEC as well as inducing Hsp27 and Hsp70. The PI3K inhibitor LY-294002, and Akt small interfering RNA inhibited Akt activation and Hsp70 expression in HEMEC. The p38 MAPK inhibitor (SB-203580) and p38 MAPK siRNA blocked Hsp27 and Hsp70 mRNA induction, suggesting a role for MAPKs in the HEMEC heat shock response. Thus acidic pH exposure protects HEMEC through induction of Hsps and activation of MAPK and PI3 kinase pathway. Acidic exposure increased HEMEC expression of VCAM-1 protein, but not ICAM-1, which may contribute to selective leukocyte (i.e., eosinophil) recruitment in esophagitis. Activation of esophageal endothelial cells exposed to acidic refluxate may contribute to GERD in the setting of a disturbed mucosal squamous epithelial barrier (i.e., erosive esophagitis, peptic ulceration).


Subject(s)
Endothelial Cells/metabolism , Esophagus/blood supply , HSP110 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Survival/physiology , Class I Phosphatidylinositol 3-Kinases , Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Esophagus/cytology , Gene Expression Regulation/genetics , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HSP110 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Humans , Hydrogen-Ion Concentration , Molecular Chaperones , Neoplasm Proteins/genetics , Phosphorylation , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Signal Transduction/physiology , Swine , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL