Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Exp Eye Res ; 145: 10-16, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26546726

RESUMEN

Topical fluoroquinolones are widely used to prevent ocular infections after ophthalmic surgery. However, they have been shown to affect the corneal cell motility, whose mechanism remains indefinite. The purpose of this study was to investigate how fluoroquinolones affect corneal stromal cell motility. Human corneal fibroblasts (HCFs) were incubated in ciprofloxacin (CIP), levofloxacin (LEV), or moxifloxacin (MOX) at 0, 10, 50, and 100 µg/ml for up to 3 days. Effect of CIP, LEV, or MOX on HCF migration was monitored using migration assay. HCF viability was determined by WST-1 assay. Expression of focal adhesion kinase (FAK), paxillin (PXN), and their phosphorylated forms were analyzed by immunoblotting. Binding affinity between FAK and PXN was determined by co-immunoprecipitation. Our results revealed that CIP and MOX, but not LEV, noticeably retarded HCF migration. HCF proliferation was significantly reduced by CIP (38.2%), LEV (29.5%), and MOX (21.3%), respectively (p = 0.002). CIP and MOX suppressed the phosphorylation of PXN at tyrosines (10.2 ± 4.3%, p < 0.001; 11.7 ± 2.4%, p < 0.001, respectively), including tyrosine 118 (33.3 ± 5.2%, p < 0.001; 34.0 ± 4.4%, p < 0.001, respectively). CIP and MOX diminished the binding affinity between FAK and PXN (8.2 ± 1.8%, p < 0.001; 9.0 ± 4.5%, p < 0.001, respectively). Nevertheless, tyrosine dephosphorylation and FAK dissociation of PXN were not found in LEV-treated HCFs. None of these fluoroquinolones affect phosphorylation of FAK-Y397. We conclude that CIP and MOX, but not LEV, might delay corneal fibroblast migration via interfering with recruitment of PXN to focal adhesions and dephosphorylation of PXN at the tyrosines.


Asunto(s)
Úlcera de la Córnea/tratamiento farmacológico , Epitelio Corneal/metabolismo , Fluoroquinolonas/administración & dosificación , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Paxillin/biosíntesis , Adulto , Anciano , Antibacterianos/administración & dosificación , Recuento de Células , Movimiento Celular/efectos de los fármacos , Úlcera de la Córnea/metabolismo , Úlcera de la Córnea/patología , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/patología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Humanos , Immunoblotting , Masculino , Persona de Mediana Edad , Soluciones Oftálmicas , Paxillin/efectos de los fármacos , Adulto Joven
2.
Neurosci Res ; 59(2): 205-14, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17707940

RESUMEN

We have previously shown that endothelin-B receptor stimulation increases neural progenitor proliferation, partly in G(i) and extracellular matrix molecule-dependent manner. In the present study, we investigated whether G(q/11) is also involved in this response and how G(i) and G(q/11) might regulate the extracellular signal-regulated kinase (ERK) pathway and integrin signaling. Endothelin-induced ERK phosphorylation was independent of integrin ligands, and an inhibitor of G(q/11), YM-254890, as well as pertussis toxin, partially inhibited endothelin-stimulated phosphorylation of Raf-1 and ERK. Endothelin-stimulated protein kinase C (PKC) was partially inhibited by both YM-254890 and pertussis toxin, while only pertussis toxin attenuated endothelin-induced Ras activation. In contrast, endothelin increased tyrosine phosphorylation of focal adhesion kinase (FAK) and paxillin in an integrin ligand-dependent manner. Both YM-254890 and pertussis toxin partially inhibited endothelin-stimulated phosphorylation of these proteins. A PKC inhibitor and down-regulation of PKC prevented endothelin-induced phosphorylation of paxillin and ERK. In addition, endothelin-induced proliferation and DNA synthesis were partially inhibited by YM-254890 and pertussis toxin. Taken together, the results indicate that endothelin activates PKC via G(q/11) and G(i), and consequently stimulates the ERK cascade in cooperation with Ras signaling stimulated by G(i). PKC appears to increase tyrosine phosphorylation of paxillin to enhance integrin signaling, which further increases DNA synthesis and proliferation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Endotelinas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Integrinas/metabolismo , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , ADN/biosíntesis , Endotelinas/farmacología , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neuronas/efectos de los fármacos , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos
3.
Oncotarget ; 8(13): 21674-21691, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28423510

RESUMEN

Genistein is one of the main components of soy-based foods, which are widely known for their many benefits, including anti-cancer, anti-inflammatory, and antioxidant effects. In this study, we investigated the anti-metastasis effects of genistein on B16F10 melanoma cells. Our results showed that genistein strongly inhibited B16F10 cell proliferation and induced apoptosis in time- and concentration-dependent manners. Genistein altered the morphology of B16F10 cells to an elongated shape with slim pseudopodia-like protrusions. Moreover, genistein inhibited the invasion and migration abilities of B16F10 cells in a dose-dependent manner. On one hand, a high concentration of genistein (100 µM) significantly inhibited cell adhesion and migration, as shown by wound healing assays and transwell-migration and invasion assays. Furthermore, the expression levels of p-FAK, p-paxillin, tensin-2, vinculin, and α-actinin were decreased by genistein. As a result, genistein is believed to strongly downregulate the migration and invasion abilities of B16F10 cells via the FAK/paxillin pathway. Moreover, p-p38, p-ERK, and p-JNK levels were also dramatically decreased by treatment with genistein. Finally, genistein significantly decreased the gene expression of FAK, paxillin, vimentin, and epithelial-to-mesenchymal transition-related transcription factor Snail, as shown by real-time PCR (qPCR) analysis. On the other hand, a lower concentration of genistein (12.5 µM) significantly promoted both invasion and migration by activating the FAK/paxillin and MAPK signaling cascades. Taken together, this study showed for the first time that genistein exerts dual functional effects on melanoma cells. Our findings suggest that genistein regulates the FAK/paxillin and MAPK signaling pathways in a highly concentration-dependent manner. Patients with melanoma should therefore be cautious of consuming soy-based foods in their diets.


Asunto(s)
Anticarcinógenos/farmacología , Genisteína/farmacología , Melanoma Experimental/patología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Invasividad Neoplásica/patología , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Periodontol ; 77(5): 883-90, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16671882

RESUMEN

BACKGROUND: Tumor necrosis factor-alpha (TNF-alpha) is associated with chronic gingival inflammation and is suspected to influence periodontal destruction. However, the exact roles of TNF-alpha in wound healing and periodontal tissue regeneration are largely unknown. In the present study, we examined the effects of TNF-alpha on migration and proliferation of human periodontal ligament (PDL) cells. METHODS: PDL cells were cultured in the presence of TNF-alpha to determine its effects on cellular migration and proliferation. The protein expression profiles of alpha5 and beta1 integrin subunits and their related molecules, paxillin and focal adhesion kinases (FAK), were investigated. Gene expression of fibronectin also was assayed. Further, the activation of Rho-family small guanosine triphosphate (GTP)-binding protein (RhoA) was evaluated using a GTP-loading pull-down assay, and focal adhesion formation by PDL cells after transfection with the expression vector of paxillin-fused green fluorescent protein (GFP) also was observed with confocal microscopy. RESULTS: Cellular migration was impaired by TNF-alpha and recovered following the addition of anti-TNF-alpha antibodies. In contrast, PDL cell proliferation was not affected by TNF-alpha. TNF-alpha upregulated the expression of the alpha5 and beta1 integrin subunits, whereas fibronectin was not overexpressed. Phosphorylation of paxillin and FAK by PDL cells was induced, and RhoA activation also was induced. Confocal microscopic analysis revealed that TNF-alpha induced focal adhesion and stress fiber formation in all parts of the cells. CONCLUSION: Our results suggested that TNF-alpha impairs cellular migration by enhancing cellular adhesive ability following significant focal adhesion and stress fiber formation.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ligamento Periodontal/citología , Factor de Necrosis Tumoral alfa/farmacología , Adolescente , Femenino , Fibronectinas/genética , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrina alfa5beta1/efectos de los fármacos , Integrina alfa5beta1/metabolismo , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
5.
Int. j. morphol ; 38(1): 165-175, Feb. 2020. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1056416

RESUMEN

An alternative hyper-ovulator inducer to replace clomiphene citrate (CC) is needed as it is unsuitable for women with polycystic ovarian syndrome and is associated with low pregnancy rates. Anastrozole is an effective hyper-ovulator inducer, but has not been well researched. In order to determine the effectiveness of anastrozole as a hyper-ovulator inducer and to an extent compare it with CC in similar situations, this study ascertained the effects of these drugs on the expression of the focal adhesion proteins, paxillin and FAK, which are uterine receptivity markers in the surface luminal uterine epithelial cells of day 1 and day 6 pregnant Wistar rats. The results show that paxillin is localized in focal adhesions at the base of the uterine epithelial cells at day 1 of pregnancy whereas at day 6, paxillin disassembles from the basal focal adhesions and localizes and increases its expression apically. FAK is faintly expressed at the basal aspect of the uterine epithelial cells while moderately expressed at the cell-to-cell contact at day 1 in all groups from where it disassembles and relocates apically and becomes more intensely expressed at day 6 of pregnancy in untreated and anastrozole treated rats. Although paxillin is localized apically at day 6, its expression is significantly down-regulated with CC treatment suggesting its interference with the implantation process. These findings seem to suggest that anastrozole could favor implantation.


Para reemplazar el citrato de clomifeno (CC) es necesario un inductor de hiperovulación alternativo, ya que no es adecuado para mujeres con síndrome de ovario poliquístico y está asociado con tasas bajas de embarazo. El anastrozol es un inductor eficaz del hiper-ovulador, pero no se ha investigado adecuadamente. Con el fin de determinar la efectividad del anastrozol como inductor del hiper-ovulador y, en cierta medida, compararlo con CC en situaciones similares, este estudio determinó los efectos de estos fármacos en la expresión de las proteínas de adhesión focal, paxillin y FAK, uterinas marcadores de receptividad en la superficie luminal de células uterinas epiteliales, del día 1 y día 6 en ratas Wistar preñadas. Los resultados muestran que la paxilina se localiza en adherencias focales en la base de las células epiteliales uterinas en el día 1 del embarazo, mientras que en el día 6, la paxilina se desmonta de las adherencias focales basales y localiza y aumenta su expresión apicalmente. FAK se expresa débilmente en el aspecto basal de las células epiteliales uterinas, mientras que se expresa moderadamente en el contacto de célula a célula en el día 1 en todos los grupos, donde se separa y se reubica apicalmente y se expresa con mayor intensidad el día 6 de la preñez, en pacientes no tratados y tratados. ratas tratadas con anastrozol. Aunque la paxillina se localiza apicalmente en el día 6, su expresión está significativamente disminuida con el tratamiento con CC, lo que sugiere su interferencia con el proceso de implantación. Estos hallazgos sugieren que el anastrozol podría favorecer el proceso de implantación.


Asunto(s)
Animales , Femenino , Ratas , Útero/efectos de los fármacos , Anastrozol/farmacología , Ovulación/efectos de los fármacos , Ratas Wistar , Adhesiones Focales/efectos de los fármacos , Epitelio/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Paxillin/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Microscopía Fluorescente
6.
J Dent Res ; 94(6): 853-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724555

RESUMEN

Mineral trioxide aggregate is the currently recommended material of choice for clinical pulp repair despite several disadvantages, including handling inconvenience. Little is known about the signaling mechanisms involved in bioceramic-mediated dental pulp repair-particularly, dental pulp cell (DPC) migration. This study evaluated the effects of iRoot BP Plus, a novel ready-to-use nanoparticulate bioceramic putty, on DPC migration in vitro and pulp repair in vivo, focusing on possible involvement of fibroblast growth factor receptor (FGFR)-related signaling, including mitogen-activated protein kinase and Akt pathways. Treatment with iRoot BP Plus extracts enhanced horizontal and vertical migration of DPCs, which was comparable with the effects induced by mineral trioxide aggregate extracts. The DPCs exposed to iRoot BP Plus extracts demonstrated no evident apoptosis. Importantly, treatment with iRoot BP Plus extracts resulted in rapid activation of FGFR, p38 mitogen-activated protein kinase, extracellular signal-regulated kinase (ERK) 1/2, c-Jun-N-terminal kinase (JNK), and Akt signaling in DPCs. Confocal immunofluorescence staining revealed that iRoot BP Plus stimulated focal adhesion formation and stress fiber assembly in DPCs, in addition to upregulating the expression of focal adhesion molecules, including p-focal adhesion kinase, p-paxillin, and vinculin. Moreover, activation of FGFR, ERK, JNK, and Akt were found to mediate the upregulated expression of focal adhesion molecules, stress fiber assembly, and enhanced DPC migration induced by iRoot BP Plus. Consistent with the in vitro results, we observed induction of homogeneous dentin bridge formation and expression of p-focal adhesion kinase, p-FGFR, p-ERK 1/2, p-JNK, and p-Akt near injury sites by iRoot BP Plus in an in vivo pulp repair model. These data demonstrate that iRoot BP Plus can promote DPC migration and pulp repair involving the FGFR-mediated ERK 1/2, JNK, and Akt pathways. These findings provide valuable insights into the signaling mechanisms underlying nanoparticulate bioceramic-mediated pulp repair.


Asunto(s)
Compuestos de Calcio/farmacología , Cerámica/química , Pulpa Dental/citología , Materiales de Recubrimiento Pulpar y Pulpectomía/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Silicatos/farmacología , Compuestos de Aluminio/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Pulpa Dental/efectos de los fármacos , Dentina Secundaria/efectos de los fármacos , Dentinogénesis/efectos de los fármacos , Combinación de Medicamentos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ensayo de Materiales , Proteínas de la Membrana/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Nanopartículas/química , Óxidos/farmacología , Paxillin/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Ratas , Fibras de Estrés/efectos de los fármacos , Vinculina/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
7.
J Endod ; 40(10): 1642-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25260738

RESUMEN

INTRODUCTION: The objective of this study was to compare the cytotoxicity, inflammatory response, osteogenic effect, and the signaling mechanism of these biologic activities of 4 calcium compound-based root canal sealers (ie, Sealapex [Sybron Kerr, WA], apatite root sealer [ARS; Dentsply Sankin, Tokyo, Japan], MTA Fillapex [Angelus Indústria de Produtos Odontológicos S/A, Londrina, PR, Brazil], and iRoot SP [Innovative BioCreamix Inc, Vancouver, Canada]) in human periodontal ligament cells. METHODS: Cytotoxicity was assessed using the 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide assay. Levels of inflammatory mediators were measured by enzyme-linked immunosorbent assay, reverse-transcription polymerase chain reaction, and Western blot analysis. Osteogenic potential was evaluated by alkaline phosphatase activity, alizarin red staining, and marker genes by reverse-transcription polymerase chain reaction. The signal transduction pathways were examined by Western blotting. RESULTS: None of the sealers were cytotoxic. ARS, MTA Fillapex, and iRoot SP induced a lower expression of proinflammatory mediators than Sealapex. All sealers increased ALP activity and the formation of mineralized nodules and up-regulated the expression of osteoblastic marker messenger RNA. ARS, MTA Fillapex, and iRoot SP showed superior osteogenic potential compared with Sealapex. The expression and/or activation of integrin receptors and downstream signaling molecules, including focal adhesion kinase, paxillin, Akt, mitogen-activated protein kinase, and nuclear factor κB, was induced by ARS, MTA Fillapex, and iRoot SP treatment but not by Sealapex treatment. CONCLUSIONS: We show for the first time that ARS, MTA Fillapex, and iRoot SP induce a lower expression of inflammatory mediators and enhance osteoblastic differentiation of PDLCs via the integrin-mediated signaling pathway compared with Sealapex.


Asunto(s)
Materiales Biocompatibles/farmacología , Mediadores de Inflamación/farmacología , Osteogénesis/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/farmacología , Fosfatasa Alcalina/análisis , Fosfatasa Alcalina/efectos de los fármacos , Compuestos de Aluminio/farmacología , Compuestos de Aluminio/toxicidad , Antraquinonas , Materiales Biocompatibles/toxicidad , Compuestos de Calcio/farmacología , Compuestos de Calcio/toxicidad , Hidróxido de Calcio/farmacología , Hidróxido de Calcio/toxicidad , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colorantes , Combinación de Medicamentos , Durapatita/farmacología , Durapatita/toxicidad , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Humanos , Mediadores de Inflamación/toxicidad , Ensayo de Materiales , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Nanopartículas , Osteoblastos/efectos de los fármacos , Óxidos/farmacología , Óxidos/toxicidad , Paxillin/efectos de los fármacos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/toxicidad , Salicilatos/farmacología , Salicilatos/toxicidad , Transducción de Señal/efectos de los fármacos , Silicatos/farmacología , Silicatos/toxicidad , Sales de Tetrazolio , Tiazoles
8.
Toxicology ; 294(1): 42-9, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22306966

RESUMEN

Statins reduce the isoprenoids farnesyl and geranylgeranyl pyrophosphate, essential intermediates, which control a diversity of cellular events such as cytoskeleton integrity, adhesion, migration and viability. Cardiac fibroblasts are the major non-myocyte cell constituent in the normal heart, and play a key role in the maintenance of extracellular matrix. The effects of simvastatin on cardiac fibroblast processes previously mentioned remain unknown. Our aims were to investigate the effects of simvastatin on cytoskeleton structure and focal adhesion complex assembly and their relationships with cell adhesion, migration and viability in cultured cardiac fibroblasts. To this end, cells were treated with simvastatin for 24 h and changes in actin cytoskeleton, levels of vimentin and paxillin as well as their subcellular localization were analyzed by Western blot and immunocytochemistry, respectively. Cell adhesion to plastic or collagen coated dishes, migration in Transwell chambers, and cell viability were analyzed after simvastatin treatment. Our results show that simvastatin disrupts actin cytoskeleton and focal adhesion complex evaluated by phalloidin stain and immunocytochemistry for paxillin and vinculin. All these effects occurred by a cholesterol synthesis-independent mechanism. Simvastatin decreased cell adhesion, migration and viability in a concentration-dependent manner. Finally, simvastatin decreased angiotensin II-induced phospho-paxillin levels and cell adhesion. We concluded that simvastatin disrupts cytoskeleton integrity and focal adhesion complex assembly in cultured cardiac fibroblasts by a cholesterol-independent mechanism and consequently decreases cell migration, adhesion and viability.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Miofibroblastos/efectos de los fármacos , Simvastatina/efectos adversos , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Miofibroblastos/fisiología , Paxillin/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Simvastatina/antagonistas & inhibidores , Terpenos/farmacología , Vinculina/efectos de los fármacos
10.
Exp Cell Res ; 313(9): 1886-96, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17428471

RESUMEN

Valproic acid (VPA), a mood stabilizer and anticonvulsant, has a variety of neurotrophic functions; however, less is known about how VPA regulates neurite outgrowth. Here, using N1E-115 neuroblastoma cells as the model, we show that VPA upregulates Gadd45a to trigger activation of the downstream JNK cascade controlling neurite outgrowth. VPA induces the phosphorylation of c-Jun N-terminal kinase (JNK) and the substrate paxillin, while VPA induction of neurite outgrowth is inhibited by JNK inhibitors (SP600125 and the small JNK-binding peptide) or a paxillin construct harboring a Ser 178-to-Ala mutation at the JNK phosphorylation. Transfection of Gadd45a, acting through the effector MEKK4, leads to the phosphorylation of the JNK cascade. Conversely, knockdown of Gadd45a with siRNA reduces the effect of VPA. Taken together, these results suggest that upregulation of Gadd45a explains one of the mechanisms whereby VPA induces the neurotrophic effect, providing a new role of Gadd45a in neurite outgrowth.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Paxillin/metabolismo , Ácido Valproico/farmacología , Animales , Antimaníacos/farmacología , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/genética , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 4/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 4/metabolismo , Ratones , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuroblastoma , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Paxillin/efectos de los fármacos , Paxillin/genética , Fosforilación/efectos de los fármacos , Interferencia de ARN/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
11.
J Cell Biochem ; 99(1): 88-95, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16598791

RESUMEN

Spreading of SNU16mAd gastric carcinoma cells was previously shown to be regulated via a signaling network from transforming growth factor beta1 (TGFbeta1) to integrins signaling, through a mediation of protein kinase C delta (PKCdelta). However, in the previous study, the roles of PKCdelta appeared complicated. In this study to clarify the roles of PKCdelta in the spreading of the gastric carcinoma cells, we questioned if PKC activation via phorbol 12-myristate 13-acetate (PMA) treatment could mimic the TGFbeta1 effects. An acute PMA treatment increased phosphorylations of focal adhesion (FA) kinase, paxillin, c-Src, and cofilin, just as TGFbeta1 did. Furthermore, cell spreading mediated by TGFbeta1- or acute PMA treatment correlated with activation of RhoA, which regulates actin reorganization and FA formation. However, stress fiber formation was prominent in TGFbeta1-treated cells, compared to cortical actin organization in PMA-treated cells. Altogether, these observations indicate that acute PMA treatment could mimic the TGFbeta1 mechanisms for cell spreading through subtly different effects on actin reorganization.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Integrinas/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Proteína Tirosina Quinasa CSK , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Cofilina 1/efectos de los fármacos , Cofilina 1/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrinas/efectos de los fármacos , Imitación Molecular , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Fosforilación , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1 , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas
12.
J Cell Sci ; 118(Pt 23): 5513-24, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16291724

RESUMEN

The carcinoembryonic antigen-related cell adhesion molecule CEACAM1 (CD66a) and the scaffolding protein filamin A have both been implicated in tumor cell migration. In the present study we identified filamin A as a novel binding partner for the CEACAM1-L cytoplasmic domain in a yeast two-hybrid screen. Direct binding was shown by surface plasmon resonance analysis and by affinity precipitation assays. The association was shown for human and rodent CEACAM1-L in endogenous CEACAM1-L expressing cells. To address functional aspects of the interaction, we used a well-established melanoma cell system. We found in different migration studies that the interaction of CEACAM1-L and filamin A drastically reduced migration and cell scattering, whereas each of these proteins when expressed alone, acted promigratory. CEACAM1-L binding to filamin A reduced the interaction of the latter with RalA, a member of the Ras-family of GTPases. Furthermore, co-expression of CEACAM1-L and filamin A led to a reduced focal adhesion turnover. Independent of the presence of filamin A, the expression of CEACAM1-L led to an increased phosphorylation of focal adhesions and to altered cytoskeletal rearrangements during monolayer wound healing assays. Together, our data demonstrate a novel mechanism for how CEACAM1-L regulates cell migration via its interaction with filamin A.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteínas Contráctiles/fisiología , Proteínas de Microfilamentos/fisiología , Animales , Antígenos CD/genética , Antígenos CD/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/farmacología , Línea Celular Tumoral , Proteínas Contráctiles/genética , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Filaminas , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas de Microfilamentos/genética , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Fosforilación , Unión Proteica , Ratas , Técnicas del Sistema de Dos Híbridos , Tirosina/efectos de los fármacos , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda