Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Neuroscience ; 147(3): 664-73, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17560040

ABSTRACT

Multiple sclerosis (MS) is a disease of the CNS in which inflammation, demyelination and neurodegeneration contribute to its initiation and progression. A frequently employed model of MS is experimental autoimmune encephalomyelitis (EAE). Here, to gain new insights into the disease process, an analysis of proteins in extracts of lumbar spinal cord from naïve and EAE rats was undertaken. The data mainly confirm that inflammation and blood-brain barrier (BBB) breakdown are the major hallmarks of disease in this model. Given their importance in the BBB, junctional proteins were further investigated. Occludin, a protein localizing to tight junctions in brain endothelial cells, showed strikingly increased migration in EAE when analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). This increased migration was mimicked by in vitro phosphatase treatment, implying its dephosphorylation in EAE. Occludin dephosphorylation coincided with the onset of inflammation, slightly preceding visible signs of disease, and was just prior to apparent changes in BBB permeability. These findings suggest occludin is a target for signaling processes in EAE, perhaps regulating the response of the BBB to the inflammatory environment as seen in MS.


Subject(s)
Encephalitis/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Membrane Proteins/metabolism , Animals , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , Encephalitis/etiology , Encephalitis/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Endothelial Cells/cytology , Female , Immunoprecipitation/methods , Mass Spectrometry/methods , Occludin , Phosphoric Monoester Hydrolases/pharmacology , Phosphorylation/drug effects , Rats , Rats, Inbred Lew , Spinal Cord/pathology , Tight Junctions/metabolism
2.
J Biol Chem ; 276(13): 10423-31, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11139571

ABSTRACT

In epithelial and endothelial cells, tight junctions regulate the paracellular permeability of ions and proteins. Disruption of tight junctions by inflammation is often associated with tissue edema, but regulatory mechanisms are not fully understood. Using ECV304 cells as a model system, lysophosphatidic acid and histamine were found to increase the paracellular permeability of the tracer horseradish peroxidase. Cytoskeletal changes induced by these agents included stimulation of stress fiber formation and myosin light chain phosphorylation. Additionally, occludin, a tight junction protein, was a target for signaling events triggered by lysophosphatidic acid and histamine, events that resulted in its phosphorylation. A dominant-negative mutant of RhoA, RhoA T19N, or a specific inhibitor of Rho-activated kinases, Y-27632, prevented stress fiber formation, myosin light chain phosphorylation, occludin phosphorylation, and the increase in tracer flux in response to lysophosphatidic acid. In contrast, although RhoA T19N and Y-27632 blocked the cytoskeletal events induced by histamine, they had no effect on the stimulation of occludin phosphorylation or increased tracer flux, indicating that occludin phosphorylation may regulate tight junction permeability independently of cytoskeletal events. Thus, occludin is a target for receptor-initiated signaling events regulating its phosphorylation, and this phosphorylation may be a key regulator of tight junction permeability.


Subject(s)
Membrane Proteins/metabolism , Tight Junctions/physiology , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Adenoviridae/genetics , Cell Line , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Gel, Two-Dimensional , Gene Transfer Techniques , Genes, Dominant , Histamine/pharmacology , Horseradish Peroxidase/metabolism , Humans , Lysophospholipids/pharmacology , Microscopy, Fluorescence , Nucleic Acid Synthesis Inhibitors/pharmacology , Occludin , Permeability , Phosphorylation , Precipitin Tests , Signal Transduction , Stress Fibers/metabolism , Time Factors
3.
J Immunol ; 164(12): 6543-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843713

ABSTRACT

During Gram-negative sepsis bacterial LPS induces endothelial cell contraction, actin reorganization, and loss of endothelial integrity by an unknown signal mechanism. In this study, we provide evidence that LPS-stimulation of endothelial cells (HUVEC) decreases myosin light chain (MLC) phosphatase, resulting in an increase in MLC phosphorylation followed by cell contraction. All of these LPS effects could be blocked by the Rho-GTPase inhibitor C3 transferase from Clostridium botulinum or the Rho kinase inhibitor Y-27632. These data suggest that LPS induces MLC phosphorylation via Rho/Rho kinase-mediated inhibition of MLC phosphatase in HUVEC. Furthermore, we observed that cAMP-elevating drugs, known to exert a vasoprotective function, mimicked the effects of C3 transferase and Y-27632, i.e., inhibited LPS-induced MLC phosphatase inactivation and MLC phosphorylation. cAMP elevation did not inhibit myosin phosphorylation induced by constitutively active V14Rho or the MLC phosphatase inhibitor calyculin and did not induce phosphorylation of RhoA in HUVEC, indicating inhibition of an upstream regulator of Rho/Rho kinase. Taken together, Rho/Rho kinase appears to be a central target for inflammatory mediators causing endothelial cell contraction such as bacterial toxins, but also for vasoprotective molecules elevating intracellular cAMP.


Subject(s)
Cyclic AMP/physiology , Endothelium, Vascular/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/immunology , Myosin Light Chains/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/immunology , rho GTP-Binding Proteins/antagonists & inhibitors , 1-Methyl-3-isobutylxanthine/pharmacology , Cells, Cultured , Cyclic AMP/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Enzyme Activation , Enzyme Inhibitors/immunology , Humans , Intracellular Signaling Peptides and Proteins , Marine Toxins , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase , Oxazoles/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/physiology , Protein Synthesis Inhibitors/pharmacology , Transcription, Genetic/drug effects , Umbilical Veins , rho GTP-Binding Proteins/physiology , rho-Associated Kinases
4.
Biochem J ; 346 Pt 1: 209-16, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657259

ABSTRACT

Vascular endothelial growth factor (VEGF) is an endothelium-specific mitogen that induces angiogenesis and increases vascular permeability. These processes involve regulation of cell-cell adhesion, but molecular mechanisms have yet to be fully established. p120, also termed p120(ctn), and its variant p100 are catenins which associate with cadherins and localize to adherens junctions. VEGF was reported to stimulate tyrosine phosphorylation of catenins in endothelial cells. In contrast, we have found that VEGF potently stimulated a rapid and dose-dependent decrease in serine/threonine phosphorylation of p120 and p100. VEGF acted via VEGF receptor 2 to achieve this effect which was independent of activation of the extracellular-signal-regulated kinase pathway. Histamine and activators of protein kinase C had a very similar effect to that of VEGF on phosphorylation of p120 and p100, suggesting that these diverse stimuli may converge on a common signalling element regulating p120/p100 serine/threonine phosphorylation. These data raise the possibility that the dephosphorylation of p120 and p100 triggered by VEGF may contribute to mechanisms regulating permeability and/or motility through modulation of cadherin adhesiveness.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Cadherins/metabolism , Catenins , Cell Adhesion Molecules/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Genetic Variation/genetics , Histidine/pharmacology , Humans , Kinetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Weight , Peptide Mapping , Phorbol 12,13-Dibutyrate/pharmacology , Phosphoamino Acids/metabolism , Phosphoproteins/genetics , Phosphorylation/drug effects , Precipitin Tests , Protein Kinase C/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Delta Catenin
5.
Anal Biochem ; 275(2): 148-55, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10552898

ABSTRACT

Cyclic nucleotides are key regulators of many cellular processes. Their immediate action is terminated through the activity of phosphodiesterases, a diverse family of enzymes. This diversity has given rise to drug discovery opportunities, and assay technology is therefore of key importance. Inhibitors of the cyclic-AMP-specific phosphodiesterases (the PDE4 family) are drug candidates for a variety of inflammatory disorders. However, PDE4 inhibitors, besides their immunomodulatory effects, also cause side effects including nausea and emesis. Recently, it has been suggested that PDE4 exists in two different conformations with respect to inhibition by the prototypical compound rolipram. Inhibition of the low-affinity conformer is thought to give rise to anti-inflammatory effects, and inhibition of the high-affinity conformer to side effects. Therefore, a selective inhibitor of the low-affinity conformer may have clinical utility. Methods are described to prepare recombinant forms of PDE4B that allow screening for compounds that could preferentially inhibit the low-affinity conformer. Furthermore, conditions for an efficient, scintillation proximity, microtiter plate-based assay are described, providing a considerable advance over previous assays in terms of throughput and automatability.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Phosphodiesterase Inhibitors/analysis , Rolipram/analysis , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Automation , Base Sequence , Cyclic Nucleotide Phosphodiesterases, Type 4 , DNA Primers , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Humans , Phosphodiesterase Inhibitors/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Rolipram/pharmacology
6.
J Neuroimmunol ; 97(1-2): 119-28, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10408965

ABSTRACT

We examined the treatment effects of two structurally distinct phosphodiesterase type IV (PDE IV) inhibitors, BBB022 and rolipram, in murine and rat models of experimental autoimmune encephalomyelitis (EAE). Based on our data, we propose a mechanism of action which may supplement immunomodulatory effects of PDE IV inhibitors. In particular, PDE inhibitors promote elevation of intracellular cAMP levels, increasing the electrical resistance of endothelial monolayers by stabilizing intercellular junctional complexes. Such an effect on central nervous system (CNS) vascular endothelium has the potential to reduce disease severity in EAE, because both inflammatory cells and humoral factors readily cross a disrupted blood-brain barrier (BBB). In this report, we demonstrate the capacity of BBB022 and rolipram to decrease clinical severity of EAE. further, PDE IV inhibitors significantly reduced BBB permeability in the spinal cords of mice with EAE. These results provide evidence that PDE IV-inhibitors may exert therapeutic effects in EAE by modifying cerebrovascular endothelial permeability, reducing tissue edema as well as entry of inflammatory cells and factors.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Blood-Brain Barrier/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Acute Disease , Animals , Blood-Brain Barrier/immunology , Brain Edema/drug therapy , Brain Edema/immunology , Central Nervous System/enzymology , Central Nervous System/immunology , Chronic Disease , Cyclic Nucleotide Phosphodiesterases, Type 4 , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred Strains , Organic Chemicals , Rats , Rats, Inbred Lew , Recurrence , Rolipram
7.
Annu Rev Neurosci ; 22: 11-28, 1999.
Article in English | MEDLINE | ID: mdl-10202530

ABSTRACT

The blood-brain barrier (BBB) is formed by brain capillary endothelial cells (ECs). In the late embryonic and early postnatal period, these cells respond to inducing factors found in the brain environment by adopting a set of defined characteristics, including high-electrical-resistance tight junctions. Although the factors have not been identified definitively, a great deal of information about brain ECs has been obtained, especially recently. This review concentrates on a cell biological analysis of the BBB, with an emphasis on regulation of the specialized intercellular junctions. The development of these junctions seems to depend on two primary processes: the appearance of high levels of the tight junction protein occludin and intracellular signaling processes that control the state of phosphorylation of junctional proteins. Recent studies have revealed that the BBB can be modulated in an ongoing way to respond to environmental stimuli.


Subject(s)
Blood-Brain Barrier/physiology , Brain/blood supply , Capillaries/physiology , Endothelium, Vascular/physiology , Animals , Capillaries/cytology , Capillary Permeability/physiology , Endothelium, Vascular/cytology , Humans , Signal Transduction/physiology , Tight Junctions/physiology
8.
Biochem J ; 338 ( Pt 2): 471-8, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10024525

ABSTRACT

Inflammatory mediators such as histamine and thrombin increase the tight-junction permeability of endothelial cells. Tight-junction permeability may be independently controlled, but is dependent on the adherens junction, where adhesion is achieved through homotypic interaction of cadherins, which in turn are associated with cytoplasmic proteins, the catenins. p120, also termed p120(cas)/p120(ctn), and its splice variant, p100, are catenins. p120, originally discovered as a substrate of the tyrosine kinase Src, is also a target for a protein kinase C-stimulated pathway in epithelial cells, causing its serine/threonine dephosphorylation. The present study shows that pharmacological activation of protein kinase C stimulated a similar pathway in endothelial cells. Activation of receptors for agents such as histamine (H1), thrombin and lysophosphatidic acid in the endothelial cells also caused serine/threonine dephosphorylation of p120 and p100, suggesting physiological relevance. However, protein kinase C inhibitors, although blocking the effect of pharmacological activation of protein kinase C, did not block the effects due to receptor activation. Calcium mobilization and the myosin-light-chain-kinase pathway do not participate in p120/p100 signalling. In conclusion, endothelial cells possess protein kinase C-dependent and -independent pathways regulating p120/p100 serine/threonine phosphorylation. These data describe a new connection between inflammatory agents, receptor-stimulated signalling and pathways potentially influencing intercellular adhesion in endothelial cells.


Subject(s)
Cytoskeletal Proteins/metabolism , Inflammation Mediators/metabolism , Calcium/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Inhibitors/pharmacology , Histamine Antagonists/pharmacology , Humans , Myosin Light Chains/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism
9.
J Cell Sci ; 110 ( Pt 14): 1603-13, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9247194

ABSTRACT

Endothelial cells provide a crucial interface between blood and tissue environments. Free diffusion of substances across endothelia is prevented by the endothelial tight junction, the permeability of which varies enormously depending on tissue. Endothelial cells of the blood-brain barrier possess tight junctions of severely limited permeability, whereas those of non-neural tissue are considerably leakier, but the molecular basis for this difference is not clear. Occludin is a major transmembrane protein localizing at the tight junction. In this study, we show, by immunocytochemistry, that occludin is present at high levels and is distributed continuously at cell-cell contacts in brain endothelial cells. In contrast, endothelial cells of non-neural tissue have a much lower expression of occludin, which is distributed in a discontinuous fashion at cell-cell contacts. The apparent differences in occludin expression levels were directly confirmed by immunoblotting. The differences in occludin protein were reflected at the message level, suggesting transcriptional regulation of expression. We also show that occludin expression is developmentally regulated, being low in rat brain endothelial cells at postnatal day 8 but clearly detectable at post-natal day 70. Our data indicate that regulation of occludin expression may be a crucial determinant of the tight junction permeability properties of endothelial cells in different tissues.


Subject(s)
Brain/blood supply , Membrane Proteins/analysis , Tight Junctions/chemistry , Tight Junctions/metabolism , Trans-Activators , Amino Acid Sequence , Animals , Astrocytes , Brain/cytology , Brain/growth & development , Cadherins/analysis , Capillaries/chemistry , Capillaries/metabolism , Cell Membrane Permeability/physiology , Cells, Cultured , Culture Media, Conditioned , Cytoskeletal Proteins/analysis , Endothelium/cytology , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Freeze Fracturing , Gene Expression Regulation, Developmental/physiology , Guinea Pigs , Membrane Proteins/genetics , Mice , Microscopy, Electron , Molecular Sequence Data , Occludin , Phosphoproteins/analysis , RNA, Messenger/analysis , Rats , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein , beta Catenin
10.
J Neurochem ; 68(3): 991-1000, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048744

ABSTRACT

Brain capillary endothelial cells are coupled by a continuous belt of complex high-electrical-resistance tight junctions that are largely responsible for the blood-brain barrier. We have investigated mechanisms regulating tight junction permeability in brain endothelial cells cultured to maintain high-resistance junctions. The phospholipid lysophosphatidic acid (LPA) was found to cause a rapid, reversible, and dose-dependent decrease in transcellular electrical resistance in brain endothelial cells. LPA also increased the paracellular flux of sucrose, which, together with the resistance decrease, indicated increased tight junction permeability. Activation of protein kinase C attenuated the effect of LPA, suggesting that it was mediated by activation of a signalling pathway. LPA did not cause any obvious relocalization of adherens junction- or tight junction-associated proteins. However, it did stimulate the formation of stress fibres, the recruitment of focal adhesion components, and the appearance of tyrosine phosphorylated protein at focal contacts. Our study shows that LPA is a modulator of tight junction permeability in brain endothelial cells in culture and raises the possibility that it triggers blood-brain barrier permeability changes under (patho)physiological conditions.


Subject(s)
Capillary Permeability/drug effects , Cerebrovascular Circulation/drug effects , Endothelium, Vascular/metabolism , Lysophospholipids/pharmacology , Tight Junctions/metabolism , Animals , Cells, Cultured , Electric Impedance , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Immunohistochemistry , Mice , Phosphorylation , Signal Transduction , Swine , Tyrosine/metabolism
11.
J Biol Chem ; 272(50): 31894-901, 1997 Dec 12.
Article in English | MEDLINE | ID: mdl-9395537

ABSTRACT

Protein kinase C signaling pathways have been implicated in the disruption of intercellular junctions, but mechanisms are not clear. p100 and p120 are members of the Armadillo family of proteins and are localized to cellular adherens junctions. In strain I Madin-Darby canine kidney cells, protein kinase C activation leads to disruption of tight junctions and an increase in permeability of cell monolayers. We show that this permeability increase is accompanied by dephosphorylation of p100/p120 on serine and threonine residues. The dephosphorylation of these proteins can also be induced by the kinase inhibitors staurosporine, KT5926, and Gö 6976. Treatment of cells with phosphatase inhibitors induced hyperphosphorylation of p100 and p120. Thus, p100 and p120 participate in a regulatable cycle of serine/threonine phosphorylation and dephosphorylation. Protein kinase C must act, directly or indirectly, by perturbing this phosphorylation cycle, by inhibition of a p100/p120 kinase and/or activation of a phosphatase. These data clearly show that p100 and p120 are targets of a novel protein kinase C signaling pathway. Dephosphorylation of these proteins precedes the permeability increase across epithelial cell monolayers seen in response to phorbol esters, raising the possibility that this pathway may play a role in the modulation of intercellular junctions.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/enzymology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Animals , Catenins , Dogs , Enzyme Activation , Models, Chemical , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Serine/metabolism , Threonine/metabolism , Delta Catenin
12.
Curr Opin Neurobiol ; 6(5): 622-7, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8937826

ABSTRACT

The blood-brain barrier regulates the movement of molecules and cells between the circulation and the CNS. Modulation of this barrier may be critical in the aetiology of various CNS pathologies. Endothelial cell tight junctions are an essential part of the barrier, and recent advances have been made in understanding how specific intracellular signalling events regulate cell-cell adhesion and tight-junction permeability.


Subject(s)
Blood-Brain Barrier , Brain/physiology , Cell Adhesion , Endothelium, Vascular/physiology , Animals , Brain/pathology , Brain/physiopathology , Cytoskeleton/physiology , Models, Neurological , Signal Transduction , Tight Junctions/physiology
14.
J Cell Biol ; 130(2): 369-81, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7615637

ABSTRACT

Cadherins and catenins play an important role in cell-cell adhesion. Two of the catenins, beta and gamma, are members of a group of proteins that contains a repeating amino acid motif originally described for the Drosophila segment polarity gene armadillo. Another member of this group is a 120-kD protein termed p120, originally identified as a substrate of the tyrosine kinase pp60src. In this paper, we show that endothelial and epithelial cells express p120 and p100, a 100-kD, p120-related protein. Peptide sequencing of p100 establishes it as highly related to p120. p120 and p100 both appear associated with the cadherin/catenin complex, but independent p120/catenin and p100/catenin complexes can be isolated. This association is shown by coimmunoprecipitation of cadherins and catenins with an anti-p120/p100 antibody, and of p120/p100 with cadherin or catenin antibodies. Immunocytochemical analysis with a p120-specific antibody reveals junctional colocalization of p120 and beta-catenin in epithelial cells. Catenins and p120/p100 also colocalize in endothelial and epithelial cells in culture and in tissue sections. The cellular content of p120/p100 and beta-catenin is similar in MDCK cells, but only approximately 20% of the p120/p100 pool associates with the cadherin/catenin complex. Our data provide further evidence for interactions among the different arm proteins and suggest that p120/p100 may participate in regulating the function of cadherins and, thereby, other processes influenced by cell-cell adhesion.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/chemistry , Endothelium/chemistry , Epithelium/chemistry , Phosphoproteins/metabolism , Trans-Activators , Amino Acid Sequence , Animals , Cadherins/analysis , Catenins , Cell Adhesion Molecules/analysis , Cell Line , Cells, Cultured , Cytoskeletal Proteins/analysis , Endothelium/cytology , Endothelium, Vascular/cytology , Epithelial Cells , Humans , Immunoblotting , Immunohistochemistry , Intercellular Junctions/chemistry , Molecular Sequence Data , Phosphoproteins/analysis , Precipitin Tests , Tumor Cells, Cultured , beta Catenin , Delta Catenin
15.
J Cell Sci ; 108 ( Pt 2): 609-19, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7769005

ABSTRACT

Tight junction permeability control is important in a variety of physiological and pathological processes. We have investigated the role of tyrosine phosphorylation in the regulation of tight junction permeability. MDCK epithelial cells and brain endothelial cells were grown on filters and tight junction permeability was determined by transcellular electrical resistance (TER). The tyrosine phosphatase inhibitor pervanadate caused a concentration- and time-dependent decrease in TER in both MDCK and brain endothelial cells. However, as expected, pervanadate resulted in the tyrosine phosphorylation of many proteins; hence interpretation of its effects are extremely difficult. Phenylarsine oxide, a more selective tyrosine phosphatase inhibitor, caused the tyrosine phosphorylation of relatively few proteins as analyzed by immunoblotting of whole cell lysates. This inhibitor, like pervanadate, also elicited a decrease in TER in the two cell types. In the MDCK cells, the action of phenylarsine oxide could be reversed by the subsequent addition of the reducing agent 2,3-dimercaptopropanol. Immunocytochemistry revealed that phenylarsine oxide rapidly stimulated the tyrosine phosphorylation of proteins associated with intercellular junctions. Because of the known influence of the adherens junction on tight junctions, we analyzed immunoprecipitates of the E-cadherin/catenin complex from MDCK cells treated with phenylarsine oxide. This revealed an increase in the tyrosine phosphorylation of beta-catenin, but not of alpha-catenin. However, the tight junction associated protein ZO-1 was also tyrosine phosphorylated after PAO treatment. These data indicate that tight junction permeability may be regulated via mechanisms involving tyrosine phosphorylation of adherens junction and tight junction proteins.


Subject(s)
Intercellular Junctions/metabolism , Tyrosine/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Line , Dogs , Endothelium/cytology , Endothelium/metabolism , Immunohistochemistry , Kidney/cytology , Kidney/metabolism , Membrane Potentials , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors
16.
J Biol Chem ; 267(35): 25239-45, 1992 Dec 15.
Article in English | MEDLINE | ID: mdl-1460024

ABSTRACT

Incubation of Swiss 3T3 cells with [2-3H]adenine, as in other cell types, reveals the ADP-ribosylation of GRP78 (the 78-kDa glucose-regulated protein, also known as BiP, the immunoglobulin heavy chain-binding protein), a resident endoplasmic reticulum protein that assists in the processing of proteins destined for secretion or cell surface expression. Here we show that Pasteurella multocida toxin, a potent growth factor for cultured fibroblasts, decreased the ADP-ribosylation of GRP78/BiP to 16 +/- 6% of the control value (n = 23). The action of the toxin occurred after a lag period, was blocked by lysosomotrophic agents, and potentiated by increased incubation time (ED50 4 ng/ml and 1 ng/ml in 4 and 8 h, respectively), thus indicating that the toxin enters the cells to act. Bombesin and platelet-derived growth factor (PDGF) similarly decreased the ADP-ribosylation of GRP78/BiP (ED50 0.5 nM and 2.5 ng/ml, respectively) but acted more rapidly than the toxin. Signaling pathways activated by the toxin, bombesin, and PDGF had effects on the ADP-ribosylation of GRP78/BiP. Thus, activation of protein kinase C alone by phorbol 12,13-dibutyrate was partially effective, and down-regulation of protein kinase C attenuated but did not block the action of the toxin, bombesin, and PDGF. Agents that mobilize Ca2+ from the endoplasmic reticulum (A23187, ionomycin, and thapsigargin) caused a decrease in the ADP-ribosylation of GRP78/BiP that was similar in magnitude to that achieved by the toxin, bombesin, and PDGF, implicating a role for inositol 1,4,5-trisphosphate-mediated Ca2+ mobilization in the action of the mitogenic agents. The growth factor-induced decrease in the ADP-ribosylation of GRP78/BiP may represent its conversion from an inactive to an active state.


Subject(s)
Bacterial Proteins , Bacterial Toxins/pharmacology , Bombesin/pharmacology , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones , Platelet-Derived Growth Factor/pharmacology , 3T3 Cells , Adenine/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Autoradiography , Carrier Proteins/isolation & purification , Cycloheximide/pharmacology , Deuterium , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum Chaperone BiP , Kinetics , Methionine/metabolism , Mice , Molecular Weight , Pasteurella , Sulfur Radioisotopes , Time Factors
17.
Proc Natl Acad Sci U S A ; 89(10): 4240-4, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1584759

ABSTRACT

The growth of many normal cells requires contact with an adhesive substratum, a requirement that is frequently abrogated in the transformed phenotype. We have explored pathways that can lead to the anchorage-independent growth of cultured Rat-1 fibroblasts. Pasteurella multocida toxin (PMT), a 146-kDa mitogenic protein, caused a striking increase in the formation of colonies (greater than 200 microns) from single cells in soft agar. The magnitude of the effect of PMT was greater than that achieved by epidermal growth factor or platelet-derived growth factor. The toxin was extremely potent, with half-maximal and maximal effects observed at 1 and 10 pM PMT, respectively. This concentration dependence of the action of the toxin is similar to that for the stimulation of DNA synthesis in adherent cultures of the cells. Stimulation of colony formation could be achieved by a transient exposure of the cells to PMT and it was blocked by methylamine, indicating that the toxin enters the cells to act. Colony formation was stimulated equally by native and recombinant PMT, but a truncated version (33.5 kDa) of the recombinant toxin was ineffective. PMT antiserum blocked colony formation in response to PMT. In the Rat-1 cells, PMT stimulated the phospholipase C-mediated hydrolysis of inositolphospholipids, as indicated by the stimulation of inositol phosphate release, Ca2+ mobilization, and phosphorylation of a protein kinase C substrate. The results indicate that the deregulation of signal-transduction pathways as elicited by an intracellularly acting bacterial toxin can induce a malignant phenotype.


Subject(s)
Bacterial Proteins , Bacterial Toxins/pharmacology , Calcium/metabolism , Cell Division/drug effects , DNA Replication/drug effects , Inositol Phosphates/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Clone Cells , Epidermal Growth Factor/drug effects , Epidermal Growth Factor/metabolism , Kinetics , Pasteurella , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Rats , Recombinant Proteins/pharmacology , Thymidine/metabolism , Tritium
18.
J Cell Biol ; 115(4): 949-58, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1835459

ABSTRACT

Certain microbial toxins are ADP-ribosyltransferases, acting on specific substrate proteins. Although these toxins have been of great utility in studies of cellular regulatory processes, a simple procedure to directly study toxin-catalyzed ADP-ribosylation in intact cells has not been described. Our approach was to use [2-3H]adenine to metabolically label the cellular NAD+ pool. Labeled proteins were then denatured with SDS, resolved by PAGE, and detected by flurography. In this manner, we show that pertussis toxin, after a dose-dependent lag period, [3H]-labeled a 40-kD protein intact cells. Furthermore, incubation of the gel with trichloroacetic acid at 95 degrees C before fluorography caused the release of label from bands other than the pertussis toxin substrate, thus, allowing its selective visualization. The modification of the 40-kD protein was ascribed to ADP-ribosylation of a cysteine residue on the basis of inhibition of labeling by nicotinamide and the release of [3H]ADP-ribose from the labeled protein by mercuric acetate. Cholera toxin catalyzed the [3H]-labeling of a 46-kD protein in the [2-3H]adenine-labeled cells. Pretreatment of the cells with pertussis toxin before the labeling of NAD+ with [2-3H]adenine blocked [2-3H]ADP-ribosylation catalyzed by pertussis toxin, but not that by cholera toxin. Thus, labeling with [2-3H]adenine permits the study of toxin-catalyzed ADP-ribosylation in intact cells. Pasteurella multocida toxin has recently been described as a novel and potent mitogen for Swiss 3T3 cell and acts to stimulate the phospholipase C-mediated hydrolysis of polyphosphoinositides. The basis of the action of the toxin is not known. Using the methodology described here, P. multocida toxin was not found to act by ADP-ribosylation.


Subject(s)
Bacterial Proteins , Bacterial Toxins/metabolism , Pasteurella multocida , Poly(ADP-ribose) Polymerases/metabolism , 3T3 Cells , Adenosine Diphosphate Ribose/metabolism , Animals , Cholera Toxin/metabolism , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/metabolism , Kinetics , Membrane Proteins/metabolism , Mice , Niacinamide/pharmacology , Pertussis Toxin , Solubility , Virulence Factors, Bordetella/metabolism
19.
J Biol Chem ; 266(8): 4840-7, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-2002031

ABSTRACT

Pasteurella multocida toxin, both native and recombinant, is an extremely potent mitogen for Swiss 3T3 cells and acts to enhance the formation of total inositol phosphates (Rozengurt, E., Higgins, T., Changer, N., Lax, A.J., and Staddon, J.M. (1990) Proc. Natl. Acad. Sci. U.S.A. 87, 123-127). P. multocida toxin also stimulates diacylglycerol production and activates protein kinase C (Staddon, J.M., Chanter, N., Lax, A.J., Higgins, T.E., and Rozengurt, E. (1990) J. Biol. Chem. 265, 11841-11848). Here we analyze, by [3H]inositol labeling and high performance liquid chromatography, the inositol phosphates in recombinant P. multocida toxin-treated cells. Recombinant P. multocida toxin stimulated increases in [3H]inositol 1,4,5-trisphosphate ([3H]Ins(1,4,5)P3) and its metabolic products, including Ins(1,3,4,5)P4, Ins(1,3,4)P3, Ins(1,4)P2, Ins(4/5)P, and Ins(1/3)P. The profile of the increase in the cellular content of these distinct inositol phosphates was very similar to that elicited by bombesin. Furthermore, recombinant P. multocida toxin, like bombesin, mobilizes an intracellular pool of Ca2+. Recombinant P. multocida toxin pretreatment greatly reduces the Ca2(+)-mobilizing action of bombesin, consistent with Ca2+ mobilization from a common pool by the two agents. The enhancement of inositol phosphates and mobilization of Ca2+ by recombinant P. multocida toxin were blocked by the lysosomotrophic agents methylamine, ammonium chloride, and chloroquine and occurred after a dose-dependent lag period. The stimulation of inositol phosphate production by recombinant P. multocida toxin persisted after removal of extracellular toxin, in contrast to the reversibility of the action of bombesin. Recombinant P. multocida toxin, unlike bombesin and guanosine 5'-O-(gamma-thiotriphosphate), did not cause the release of inositol phosphates in permeabilized cells. These data demonstrate that recombinant P. multocida toxin, acting intracellularly, stimulates the phospholipase C-mediated hydrolysis of phosphatidylinositol 4,5-bisphosphate.


Subject(s)
Bacterial Proteins , Bacterial Toxins/toxicity , Calcium/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Mitogens , Pasteurella/metabolism , Ammonium Chloride/pharmacology , Animals , Bacterial Toxins/antagonists & inhibitors , Bombesin/antagonists & inhibitors , Bombesin/toxicity , Cells, Cultured , Chloroquine/pharmacology , Chromatography, High Pressure Liquid , Methylamines/pharmacology , Mice , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/toxicity
20.
FEBS Lett ; 277(1-2): 59-64, 1990 Dec 17.
Article in English | MEDLINE | ID: mdl-2269370

ABSTRACT

Pasteurella multocida toxin is a potent mitogen for cultured Swiss 3T3 cells where it causes an accumulation of inositol phosphates and activation of protein kinase C. The gene sequence described here coded for a 146 kDa protein. The ORF was preceded by a ribosome binding site and followed by a stem loop. There was no evidence for a signal sequence. The gene had a low G + C base ratio which differs from the rest of the Pasteurella genome. There was no significant homology with other known proteins, although a motif found in certain bacterial toxins which are ADP-ribosyl transferases is present. A recombinant expressing only part of the PMT gene was not mitogenic.


Subject(s)
Bacterial Proteins , Bacterial Toxins/genetics , Genes, Bacterial , Mitogens , Pasteurella/genetics , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...