Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Cell ; 24(19): 3056-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23924897

ABSTRACT

In vitro and in vivo studies implicate occludin in the regulation of paracellular macromolecular flux at steady state and in response to tumor necrosis factor (TNF). To define the roles of occludin in these processes, we established intestinal epithelia with stable occludin knockdown. Knockdown monolayers had markedly enhanced tight junction permeability to large molecules that could be modeled by size-selective channels with radii of ~62.5 Å. TNF increased paracellular flux of large molecules in occludin-sufficient, but not occludin-deficient, monolayers. Complementation using full-length or C-terminal coiled-coil occludin/ELL domain (OCEL)-deficient enhanced green fluorescent protein (EGFP)-occludin showed that TNF-induced occludin endocytosis and barrier regulation both required the OCEL domain. Either TNF treatment or OCEL deletion accelerated EGFP-occludin fluorescence recovery after photobleaching, but TNF treatment did not affect behavior of EGFP-occludin(ΔOCEL). Further, the free OCEL domain prevented TNF-induced acceleration of occludin fluorescence recovery, occludin endocytosis, and barrier loss. OCEL mutated within a recently proposed ZO-1-binding domain (K433) could not inhibit TNF effects, but OCEL mutated within the ZO-1 SH3-GuK-binding region (K485/K488) remained functional. We conclude that OCEL-mediated occludin interactions are essential for limiting paracellular macromolecular flux. Moreover, our data implicate interactions mediated by the OCEL K433 region as an effector of TNF-induced barrier regulation.


Subject(s)
Intestinal Mucosa/metabolism , Occludin/metabolism , Tight Junctions/genetics , Tumor Necrosis Factor-alpha/metabolism , Caco-2 Cells , Endocytosis/genetics , Gene Knockdown Techniques , Humans , Occludin/chemistry , Occludin/genetics , Permeability , Protein Binding , Protein Structure, Tertiary/genetics , Sequence Deletion , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
2.
Gastroenterology ; 145(2): 407-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23619146

ABSTRACT

BACKGROUND & AIMS: Tight junction dysregulation and epithelial damage contribute to barrier loss in patients with inflammatory bowel disease. However, the mechanisms that regulate these processes and their relative contributions to disease pathogenesis are not completely understood. We investigated these processes using colitis models in mice. METHODS: We induced colitis by adoptive transfer of CD4(+)CD45RB(hi) cells or administration of dextran sulfate sodium to mice, including those deficient in tumor necrosis factor receptor (TNFR) 1, TNFR2, or the long isoform of myosin light chain kinase (MLCK). Intestinal tissues and isolated epithelial cells were analyzed by immunoblot, immunofluorescence, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction assays. RESULTS: Induction of immune-mediated colitis by CD4(+)CD45RB(hi) adoptive transfer increased intestinal permeability, epithelial expression of claudin-2, the long isoform of MLCK, and TNFR2 (but not TNFR1) and phosphorylation of the myosin II light chain. Long MLCK upregulation, myosin II light chain phosphorylation, barrier loss, and weight loss were attenuated in TNFR2(-/-) , but not TNFR1(-/-) , recipients of wild-type CD4(+)CD45RB(hi) cells. Similarly, long MLCK(-/-) mice had limited increases in myosin II light chain phosphorylation, claudin-2 expression, and intestinal permeability and delayed onset of adoptive transfer-induced colitis. However, coincident with onset of epithelial apoptosis, long MLCK(-/-) mice ultimately developed colitis. This indicates that disease progresses via apoptosis in the absence of MLCK-dependent tight junction regulation. In support of this conclusion, long MLCK(-/-) mice were not protected from epithelial apoptosis-mediated, damage-dependent dextran sulfate sodium colitis. CONCLUSIONS: In immune-mediated inflammatory bowel disease models, TNFR2 signaling increases long MLCK expression, resulting in tight junction dysregulation, barrier loss, and induction of colitis. At advanced stages, colitis progresses by apoptosis and mucosal damage that result in tight junction- and MLCK-independent barrier loss. Therefore, barrier loss in immune-mediated colitis occurs via two temporally and morphologically distinct mechanisms. Differential targeting of these mechanisms can lead to improved inflammatory bowel disease therapies.


Subject(s)
Colitis/physiopathology , Epithelial Cells/physiology , Intestinal Mucosa/physiopathology , Myosin-Light-Chain Kinase/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tight Junctions/physiology , Adoptive Transfer , Animals , Apoptosis/physiology , Claudin-2/metabolism , Colitis/immunology , Colitis/metabolism , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Inflammatory Bowel Diseases , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Myosin-Light-Chain Kinase/genetics , Permeability , Phosphorylation , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction , Tight Junctions/metabolism , Up-Regulation , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...