Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 34(36): 4702-12, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-25486430

RESUMO

Nuclear Foxc2 is a transcriptional regulator of mesenchymal transformation during developmental epithelial-mesenchymal transition (EMT) and has been associated with EMT in malignant epithelia. Our laboratory has shown that in normal epithelial cells Foxc2 is maintained in the cytoplasm where it promotes an epithelial phenotype. The Foxc2 amino terminus has a consensus casein kinase 2 (CK2) phosphorylation site at serine 124, and we now show that CK2 associates with Foxc2 and phosphorylates this site in vitro. Knockdown or inhibition of the CK2α/α' kinase subunit in epithelial cells causes de novo accumulation of Foxc2 in the nucleus. Mutation of serine 124 to leucine promotes constitutive nuclear localization of Foxc2 and expression of mesenchymal genes, whereas an S124D phosphomimetic leads to constitutive cytoplasmic localization and epithelial maintenance. In malignant breast cancer cells, the CK2ß regulatory subunit is downregulated and FOXC2 is found in the nucleus, correlating with an increase in α-smooth muscle actin (SMA) expression. Restoration of CK2ß expression in these cells results in cytoplasmic localization of Foxc2, decreased α-SMA expression and reduced cell migration and invasion. In contrast, knockdown of CK2ß in normal breast epithelial cells leads to FOXC2 nuclear localization, decreased E-cadherin expression, increased α-SMA and vimentin expression, and enhanced cell migration and invasion. Based on these findings, we propose that Foxc2 is functionally maintained in the cytoplasm of normal epithelial cells by CK2α/α'-mediated phosphorylation at serine 124, which is dependent on proper targeting of the holoenzyme via the CK2ß regulatory subunit.


Assuntos
Neoplasias da Mama/genética , Caseína Quinase II/biossíntese , Transição Epitelial-Mesenquimal/genética , Fatores de Transcrição Forkhead/genética , Mama/patologia , Neoplasias da Mama/patologia , Caderinas/biossíntese , Caseína Quinase II/genética , Movimento Celular/genética , Citoplasma/genética , Citoplasma/metabolismo , Células Epiteliais/patologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Vimentina/biossíntese
2.
Proc Natl Acad Sci U S A ; 98(22): 12509-14, 2001 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-11606725

RESUMO

Endostatin (ES) inhibits endothelial cell migration and has been found to bind to glypicans (Gpcs) on both endothelial cells and renal epithelial cells. We examined the possibility that ES might regulate epithelial cell morphogenesis. The addition of ES to cultured epithelial cells causes an inhibition of both hepatocyte growth factor- and epidermal growth factor-dependent process formation and migration. In contrast, ES does not inhibit epidermal growth factor-dependent morphogenesis in renal epithelial cells derived from Gpc-3 -/mice, whereas expression of Gpc-1 in these cells reconstitutes ES responsiveness. Gpc-3 -/mice have been shown to display enhanced ureteric bud (UB) branching early in development, and cultured UB cells release ES into the media, suggesting that ES binding to Gpcs may regulate UB branching. The addition of ES inhibits branching of the explanted UB, whereas a neutralizing Ab to ES enhances UB outgrowth and branching. Thus, local expression of ES at the tips of the UB may play a role in the regulation of UB arborization.


Assuntos
Colágeno/fisiologia , Rim/embriologia , Fragmentos de Peptídeos/fisiologia , Ureter/embriologia , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Endostatinas , Fator de Crescimento Epidérmico/farmacologia , Glipicanas , Proteoglicanas de Heparan Sulfato/análise , Proteoglicanas de Heparan Sulfato/fisiologia , Fator de Crescimento de Hepatócito/farmacologia , Camundongos , Morfogênese , Ratos
3.
J Biol Chem ; 276(35): 32552-8, 2001 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-11445578

RESUMO

Based on our previous observations that active ERK associates with and phosphorylates Gab1 in response to HGF, and the prediction that the ERK phosphorylation site is adjacent to one of the phosphatidylinositol 3-kinase (PI3K) SH2 binding motifs, we examined the possibility that ERK phosphorylation can regulate the Gab1/PI3K association. The HGF-mediated association of Gab1 with either full-length GST-p85 or its isolated N- or C-terminal SH2 domains was inhibited by approximately 50% in the setting of ERK inhibition, a result confirmed by co-immunoprecipitation of the native proteins. A 14-amino acid peptide encoding (472)YVPMTP(477) (one of the major p85 binding sites in Gab1 and the predicted ERK phosphorylation site) was synthesized with either phosphotyrosine alone (pY), or phosphotyrosine + phosphothreonine (pYT). In both pull-down assays and competition assays, pYT demonstrated a higher affinity for p85 than did pY alone. Finally, examination of the phosphorylation state of Akt after HGF stimulation revealed that ERK inhibition resulted in a decrease in Akt activation at both 5 and 10 min. These results suggest that activated ERK can phosphorylate Gab1 in response to HGF stimulation and thereby potentiate the Gab1/PI3K association and subsequent PI3K activation.


Assuntos
Células Epiteliais/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Butadienos/farmacologia , Linhagem Celular , Meios de Cultura Livres de Soro , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Cinética , Nitrilas/farmacologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosfopeptídeos/síntese química , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Fosfoproteínas/química , Fosfotreonina , Fosfotirosina , Subunidades Proteicas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Domínios de Homologia de src
4.
Am J Physiol Renal Physiol ; 281(1): F62-70, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11399647

RESUMO

Hepatocyte growth factor (HGF) has been shown to enhance recovery from renal tubular ischemia. We investigated the possibility that HGF improves recovery by preventing ischemia-induced loss of cell adhesion. Murine inner medullary collecting duct-3 (mIMCD-3) cells subjected to 90% ATP depletion demonstrated a 55% decrease in adhesion, an effect that was completely reversed by the addition of HGF. Assays examining release of adherent cells revealed similar results with 30 min of ATP depletion causing loss of adhesion of 25% of mIMCD-3 cells and HGF completely reversing this effect. In contrast, HGF was unable to reverse the loss of adhesion of cells exposed to 99% ATP depletion. Examination of the mitogen-activated protein kinase (MAPK) signaling pathway revealed that HGF could induce extracellular signal-regulated kinase (ERK) phosphorylation in control and 90% ATP-depleted cells but not in 99% ATP-depleted cells. Inhibition of ERK activation with U0126 completely blocked the HGF-dependent reversal of ATP-depleted cell adhesion. Thus ATP-depleted cells demonstrate a marked decrease in cell adhesion that is reversible by the addition of HGF. This effect of HGF requires activation of the MAPK pathway.


Assuntos
Adesão Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Túbulos Renais/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Trifosfato de Adenosina/deficiência , Animais , Butadienos/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas/farmacologia
5.
J Biol Chem ; 276(12): 9166-73, 2001 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-11118451

RESUMO

Cells derived from the inner medullary collecting duct undergo in vitro branching tubulogenesis to both the c-met receptor ligand hepatocyte growth factor (HGF) as well as epidermal growth factor (EGF) receptor ligands. In contrast, many other cultured renal epithelial cells respond in this manner only to HGF, suggesting that these two receptors may use independent signaling pathways during morphogenesis. We have therefore compared the signaling pathways for mIMCD-3 cell morphogenesis in response to EGF and HGF. Inhibition of the p42/44 mitogen-activated protein kinase (MAPK) pathway with the mitogen-activated protein kinase kinase (MKK1) inhibitor PD98059 (50 microm) markedly inhibits HGF-induced cell migration with only partial inhibition of EGF-induced cell motility. Similarly, HGF-dependent, but not EGF-dependent, branching morphogenesis was more greatly inhibited by the MKK1 inhibitor. Examination of EGF-stimulated cells demonstrated that extracellular-regulated kinase 5 (ERK5) was activated in response to EGF but not HGF, and that activation of ERK5 was only 60% inhibited by 50 microm PD98059. In contrast, the MKK inhibitor U0126 markedly inhibited both ERK1/2 and ERK5 activation and completely prevented HGF- and EGF-dependent migration and branching process formation. Expression of dominant negative ERK5 (dnBMK1) likewise inhibited EGF-dependent branching process formation, but did not affect HGF-dependent branching process formation. Our results indicate that activation of the ERK1/ERK2 signaling pathway is critical for HGF-induced cell motility/morphogenesis in mIMCD-3 cells, whereas ERK5 appears to be required for EGF-dependent morphogenesis.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Rim/crescimento & desenvolvimento , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Linhagem Celular Transformada , Movimento Celular , Rim/citologia , Rim/enzimologia , Rim/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Morfogênese
6.
J Biol Chem ; 275(26): 20027-32, 2000 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-10748075

RESUMO

Goodpasture syndrome is an autoimmune disease of the kidneys and lungs mediated by antibodies and T-cells directed to cryptic epitopes hidden within basement membrane hexamers rich in alpha3 non-collagenous globular (NC1) domains of type IV collagen. These epitopes are normally invisible to the immune system, but this privilege can be obviated by chemical modification. Endogenous drivers of immune activation consequent to the loss of privilege have long been suspected. We have examined the ability of reactive oxygen species (ROS) to expose Goodpasture epitopes buried within NC1 hexamers obtained from renal glomeruli abundant in alpha3(IV) NC1 domains. For some hexameric epitopes, like the Goodpasture epitopes, exposure to ROS specifically enhanced recognition by Goodpasture antibodies in a sequential and time-dependent fashion; control binding of epitopes to alpha3(IV) alloantibodies from renal transplant recipients with Alport syndrome was decreased, whereas epitope binding to heterologous antibodies recognizing all alpha3 NC1 epitopes remained the same. Inhibitors of hydrogen peroxide and hydroxyl radical scavengers were capable of attenuating the effects of ROS in cells and kidney by 30-50%, respectively, thereby keeping the Goodpasture epitopes largely concealed when compared with a 70% maximum inhibition by iron chelators. Hydrogen peroxide administration to rodents was sufficient to expose Goodpasture epitope in vivo and initiate autoantibody production. Our findings collectively suggest that ROS can alter the hexameric structure of type IV collagen to expose or destroy selectively immunologic epitopes embedded in basement membrane. The reasons for autoimmunity in Goodpasture syndrome may lie in an age-dependent deterioration in inhibitor function modulating oxidative damage to structural molecules. ROS therefore may play an important role in shaping post-translational epitope diversity or neoantigen formation in organ tissues.


Assuntos
Doença Antimembrana Basal Glomerular/metabolismo , Epitopos/metabolismo , Espécies Reativas de Oxigênio , Animais , Doença Antimembrana Basal Glomerular/imunologia , Autoanticorpos/sangue , Membrana Basal/metabolismo , Colágeno/química , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Imunofluorescência , Humanos , Peróxido de Hidrogênio/farmacologia , Rim/imunologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Ratos , Fatores de Tempo
7.
J Biol Chem ; 274(51): 36362-8, 1999 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-10593929

RESUMO

Grb2-associated binder 1 (GAB1) is a docking protein found to associate with the activated c-MET receptor via the MET-binding domain (MBD) and appears to be critical for the tubulogenic actions of this receptor. Pull-down experiments with bacterially expressed MBD and full-length GAB1 revealed the presence of c-MET as well as phosphorylated ERK2 (pERK2). By using purified pERK2 and non-pERK2, we found that GAB1 associates exclusively with the phosphorylated form of the enzyme and that this association does not require mediation by a third protein. When epitope-tagged GAB1 was co-transfected with constitutively active MEK1 into A293 cells, co-immunoprecipitation of GAB1 and pERK2 was observed, demonstrating that this interaction can occur in intact cells. In vitro, both the MBD and full-length GAB1 were found to be substrates for activated ERK2. In intact cells, epitope-tagged GAB1 was found to be basally phosphorylated on serine with an increase following co-transfection with constitutively active MEK1 and the appearance of novel phosphorylation sites detected by phosphopeptide mapping. Thus, it appears that GAB1 can associate directly with phosphorylated ERK2 via the MET-binding domain and that GAB1 then acts as a substrate for the enzyme.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Cães , Ativação Enzimática , Fosforilação , Especificidade por Substrato
8.
J Biol Chem ; 273(37): 23750-7, 1998 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-9726983

RESUMO

It has been demonstrated that the lipid products of the phosphoinositide 3-kinase (PI3K) can associate with the Src homology 2 (SH2) domains of specific signaling molecules and modify their actions. In the current experiments, phosphatidylinositol 3,4, 5-trisphosphate (PtdIns-3,4,5-P3) was found to bind to the C-terminal SH2 domain of phospholipase Cgamma (PLCgamma) with an apparent Kd of 2.4 microM and to displace the C-terminal SH2 domain from the activated platelet-derived growth factor receptor (PDGFR). To investigate the in vivo relevance of this observation, intracellular inositol trisphosphate (IP3) generation and calcium release were examined in HepG2 cells expressing a series of PDGFR mutants that activate PLCgamma with or without receptor association with PI3K. Coactivation of PLCgamma and PI3K resulted in an approximately 40% increase in both intracellular IP3 generation and intracellular calcium release as compared with selective activation of PLCgamma. Similarly, the addition of wortmannin or LY294002 to cells expressing the wild-type PDGFR inhibited the release of intracellular calcium. Thus, generation of PtdIns-3,4,5-P3 by receptor-associated PI3K causes an increase in IP3 production and intracellular calcium release, potentially via enhanced PtdIns-4, 5-P2 substrate availability due to PtdIns-3,4,5-P3-mediated recruitment of PLCgamma to the lipid bilayer.


Assuntos
Cálcio/metabolismo , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo , Androstadienos/farmacologia , Sítios de Ligação , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Morfolinas/farmacologia , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipase C gama , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas , Wortmanina
9.
J Biol Chem ; 273(8): 4465-9, 1998 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-9468499

RESUMO

Signal transduction across cell membranes often involves the activation of both phosphatidylinositol (PI)-specific phospholipase C (PLC) and phosphoinositide 3-kinase (PI 3-kinase). Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a substrate for both enzymes, is converted to phosphatidylinositol 3,4, 5-trisphosphate (PI(3,4,5)P3) by the action of PI 3-kinase. Here, we show that PI(3,4,5)P3 activates purified PLC-gamma isozymes by interacting with their Src homology 2 domains. Furthermore, the expression of an activated catalytic subunit of PI 3-kinase in COS-7 cells resulted in an increase in inositol phosphate formation, whereas platelet-derived growth factor-induced PLC activation in NIH 3T3 cells was markedly inhibited by the specific PI 3-kinase inhibitor LY294002. These results suggest that receptors coupled to PI 3-kinase may activate PLC-gamma isozymes indirectly, in the absence of PLC-gamma tyrosine phosphorylation, through the generation of PI(3,4,5)P3.


Assuntos
Isoenzimas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Células COS , Ativação Enzimática , Células HeLa , Humanos , Fosfolipase C gama , Transdução de Sinais , Especificidade por Substrato , Domínios de Homologia de src
10.
Am J Physiol ; 273(3 Pt 2): F463-72, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9321921

RESUMO

Much attention has recently focused upon hepatocyte growth factor (HGF) as a potential regulator of epithelial branching morphogenesis. However, since neither the HGF nor c-met "knockout" mice show abnormal kidney branching morphogenesis, we sought to analyze the relative importance of HGF in in vitro branching morphogenesis compared with other factors secreted by the embryonic kidney. Exploiting an assay that employs kidney epithelial cells (murine inner medullary collecting duct, mIMCD3) seeded in collagen cocultured with the embryonic kidney, we found that a tyrosine kinase inhibitor that is highly specific for the epidermal growth factor (EGF) receptor (EGFR), tyrphostin AG1478, inhibited mIMCD3 cell process formation (an early step in branching tubulogenesis) by 40%, whereas high concentrations of neutralizing anti-HGF antibodies had a lesser effect (20% inhibition), suggesting that EGFR ligands account for a larger fraction of branching morphogens secreted by the embryonic kidney than HGF. In addition, when an embryonic epithelial cell line derived from c-met (-/-) mice was cocultured with the embryonic kidney, these c-met (-/-) cells underwent process formation. EGFR ligands but not HGF were able to induce branching tubulogenesis in these cells. All EGFR ligands tested, including EGF, transforming growth factor-alpha, heparin-binding EGF, betacellulin, and amphiregulin, induced mIMCD3 cell tubulogenesis. EGFR ligands caused upregulation of urokinase, urokinase receptor, and matrix metalloprotease-1, and tubulogenesis could be inhibited by the metalloprotease inhibitor 1,10-phenanthroline. Our results support the notion that multiple parallel and potentially redundant growth factor-dependent pathways regulate branching tubulogenesis.


Assuntos
Receptores ErbB/metabolismo , Fator de Crescimento de Hepatócito/fisiologia , Rim/fisiologia , Nitrilas/farmacologia , Quinazolinas/farmacologia , Tirfostinas , Animais , Aprotinina/farmacologia , Linhagem Celular , Técnicas de Cocultura , Colágeno , Inibidores de Cisteína Proteinase/farmacologia , Indução Embrionária , Inibidores Enzimáticos/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/fisiologia , Fator de Crescimento de Hepatócito/deficiência , Rim/efeitos dos fármacos , Rim/embriologia , Medula Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Leucina/análogos & derivados , Leucina/farmacologia , Ligantes , Camundongos , Camundongos Knockout , Morfogênese/efeitos dos fármacos , Fenantrolinas/farmacologia , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met , Proto-Oncogenes , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Fator de Crescimento Transformador alfa/farmacologia
11.
Kidney Int ; 52(1): 130-9, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9211355

RESUMO

The isolated perfused rat kidney provides a model of selective hypoxia to the medullary thick ascending limb. To investigate the relationship between immediate early gene expression and the extent of hypoxic damage, we determined expression of the immediate early genes (IEG) c-fos and egr-1 in isolated perfused kidneys during standard perfusion and after various measures shown previously to be protective. mRNA levels of c-fos and egr-1 were markedly increased in kidneys after 90 minutes of standard perfusion with Krebs-Henseleit buffer containing albumin. Gene expression was most prominent in the outer medulla followed by papilla and cortex, a pattern reflected by the immunohistochemical demonstration of a prominent accumulation of both egr-1 and c-fos polypetides mainly in the medullary thick ascending limb (mTAL). Protective measures known to minimize morphological damage to the mTAL, including hyperoncotic perfusion, perfusion with glycine, or perfusion with a mixture of amino acids, decreased mRNA levels of c-fos and egr-1 in the outer medulla (by 50% and 35%, respectively) and the papilla (by 60 and 30%, respectively). Renal cortex showed only minor changes. In contrast, prevention of tubular transport by perfusion with 1 mM ouabain increased mRNA levels of c-fos and egr-1 in the outer medulla by 100% and 60%, respectively. Ouabain also dramatically increased mRNA levels of both IEGs in two lines of cultured renal epithelial cells. Changes in the level and distribution of the protein products of these IEGs were not detectable in perfused kidneys by immunohistochemistry. Hypoxic injury of the kidney stimulates IEG expression even in the absence of reperfusion. Protection against hypoxic injury in the mTAL correlates with suppression of IEG mRNA levels when protection is provided by amino acids or hyperoncotic perfusion, but not when provided by inhibition of Na,K-ATPase, which stimulates IEG expression. We conclude that diminished IEG expression is not a necessary concomitant of protection against hypoxic injury.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Imediatamente Precoces , Rim/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição/metabolismo , Aminoácidos/farmacologia , Animais , Northern Blotting , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Cães , Proteína 1 de Resposta de Crescimento Precoce , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica , Glicina/farmacologia , Imuno-Histoquímica , Técnicas In Vitro , Rim/efeitos dos fármacos , Masculino , Ouabaína/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
12.
J Biol Chem ; 272(10): 6465-70, 1997 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-9045671

RESUMO

Phosphoinositide 3-kinase has been implicated as an activator of cell motility in a variety of recent studies, yet the role of its lipid product, phosphatidylinositol 1,4,5-trisphosphate (PtdIns-3,4,5-P3), has yet to be elucidated. In this study, three independent preparations of PtdIns-3,4,5-P3 were found to increase the motility of NIH 3T3 cells when examined utilizing a microchemotaxis chamber. Dipalmitoyl L-alpha-phosphatidyl-D-myo-inositol 3,4,5-triphosphate (Di-C16-PtdIns-3,4,5-P3) also produced actin reorganization and membrane ruffling. Cells pretreated with 12-O-tetradecanoylphorbol-13-acetate to cause down-regulation of protein kinase C (PKC) exhibited complete inhibition of cell motility induced by Di-C16-PtdIns-3,4,5-P3. These results are consistent with previous observations that PtdIns-3,4,5-P3 activates Ca2+-independent PKC isoforms in vitro and in vivo and provide the first demonstration of an in vivo role for the lipid products of the phosphoinositide 3-kinase. PtdIns-3,4,5-P3 appears to directly initiate cellular motility via activation of a PKC family member.


Assuntos
Movimento Celular , Fosfatos de Fosfatidilinositol/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteína Quinase C/metabolismo , Células 3T3 , Androstadienos/farmacologia , Animais , Membrana Celular/ultraestrutura , Regulação para Baixo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Camundongos , Naftalenos/farmacologia , Fosfatidilinositol 3-Quinases , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Wortmanina
13.
Am J Physiol ; 272(2 Pt 2): F222-8, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9124399

RESUMO

The growth factor/receptor combination of hepatocyte growth factor (HGF)/c-met has been postulated to be critical for mesenchymal-to-epithelial conversion and tubule formation in the developing kidney. We therefore isolated and immortalized cells from embryonic kidneys of met -/- transgenic mice to determine whether these cells were epithelial and able to chemotax and form tubules in vitro. The cells were immortalized with retrovirus expressing human papillomavirus 16 (HPV 16) E6/E7 genes. Two rapidly dividing clones were isolated and found to express the epithelial cell markers cytokeratin, zonula occludens-1, and E-cadherin but not to express the fibroblast marker vimentin. The met -/- cells were able to chemotax in response to epidermal growth factor and transforming growth factor-alpha (TGF-alpha) and form tubules in vitro in response to TGF-alpha but not HGF. These experiments suggest that the HGF/c-met axis is not essential for epithelial cell development in the embryonic kidney and demonstrate that other growth factors are capable of supporting early tubulogenesis.


Assuntos
Quimiotaxia , Receptores ErbB/metabolismo , Túbulos Renais/embriologia , Rim/embriologia , Rim/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células-Tronco/fisiologia , Animais , Biomarcadores , Northern Blotting , Chlorocebus aethiops , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário e Fetal , Células Epiteliais , Epitélio/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Rim/citologia , Ligantes , Masculino , Camundongos , Camundongos Transgênicos/genética , Testes de Precipitina , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/genética
14.
Am J Physiol ; 271(6 Pt 2): F1103-13, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8997383

RESUMO

The control of epithelial cell movement and shape change is complex and requires regulation of a broad range of events including cell-cell adhesion contacts, cell-substratum interactions, and the actin cytoskeleton. Utilizing the hepatocyte growth factor tyrosine kinase receptor, c-met, the present review examines how growth factor receptors activate intracellular signaling pathways, which can then regulate the events necessary for epithelial cells to disassemble their existing structure, undergo extensive shape change and cell body movement, and reassemble into a polarized epithelium. The role of growth factor-mediated activation of the phosphoinositide 3-kinase, phospholipase C-gamma, c-src family members, and ras family members is addressed in relation to integrin-mediated cell-basement membrane contacts, cadherin-mediated cell-cell adhesions, and regulation of the actin cytoskeleton.


Assuntos
Substâncias de Crescimento/fisiologia , Rim/fisiologia , Animais , Comunicação Celular , Movimento Celular , Citoesqueleto/fisiologia , Células Epiteliais , Humanos , Rim/citologia , Rim/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais
15.
J Biol Chem ; 271(8): 4251-5, 1996 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-8626770

RESUMO

Interaction of hepatocyte growth factor with its high affinity receptor c-met initiates a cascade of intracellular events leading to epithelial motility. An 11-amino acid sequence from the c-met receptor has been found to cause cell transformation in transfected fibroblasts (Ponzetto, C., Bardelli, A., Zhen, Z., Maina, F., Dalla, Z. P., Giordano, S., Graziani, A., Panayotou, G., and Comoglio, P. M.(1994) Cell 77, 261-271). We inserted this sequence into a mutant platelet-derived growth factor receptor (F5) to determine if this region of c-met can initiate cell motility and which signaling pathways it activates. The platelet-derived growth factor (PDGF) receptor/c-met hybrid (F5 met) initiated PDGF-dependent chemotaxis in renal epithelial cells (8.0 +/- 2.3 versus 70.5 +/- 4.8 cells/mm2), while the parental construct, F5, did not. Addition of PDGF to cells expressing F5 met caused activation of the phosphatidylinositol (PI) 3-kinase (control 2.0 +/- 0.8, +PDGF 17.1 +/- 5.1, n = 3, p < 0.05) and phospholipase C (control 478.5 +/- 67 dpm/well, +PDGF 1049.3 +/- 93, n = 4, p = 0.003), while neither pathway was activated in cells expressing F5. The chemotactic response of F5 met was inhibited by both the PI 3-kinase inhibitor wortmannin and the phospholipase C inhibitor U-71322. Selective activation of the PI 3-kinase utilizing a PDGF receptor mutant (F3) containing the native high affinity PI 3-kinase binding site also resulted in PDGF stimulated chemotaxis, although less than that generated by the c-met sequence. These findings demonstrate that the 11-amino acid sequence from c-met initiates epithelial motility via coincident activation of the PI 3-kinase and phospholipase C and that selective activation of the PI 3-kinase can initiate a partial chemotactic response.


Assuntos
Quimiotaxia/fisiologia , Fator de Crescimento de Hepatócito/farmacologia , Fragmentos de Peptídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Fosfolipases Tipo C/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Linhagem Celular , Quimiotaxia/efeitos dos fármacos , Clonagem Molecular , Epitélio/efeitos dos fármacos , Epitélio/fisiologia , Túbulos Renais , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fosfatidilinositol 3-Quinases , Fator de Crescimento Derivado de Plaquetas/farmacologia , Mutação Puntual , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/química , Receptores do Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transfecção
16.
Am J Physiol ; 268(6 Pt 2): F1211-7, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7611461

RESUMO

The association of hepatocyte growth factor (HGF) with its high-affinity receptor, c-met, has been shown to induce mitogenesis, motogenesis, and morphogenesis in renal epithelial cells (L. G. Cantley, E. J. G. Barros, M. Gandhi, M. Rauchman, and S. K. Nigam. Am. J. Physiol. 267 (Renal Fluid Electrolyte Physiol. 36): F271-F280, 1994), suggesting that HGF may be critical to the orchestration of both renal development and regeneration following injury. Although signal transduction pathways activated by c-met include the phosphatidylinositol 3-kinase (PI-3-kinase), phospholipase C gamma, ras, and others, the activation of PI-3-kinase has been the most striking in vivo. We therefore investigated whether the pathways that mediate phenotypic changes in inner medullary collecting duct cells are altered by inhibition of PI-3-kinase with the fungal metabolite, wortmannin. In these cells, the mean inhibitory concentration for in vitro wortmannin inhibition of PI-3-kinase was approximately 0.2 nM. At this low concentration, motogenesis (quantified by chemotaxis) and morphogenesis (by branching-process formation within collagen matrix) were inhibited in a striking and parallel fashion, while mitogenesis was inhibited to a lesser degree. These experiments suggest that activation of PI-3-kinase is critical for c-met-mediated chemotaxis and tubulogenesis.


Assuntos
Quimiotaxia/fisiologia , Fator de Crescimento de Hepatócito/farmacologia , Medula Renal/citologia , Medula Renal/fisiologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Androstadienos/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Quimiotaxia/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Genes ras , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Cinética , Mitose/efeitos dos fármacos , Morfogênese/efeitos dos fármacos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-met , Proto-Oncogenes , Receptores Proteína Tirosina Quinases/biossíntese , Transdução de Sinais , Timidina/metabolismo , Wortmanina
17.
Kidney Int ; 47(6): 1752-9, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7643546

RESUMO

The possible protective effect of heat-shock proteins (HSPs) on ischemic injury to renal cells was assessed in two different experimental models: ischemia-reflow in intact rats and medullary hypoxic injury as seen in the isolated perfused rat kidney. Heat shock was induced by raising the core temperature of rats to 42 degrees C for 15 minutes. Following this, Northern blots showed enhanced gene expression of HSP70, HSP60 and ubiquitin at one hour and reaching a maximum by six hours after heat shock in all regions of the kidney, but most prominently in medulla and papilla. The HSP70 protein in the kidney, estimated by immunohistochemical means, was detectable 24 hours following heat shock and further increased at 48 hours following heat shock. In the first set of experiments, the animals underwent uninephrectomy followed by cross clamping of the remaining renal artery for 40 minutes prior to reflow. Serum creatinine and urea nitrogen rose to 3.15 +/- 0.98 and 126.4 +/- 62.5 mg/dl at 24 hours. No significant differences were observed at 24, 48 and 72 hours after reflow between these values in control rats and rats pretreated with heat shock 48 hours earlier. Severe morphological damage to proximal tubules of the renal cortex was observed to the same extent in both groups. In a second set of experiments, the right kidney was removed either 24 or 48 hours after heat shock and perfused in isolation for 90 minutes. Functional and morphological parameters were compared with those of isolated perfused kidneys obtained from animals that had not been subjected to heat shock.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Proteínas de Choque Térmico/metabolismo , Isquemia/metabolismo , Isquemia/patologia , Túbulos Renais/patologia , Circulação Renal , Animais , Proteínas de Choque Térmico/genética , Temperatura Alta , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Perfusão , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reperfusão
18.
J Am Soc Nephrol ; 5(11): 1872-81, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7620084

RESUMO

Signal transduction by tyrosine kinase growth factor receptors involves the activation of multiple intracellular signaling pathways. In many cases, this occurs via direct binding of a downstream signaling protein to the phosphorylated receptor via src-homology 2 domains on the signaling protein. In this review of the hepatocyte growth factor receptor c-met, the ability of the amino acid sequence of the receptor to dictate which signaling proteins are activated is described, with particular emphasis on association with the phosphatidylinositol 3-kinase. Recent developments that provide new understanding of the mechanisms of downstream signal transduction by the phosphatidylinositol 3-kinase are discussed, including how these might be involved in the mitogenic, motogenic, and tubulogenic effects of hepatocyte growth factor on renal epithelial cells.


Assuntos
Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Ativação Enzimática , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Rim/metabolismo , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/química
19.
Am J Physiol ; 267(2 Pt 2): F231-6, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8067383

RESUMO

Hepatocyte growth factor (HGF) and its high-affinity receptor, c-met, have been found to increase in the whole kidney of the rat following several types of renal injury or renal hypertrophy. In an attempt to determine whether the upregulation of this growth factor and its receptor is selective for the regions of greatest anatomic change, and therefore likely to be important in regulating renal tubular hyperplasia and/or hypertrophy, we examined their expression in liver, whole kidney, and subsections of the kidney following either sham operation, transient ischemia of one kidney, or unilateral nephrectomy. The message for HGF was increased in both liver and kidney by all surgical procedures tested, including sham operation, and was seen predominantly in the outer cortex, the site of least morphological change. However, c-met was not upregulated by sham operation or in the liver, but rather was selectively upregulated only in the kidney in both the hypertrophy and hyperplasia (ischemia/reflow) models. Renal subsections revealed that this increase was confined to the renal medulla, with the greatest change in the outer medulla. Thus induction of the message for HGF can occur nonselectively and at sites distant to the injurious stimulus, whereas the target for HGF, c-met, is upregulated selectively at the site of greatest tubular injury or hypertrophy. These results support a role for HGF/c-met in regulation of these renal tubular events.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Rim/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Fator de Crescimento de Hepatócito/genética , Hiperplasia , Hipertrofia , Isquemia/metabolismo , Isquemia/patologia , Masculino , Proteínas Proto-Oncogênicas c-met , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/genética , Circulação Renal , Reperfusão , Distribuição Tecidual , Regulação para Cima
20.
Am J Physiol ; 267(2 Pt 2): F271-80, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8067388

RESUMO

Hepatocyte growth factor (HGF) has been implicated in branching tubulogenesis of the developing kidney and in response to renal injury. We therefore examined the effects of response to renal injury. We therefore examined the effects of HGF on a recently described murine inner medullary collecting duct epithelial cell line (mIMCD-3 cells) in comparison with Madin-Darby canine kidney (MDCK) cells. HGF induced mitosis, scattering, and tubulogenesis in both mIMCD-3 cells and MDCK cells. However, mIMCD-3 cells underwent branching tubulogenesis under matrix conditions that did not support these morphogenetic changes in MDCK cells, suggesting substantial differences in regulation of tubulogenesis in these two cell types. In quiescent mIMCD-3 cells, the high-affinity receptor for HGF, c-met, was expressed in a nonphosphorylated state. After stimulation with HGF, there was a > 10-fold increase in receptor tyrosine phosphorylation and selective association with at least two intracellular proteins, including the phosphatidylinositol-3-kinase. Thus mIMCD-3 cells, which undergo HGF-dependent mitosis, scattering, and branching tubulogenesis, express the c-met receptor in a highly regulated state and therefore should make an excellent model for examining the mechanisms of HGF-dependent tubulogenesis in the renal collecting duct.


Assuntos
Fator de Crescimento de Hepatócito/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais/crescimento & desenvolvimento , Mitose , Animais , Linhagem Celular , Cães , Rim/crescimento & desenvolvimento , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Camundongos , Mitógenos/farmacologia , Fosfatidilinositol 3-Quinases , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores Proteína Tirosina Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...