Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Med Sci Monit ; 28: e935055, 2022 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-35277469

RESUMEN

BACKGROUND Tongue cancer is the most prevalent of head and neck squamous cell carcinomas, including base of tongue cancer (BOT) and oral squamous cell carcinoma of the mobile tongue (OTSCC). We aimed to investigate the role of RIPOR3 in tumorigenesis and its development as a potential prognostic biomarker for tongue cancer, especially OTSCC. MATERIAL AND METHODS Associations of expression, clinical pathologic features, and overall survival were analyzed by logistic regression, multivariate Cox analysis, and Kaplan-Meier methods. Gene set enrichment analysis (GSEA) and the CIBERSORT algorithm were performed to determine the correlation between RIPOR3 and tumor immune infiltration. cBioPortal was used for methylation and copy number variation (CNV) analysis. The Human Protein Atlas (HPA) and GSE31056 dataset were used for further external validation. RESULTS RIPOR3 expression in OTSCC was significantly associated with various clinicopathological parameters. Kaplan-Meier survival analysis showed that OTSCC with low RIPOR3 expression had a worse prognosis than that with high RIPOR3 expression. Multivariate analysis revealed that lower RIPOR3 expression was an independent prognostic factor for poor prognosis. GSEA and Neighbor Gene Network analysis showed RIPOR3 expression was related with the modulation and function of the immune-related pathway. Methylation level and CNV analysis showed that the downregulated expression of RIPOR3 was significantly related to hypermethylation but not to CNV. Finally, high RIPOR3 expression was validated at the protein level using the HPA database and GSE31056 dataset. CONCLUSIONS These findings suggested that RIPOR3 might serve as a promising prognostic biomarker and is related to the immune cell infiltration of OTSCC.


Asunto(s)
Carcinogénesis/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/genética , ARN Neoplásico/genética , Proteínas de Unión al GTP rac/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Variaciones en el Número de Copia de ADN , Metilación de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Neoplasias de la Lengua/genética , Proteínas de Unión al GTP rac/biosíntesis
2.
DNA Cell Biol ; 40(3): 469-481, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33600260

RESUMEN

Bladder cancer (BC) is a common genitourinary malignancy worldwide. However, the molecular pathogenesis of BC remains unclear. The current study conducted bioinformatic analyses to discover key genes involved in BC progression. A total of 375 differentially expressed genes (DEGs) were screened in the GEO database and The Cancer Genome Atlas (TCGA) database, which were further evaluated by the core level in the protein-protein interaction network. RAC3 (Rac family small GTPase 3), one of the top hub genes, was focused on for its gene expression and prognostic value in BC. Immunohistochemical assays indicated elevated RAC3 levels in BC tissues compared with normal tissues. Overexpression of RAC3 expression was closely associated with poor differentiation (p = 0.035), advanced TNM stage (p = 0.014), lymph metastasis (p = 0.033), and recurrence (p < 0.001). Kaplan-Meier and Cox proportional hazards analyses demonstrated that high RAC3 expression indicated poor survival of BC patients, which could serve as an independent prognostic factor for overall survival (HR = 3.159, p = 0.023) and disease-free survival (HR = 4.633, p = 0.002). Moreover, bioinformatic analyses indicated that RAC3 might be correlated with malignant phenotypes and immune infiltration of BC. Taken together, RAC3 could be a novel prognostic biomarker for BC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Vejiga Urinaria , Proteínas de Unión al GTP rac/biosíntesis , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Estadificación de Neoplasias , Tasa de Supervivencia , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/mortalidad , Proteínas de Unión al GTP rac/genética
3.
Appl Immunohistochem Mol Morphol ; 28(2): 111-122, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32044879

RESUMEN

Genome-wide screening of transcriptional changes among normal, cancer, and nodal metastases provides insights into the molecular basis of breast cancer (BC) progression and metastasis. To identify transcriptional changes and differentially expressed genes (DEGs) in the metastatic progression of BC and to determine the prognostic role of these DEGs in clinical outcome, we compared transcriptome profiling in matched normal, cancer, and lymph node metastatic tissues of 7 patients with estrogen receptor-positive, HER2-negative BC by using massive parallel RNA sequencing. The global profiles of gene expression in cancer and nodal metastases were highly correlated (r=0.962, P<0.001). In 6 (85.8%) patients, cancer and corresponding nodal metastases from the same patient clustered together. We identified 1522 and 664 DEGs between normal and cancer and between cancer and nodal metastases, respectively. The DEGs in normal versus cancer and cancer versus nodal metastases were significantly clustered in 1 and 8 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, respectively. The chemokine signaling pathway was the most significant pathway in the cancer-to-nodal metastasis transition (false discovery rate=2.15E-13). The expression of 2 dysregulated RAC2 and PTGDS genes was confirmed by quantitative real-time polymerase chain reaction and immunohistochemistry. Interestingly, the lower RAC2 and PTGDS expression were associated with significantly worse disease-free survival in patients with BC. Our results show a high concordance of gene expression in BC and their nodal metastases, and identify DEGs associated with the metastatic progression of BC. The DEGs identified in this study represent novel biomarkers for predicting the prognosis of patients with BC.


Asunto(s)
Neoplasias de la Mama , Regulación Neoplásica de la Expresión Génica , Oxidorreductasas Intramoleculares/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas de Unión al GTP rac/biosíntesis , Adulto , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Oxidorreductasas Intramoleculares/genética , Metástasis Linfática , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Valor Predictivo de las Pruebas , Tasa de Supervivencia , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
4.
Oxid Med Cell Longev ; 2019: 5254798, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281584

RESUMEN

Radioresistance is the major obstacle in the radiotherapy of the malignant melanoma. Thus, it is of importance to increase the radiosensitivity of melanoma cells. In the present study, the radioresistant melanoma cell line OCM-1 with inducible overexpression of Ras-related C3 botulinum toxin substrate 2 was established based on a radiation-inducible early growth response gene (Egr-1) promoter. The effects of Ras-related C3 botulinum toxin substrate 2 overexpression on the radiosensitivity of melanoma cells exposed to either X-rays or carbon ion beams were evaluated in cultured cells as well as xenograft tumor models. In addition, both reactive oxygen species yield and the NADPH oxidase activity were measured in the irradiated melanoma cells. It was found that the radiation-inducible overexpression of Ras-related C3 botulinum toxin substrate 2 sensitized the melanoma cells to both X-rays and carbon ion irradiation by enhancing the NADPH oxidase activity and the subsequent reactive oxygen species production. Besides, the overexpression of Ras-related C3 botulinum toxin substrate 2 enhanced the tumor-killing effect of radiotherapy in xenograft tumors significantly. The results of this study indicate that Ras-related C3 botulinum toxin substrate 2 is promising in increasing the radiosensitivity of melanoma cells, which provides experimental evidence and theoretical basis for clinical radiosensitization of the malignant melanoma.


Asunto(s)
Melanoma/metabolismo , Melanoma/radioterapia , Proteínas de Unión al GTP rac/biosíntesis , Animales , Toxinas Botulínicas/metabolismo , Línea Celular Tumoral , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Tolerancia a Radiación/fisiología , Proteína RCA2 de Unión a GTP
5.
PLoS One ; 14(3): e0213529, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30845261

RESUMEN

Plasmodium falciparum severe malaria causes more than 400,000 deaths every year. One feature of P. falciparum-parasitized erythrocytes (pRBC) leading to cerebral malaria (CM), the most dangerous form of severe malaria, is cytoadherence to endothelium and blockage of the brain microvasculature. Preventing ligand-receptor interactions involved in this process could inhibit pRBC sequestration and insurgence of severe disease whilst reversing existing cytoadherence could be a saving life adjunct therapy. Increasing evidence indicate the endothelial Rho signaling as a crucial player in malaria parasite cytoadherence. Therefore, we have used the cytotoxic necrotizing factor 1 (CNF1), an Escherichia coli protein able to modulate the activity of Cdc42, Rac, and Rho, three subfamilies of the Rho GTPases family, to study interactions between infected erythrocytes and cerebral endothelium in co-culture models. The main results are that CNF1 not only prevents cytoadherence but, more importantly, induces the detachment of pRBCs from endothelia monolayers. We first observed that CNF1 does affect neither parasite growth, nor the morphology and concentration of knobs that characterize the parasitized erythrocyte surface, as viewed by scanning electron microscopy. On the other hand, flow cytometry experiments show that cytoadherence reversion induced by CNF1 occurs in parallel with a decreased ICAM-1 receptor expression on the cell surface, suggesting the involvement of a toxin-promoted endocytic activity in such a response. Furthermore, since the endothelial barrier functionality is compromised by P. falciparum, we conducted a permeability assay on endothelial cells, revealing the CNF1 capacity to restore the brain endothelial barrier integrity. Then, using pull-down assays and inhibitory studies, we demonstrated, for the first time, that CNF1 is able not only to prevent but also to cause the parasite detachment by simultaneously activating Rho, Rac and Cdc42 in endothelial cells. All in all our findings indicate that CNF1 may represent a potential novel therapeutic strategy for preventing neurological complications of CM.


Asunto(s)
Toxinas Bacterianas/farmacología , Adhesión Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas de Escherichia coli/farmacología , Escherichia coli/química , Plasmodium falciparum/metabolismo , Toxinas Bacterianas/química , Línea Celular , Células Endoteliales/parasitología , Células Endoteliales/patología , Proteínas de Escherichia coli/química , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Proteína de Unión al GTP cdc42/biosíntesis , Proteínas de Unión al GTP rac/biosíntesis
6.
Mol Med Rep ; 18(1): 333-341, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749511

RESUMEN

Brain ischemia leads to energy depletion, mitochondrial dysfunction and neuronal cell death. The present study was designed to identify key genes and pathways associated with brain ischemia. The gene expression profile GSE52001, including 3 normal brain samples and 3 cerebral ischemia samples, was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using the limma package. Then functional and pathway enrichment analyses were performed by the MATHT tool. Protein­protein interaction (PPI) network, module selection and microRNA (miRNA)­target gene network were constructed utilizing Cytoscape software. A total of 488 DEGs were identified (including 281 upregulated and 207 downregulated genes). In the PPI network, Rac family small GTPase 2 (RAC2) had higher degrees. RAC2 was significantly enriched in the FcγR­mediated phagocytosis pathway. miR­29A/B/C had a higher degree in the miRNA­target gene network. Insulin like growth factor 1 (Igf1) was identified as the target gene for miR­29A/B/C. RAC2 may function in brain ischemia through mediating the FcγR­mediated phagocytosis pathway. Meanwhile, miR­29A/B/C and their targets gene Igf1 may serve important roles in the development and progression of brain ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Biología Computacional , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/patología , MicroARNs/biosíntesis , MicroARNs/genética , Ratas , Ratas Sprague-Dawley , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
7.
J Biol Chem ; 290(10): 6408-18, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25586182

RESUMEN

Engulfment and cell motility 1/dedicator of cytokinesis 180 (Elmo1/Dock180) is a bipartite guanine nucleotide exchange factor for the monomeric GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1). Elmo1/Dock180 regulates Rac1 activity in a specific spatiotemporal manner in endothelial cells (ECs) during zebrafish development and acts downstream of the Netrin-1/Unc5-homolog B (Unc5B) signaling cascade. However, mechanistic details on the pathways by which Elmo1/Dock180 regulates endothelial function and vascular development remained elusive. In this study, we aimed to analyze the vascular function of Elmo1 and Dock180 in human ECs and during vascular development in zebrafish embryos. In vitro overexpression of Elmo1 and Dock180 in ECs reduced caspase-3/7 activity and annexin V-positive cell number upon induction of apoptosis. This protective effect of Elmo1 and Dock180 is mediated by activation of Rac1, p21-activated kinase (PAK) and AKT/protein kinase B (AKT) signaling. In zebrafish, Elmo1 and Dock180 overexpression reduced the total apoptotic cell and apoptotic EC number and promoted the formation of blood vessels during embryogenesis. In conclusion, Elmo1 and Dock180 protect ECs from apoptosis by the activation of the Rac1/PAK/AKT signaling cascade in vitro and in vivo. Thus, Elmo1 and Dock180 facilitate blood vessel formation by stabilization of the endothelium during angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Vasos Sanguíneos/metabolismo , Neovascularización Fisiológica , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/genética , Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética
8.
Mol Pharmacol ; 87(3): 378-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25488183

RESUMEN

Gefitinib is widely used for the treatment of lung cancer in patients with sensitizing epidermal growth factor receptor mutations, but patients tend to develop resistance after an average of 10 months. Low molecular weight heparins, such as enoxaparin, potently inhibit experimental metastasis. This study aimed to determine the potential of combined enoxaparin and gefitinib (enoxaparin + gefitinib) treatment to inhibit tumor resistance to gefitinib both in vitro and in vivo. A549 and H1975 cell migration was analyzed in wound closure and Transwell assays. Akt and extracellular signal-related kinase 1/2 signaling pathways were identified, and a proteomics analysis was conducted using SDS-PAGE/liquid chromatography-tandem mass spectrometry analysis. Molecular interaction networks were visualized using the Cytoscape bioinformatics platform. Protein expression of dedicator of cytokinesis 1 (DOCK1) and cytoskeleton intermediate filament vimentin were identified using an enzyme-linked immunosorbent assay, Western blot, and small interfering RNA transfection of A549 cells. In xenograft A549-luc-C8 tumors in nude mice, enoxaparin + gefitinib inhibited tumor growth and reduced lung colony formation compared with gefitinib alone. Furthermore, the combination had stronger inhibitory effects on cell migration than either agent used individually. Additional enoxaparin administration resulted in better effective inhibition of Akt activity compared with gefitinib alone. Proteomics and network analysis implicated DOCK1 as the key node molecule. Western blot verified the effective inhibition of the expression of DOCK1 and vimentin phosphorylation by enoxaparin + gefitinib compared with gefitinib alone. DOCK1 knockdown confirmed its role in cell migration, Akt expression, and vimentin phosphorylation. Our data indicate that enoxaparin sensitizes gefitinib antitumor and antimigration activity in lung cancer by suppressing DOCK1 expression, Akt activity, and vimentin phosphorylation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Enoxaparina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/administración & dosificación , Vimentina/metabolismo , Proteínas de Unión al GTP rac/biosíntesis , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Gefitinib , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rac/antagonistas & inhibidores
9.
Anticancer Res ; 34(12): 7111-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25503138

RESUMEN

BACKGROUND: Resistance to chemotherapeutic agents, as well as enhanced metastasis, have been frequently reported in lung cancer. MATERIALS AND METHODS: Cytotoxicity and proliferative effects of cisplatin on H460 lung cancer cells were evaluated by the MTT assay. Migration capacity was evaluated by the wound healing assay. The number of filopodia per cell were detected by rhodamine-phalloidin staining assay. The changes of protein levels of integrins, and migration-related proteins in response to cisplatin at sub-toxic concentrations were determined by western blotting. RESULTS: Herein we demonstrate for the first time that exposure to low concentrations of cisplatin results in increase of cell motility with the alteration of integrin expression. Cisplatin-treated cells exhibited a significant increase in the number of filopodia per cell in correlation with enhanced migration. Migration regulatory proteins, namely activated forms of focal-adhesion kinase (FAK) and ATP-dependent tyrosine kinase (AKT), were found to significantly be up-regulated in cisplatin-treated cells in comparison to those of the non-treated control. Active Rho A-GTP and Rac-GTP were found to be increased in accordance with activation of FAK/AKT signals. Furthermore, we found that such migration enhancement may be in part due to the integrin switch mediated by cisplatin treatment. Cisplatin induced a dramatic alteration in the integrin expression pattern by up-regulating integrin α4, αv, ß1, and ß5 which were previously reported to increase cell motility, while it had no effect on integrin α5, and ß3. CONCLUSION: As the integrin switch is a hallmark of highly aggressive cancer, these findings may provide insights for better understanding of cancer cell adaptation after exposure to cisplatin.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Cisplatino/farmacología , Cadenas alfa de Integrinas/biosíntesis , Cadenas beta de Integrinas/biosíntesis , Neoplasias Pulmonares/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Humanos , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Seudópodos/fisiología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba , Proteínas de Unión al GTP rac/biosíntesis , Quinasas Asociadas a rho/biosíntesis
10.
Am J Physiol Cell Physiol ; 302(11): C1676-86, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22460711

RESUMEN

This study utilized the glutathione transferase (GST) pull-down assay to identify novel substrates of an osteoclastic protein-tyrosine phosphatase, PTP-oc. Consistent with the previous findings that the phosphorylated tyr-527 (pY527) of Src is a substrate of PTP-oc, the major protein pulled down with the phosphatase-deficient (PD)-PTP-oc-GST trapping mutant in RAW264.7 cells was Src. The GST-PD-PTP-oc also pulled down pY-Syk and pY-ß(3)-integrin, but not after PP2 pretreatment. However, PTP-oc transgenic osteoclasts or PTP-oc-overexpressing RAW264.7 cells had elevated, and not reduced, levels of pY525/526-Syk and pY759-ß(3) integrin, and the PTP-oc siRNA treatment drastically reduced levels of pY525/526 Syk and pY759-ß(3)-integrin in RAW264.7 cells. These findings are incompatible with the premise that they are substrates of PTP-oc. The PTP-oc-dependent increases in pY525/526-Syk and pY759-ß(3)-integrin levels were completely blocked by PP2, indicating that these effects are secondary to PTP-oc-mediated activation of the Src protein-tyrosine kinase (PTK). Overexpression of PTP-oc increased, and siRNA-mediated suppression of PTP-oc reduced, pY160-Vav1, pY173-Vav3, and pY783-PLCγ levels, and Rac1 activation, which are downstream mediators of the ITAM/Syk signaling. Overexpression of PTP-oc also increased, and PTP-oc siRNA treatment decreased, the pY-Shp1 levels, which were blocked by PP2. Since Shp1 is a negative regulator of osteoclast activity and is a key mediator of the ITIM signaling, these findings suggest that PTP-oc is an upstream suppressor of the ITIM/Shp1 signaling through PTP-oc-induced Src-dependent Shp1 phosphorylation. In summary, PTP-oc plays a central regulatory role in the concerted regulation of the ß(3)-integrin, the ITAM/Syk, and the ITIM/Shp1 signaling indirectly through activation of Src PTK.


Asunto(s)
Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Osteoclastos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Resorción Ósea , Línea Celular , Integrina beta3/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Neuropéptidos/biosíntesis , Neuropéptidos/metabolismo , Osteoclastos/fisiología , Fosfolipasa C gamma/biosíntesis , Fosforilación , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-vav/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Quinasa Syk , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
11.
Histopathology ; 59(6): 1163-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22175896

RESUMEN

AIMS: Dedicator of cytokinesis I (Dock180) is a novel guanine nucleotide exchange factor for Rho guanosine triphosphates (GTPases) important for cell migration. The aim of this study was to evaluate the role of Dock180 in ovarian carcinogenesis. METHODS AND RESULTS: Using immunohistochemistry, real-time polymerase chain reaction and Western blotting, overexpression of Dock180 RNA and protein was demonstrated in the nucleus and cytoplasm of ovarian cancer cell lines (n = 5) and clinical samples of ovarian borderline tumours (n = 21) and invasive cancers (n = 108) when compared with ovarian epithelial cell lines (n = 3) and benign cystadenomas (n = 10) (P < 0.05). High Dock180 cytoplasmic expression in ovarian cancer (n = 108) was associated significantly with serous histological type, high-grade cancer and advanced stage (P < 0.05), as well as poor overall and disease-free survival (P = 0.004). Using multivariate progression analysis, high Dock180 cytoplasmic expression and advanced cancer stage were found to be independent prognostic factors for short overall survival and disease-free survival (P < 0.05). Exogenous expression of Dock180 by transient transfection enhanced cancer cell migration and invasion, whereas knockdown of Dock180 by an siRNA approach retarded cancer cell migration and invasion in association with down-regulation of matrix metalloproteinase 2. CONCLUSIONS: Our findings suggest that Dock180 contributes to ovarian carcinogenesis and dissemination and is a potential prognostic marker and therapeutic target.


Asunto(s)
Carcinoma/enzimología , Cistoadenoma/enzimología , Neoplasias Ováricas/enzimología , Proteínas de Unión al GTP rac/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Western Blotting , Carcinoma/mortalidad , Carcinoma/patología , Movimiento Celular/genética , Cistoadenoma/mortalidad , Cistoadenoma/patología , Femenino , Humanos , Inmunohistoquímica , Microscopía Confocal , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Fenotipo , Pronóstico , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba , Adulto Joven , Proteínas de Unión al GTP rac/análisis
12.
Neurodegener Dis ; 8(6): 504-14, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21849797

RESUMEN

BACKGROUND: Because amyotrophic lateral sclerosis (ALS) is a progressive inflammatory disease, treatment of the pulmonary system plays a key role in ALS patients' care. Previous studies have mainly examined the pathological mechanism of ALS in the central nervous system; however, there has been relatively little research regarding the pulmonary system in ALS animal models. In inflammatory diseases, including asthma and arthritis, electroacupuncture (EA) is commonly used for its anti-inflammatory effects. The goal of this study was to determine whether EA treatment affects inflammation in the pulmonary system in an ALS animal model. METHODS: EA treatment at ST36 (Zusanli) acupoint was performed with 14-week-old hSOD1(G93A) transgenic mice. Immunohistochemical analysis was performed using anti-ionized calcium binding adaptor molecule 1 (Iba-1) and anti-tumor necrosis factor alpha (TNF-α) antibodies. To investigate the expression level of inflammatory proteins, Western blot analyses were performed using anti-Iba-1, anti-TNF-α, anti-nuclear factor kappa B (NF-κB), and anti-interleukin 6 (IL-6) antibodies. The activation of Ser435-phospho-specific RAC-alpha serine/threonine-protein kinase 1 (pAKT) and the increase of phosphorylated extracellular-signal-regulated kinases (pERK) protein in lung tissues of EA-treated and untreated hSOD1(G93A) mice were also evaluated by Western blot. RESULTS: EA treatment decreased the expression of the proinflammatory proteins such as TNF-α and IL-6, pNF-κB, and Iba-1 and increased the level of activated pAKT and pERK compared to control hSOD1(G93A) mice. CONCLUSIONS: Our findings suggest that EA could be an effective anti-inflammatory treatment for the respiratory impairment that occurs in ALS animal models.


Asunto(s)
Esclerosis Amiotrófica Lateral/complicaciones , Electroacupuntura , Inflamación/terapia , Enfermedades Respiratorias/etiología , Enfermedades Respiratorias/terapia , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Esclerosis Amiotrófica Lateral/patología , Animales , Western Blotting , Recuento de Células , Supervivencia Celular/fisiología , Proteínas del Citoesqueleto/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Femenino , Inmunohistoquímica , Interleucina-6/biosíntesis , Pulmón/metabolismo , Masculino , Ratones , Ratones Transgénicos , FN-kappa B/biosíntesis , Proteínas Nucleares/biosíntesis , Proteína Oncogénica v-akt/biosíntesis , Enfermedades Respiratorias/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas de Unión al GTP rac/biosíntesis
13.
Mol Cell Biol ; 31(11): 2227-40, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21444720

RESUMEN

We document a biphasic effect of Rac2 on the activation and inhibition of PLD2. Cells overexpressing Rac2 and PLD2 simultaneously show a robust initial (<10 min) response toward a chemoattractant that is later (>30 min) greatly diminished over PLD2-only controls. The first phase is due to the presence of a Rac2-PLD2 positive-feedback loop. To explain the mechanism for the Rac2-led PLD2 inhibition (the second phase), we used leukocytes from wild-type (WT) and Rac2(-/-) knockout mice. Rac2(-/-) cells displayed an enhanced PLD2 (but not PLD1) enzymatic activity, confirming the inhibitory role of Rac2. Late inhibitory responses on PLD2 due to Rac2 were reversed in the presence of phosphatidylinositol 4,5-bisphosphate (PIP(2)) both in vitro (purified GST-PH-PLD2, where GST is glutathione S-transferase and PH is pleckstrin homology) and in vivo. Coimmunoprecipitation and immunofluorescence microscopy indicated that PLD2 and Rac2 remain together. The presence of an "arc" of Rac2 at the leading edge of leukocyte pseudopodia and PLD2 physically posterior to this wave of Rac2 was observed in late chemotaxis. We propose Rac-led inhibition of PLD2 function is due to sterical interference of Rac with PLD2's PH binding site to the membrane and deprivation of the PIP(2). This work supports the importance of functional interactions between PLD and Rac in the biological response of cell migration.


Asunto(s)
Quimiotaxis , Macrófagos/fisiología , Fosfolipasa D/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Proteínas Sanguíneas , Quimiotaxis de Leucocito , Glutatión Transferasa/metabolismo , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Fosfatidilinositol 4,5-Difosfato/farmacología , Fosfolipasa D/biosíntesis , Fosfolipasa D/genética , Fosfoproteínas , Unión Proteica , Transducción de Señal , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
14.
J Immunol ; 186(3): 1467-76, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21178006

RESUMEN

G protein-coupled receptor (GPCR) activation elicits neutrophil responses such as chemotaxis and reactive oxygen species (ROS) formation, which depend on the small G protein Rac and are essential for host defense. P-Rex and Vav are two families of guanine-nucleotide exchange factors (GEFs) for Rac, which are activated through distinct mechanisms but can both control GPCR-dependent neutrophil responses. It is currently unknown whether they play specific roles or whether they can compensate for each other in controlling these responses. In this study, we have assessed the function of neutrophils from mice deficient in P-Rex and/or Vav family GEFs. We found that both the P-Rex and the Vav family are important for LPS priming of ROS formation, whereas particle-induced ROS responses and cell spreading are controlled by the Vav family alone. Surprisingly, fMLF-stimulated ROS formation, adhesion, and chemotaxis were synergistically controlled by P-Rex1 and Vav1. These responses were more severely impaired in neutrophils lacking both P-Rex1 and Vav1 than those lacking the entire P-Rex family, the entire Vav family, or both P-Rex1 and Vav3. P-Rex1/Vav1 (P1V1) double-deficient cells also showed the strongest reduction in fMLF-stimulated activation of Rac1 and Rac2. This reduction in Rac activity may be sufficient to cause the defects observed in fMLF-stimulated P1V1 neutrophil responses. Additionally, Mac-1 surface expression was reduced in P1V1 cells, which might contribute further to defects in responses involving integrins, such as GPCR-stimulated adhesion and chemotaxis. We conclude that P-Rex1 and Vav1 together are the major fMLFR-dependent Dbl family Rac-GEFs in neutrophils and cooperate in the control of fMLF-stimulated neutrophil responses.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Proteínas Proto-Oncogénicas c-vav/fisiología , Animales , Adhesión Celular/inmunología , Línea Celular , Quimiotaxis de Leucocito/inmunología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/biosíntesis , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-vav/deficiencia , Proteínas Proto-Oncogénicas c-vav/genética , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Proteínas de Unión al GTP rac/biosíntesis , Proteína de Unión al GTP rac1 , Proteína RCA2 de Unión a GTP
15.
Clin Exp Pharmacol Physiol ; 37(12): 1147-53, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20804509

RESUMEN

1. Diallyl disulphide (DADS) has potential as a chemopreventive and therapeutic agent. Previous studies have reported that Ras-related C3 botulinum toxin substrate 2 (Rac2), a regulatory subunit of the NADPH oxidase complex, is upregulated in DADS-induced apoptosis in human leukaemia HL-60 cells. The aim of the present study was to investigate the role of Rac2, NADPH oxidase and reactive oxygen species (ROS) in DADS-induced apoptosis. 2. Expression of the Rac2 gene along with that of five other genes of NADPH oxidase subunits were in HL-60 cells measured by Sybergreen quantitative real-time polymerase chain reaction. RNA interference was used to test the effect of Rac2. Protein expression was evaluated using western blot analysis and ROS levels were measured by 2',7'-dichlorofluorescein diacetate (DCFH-DA) fluorescence. DNA fragmentation and flow cytometry analysis were used to detect apoptotic cells. 3. Levels of Rac2 gene and protein were significantly upregulated and NADPH oxidase was activated in DADS-induced apoptosis. Pretreatment of HL-60 cells with small interfering (si) RNAs to inhibit Rac2 blocked DADS-induced apoptosis. Diallyl disulphide-induced intracellular ROS production was increased in phorbol myristate acetate-stimulated cells, but decreased in Rac2 siRNA-treated cells. In Rac2 siRNA-treated cells, activator protein-1 and caspase 3 levels decreased, c-myc protein levels were increased and p38 protein levels were unchanged compared with Rac2-competent, DADS-treated cells. 4. These results demonstrate that NADPH oxidase is the main source of DADS-induced ROS. In addition, Rac2 selectively activates the c-Jun N-terminal kinase pathway, but not the p38 pathway, in DADS-induced apoptosis. So, Rac2, NADPH oxidase and ROS have a critical role in DADS-induced apoptosis in human leukaemia HL-60 cells.


Asunto(s)
Compuestos Alílicos/farmacología , Apoptosis/efectos de los fármacos , Disulfuros/farmacología , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/farmacología , Apoptosis/fisiología , Caspasa 3/biosíntesis , Línea Celular Tumoral , Fragmentación del ADN , Citometría de Flujo , Células HL-60 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción AP-1/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína RCA2 de Unión a GTP
16.
Biochem Biophys Res Commun ; 399(4): 677-82, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20691665

RESUMEN

Apoptotic cell removal (efferocytosis) is an essential process in the regulation of inflammation and tissue repair. We have shown that monocyte chemoattractant protein-1/CC chemokine ligand 2 (MCP-1/CCL2) enhances efferocytosis by alveolar macrophages in murine bacterial pneumonia. However, the mechanism by which MCP-1 exerts this effect remains to be determined. Here we explored that hypothesis that MCP-1 enhances efferocytosis through a Rac1/phosphatidylinositol 3-kinase (PI3-kinase)-dependent mechanism. We assessed phagocytosis of apoptotic cells by MCP-1 treated murine macrophages in vitro and in vivo. Rac activity in macrophages was measured using a Rac pull down assay and an ELISA based assay (GLISA). The downstream Rac1 activation pathway was studied using a specific Rac1 inhibitor and PI3-kinase inhibitor in in vitro assays. MCP-1 enhanced efferocytosis of apoptotic cells by murine alveolar macrophages (AMs), peritoneal macrophages (PMs), the J774 macrophage cell line (J774s) in vitro, and murine AMs in vivo. Rac1 activation was demonstrated in these cell lines. The effect of MCP-1 on efferocytosis was completely negated by the Rac1 inhibitor and PI3-kinase inhibitor. We demonstrated that MCP-1 enhances efferocytosis in a Rac1-PI3 kinase-dependent manner. Therefore, MCP-1-Rac1-PI3K interaction plays a critical role in resolution of acute lung inflammation.


Asunto(s)
Apoptosis , Quimiocina CCL2/metabolismo , Macrófagos Alveolares/inmunología , Neuropéptidos/biosíntesis , Neumonía/inmunología , Proteínas de Unión al GTP rac/biosíntesis , Animales , Quimiocina CCL2/farmacología , Activación Enzimática , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Fagocitosis , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores CCR2/metabolismo , Proteínas Recombinantes/farmacología , Regulación hacia Arriba , Proteína de Unión al GTP rac1
17.
Cardiovasc Res ; 85(3): 484-93, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19755413

RESUMEN

AIMS: Sphingosine kinase 1 (SPHK1), its product sphingosine-1-phosphate (S1P), and S1P receptor subtypes have been suggested to play protective roles for cardiomyocytes in animal models of ischaemic preconditioning and cardiac ischaemia/reperfusion injury. To get more insight into roles for SPHK1 in vivo, we have generated SPHK1-transgenic (TG) mice and analysed the cardiac phenotype. METHODS AND RESULTS: SPHK1-TG mice overexpressed SPHK1 in diverse tissues, with a nearly 20-fold increase in enzymatic activity. The TG mice grew normally with normal blood chemistry, cell counts, heart rate, and blood pressure. Unexpectedly, TG mice with high but not low expression levels of SPHK1 developed progressive myocardial degeneration and fibrosis, with upregulation of embryonic genes, elevated RhoA and Rac1 activity, stimulation of Smad3 phosphorylation, and increased levels of oxidative stress markers. Treatment of juvenile TG mice with pitavastatin, an established inhibitor of the Rho family G proteins, or deletion of S1P3, a major myocardial S1P receptor subtype that couples to Rho GTPases and transactivates Smad signalling, both inhibited cardiac fibrosis with concomitant inhibition of SPHK1-dependent Smad-3 phosphorylation. In addition, the anti-oxidant N-2-mercaptopropyonylglycine, which reduces reactive oxygen species (ROS), also inhibited cardiac fibrosis. In in vivo ischaemia/reperfusion injury, the size of myocardial infarct was 30% decreased in SPHK1-TG mice compared with wild-type mice. CONCLUSION: These results suggest that chronic activation of SPHK1-S1P signalling results in both pathological cardiac remodelling through ROS mediated by S1P3 and favourable cardioprotective effects.


Asunto(s)
Miocardio/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Lisoesfingolípidos/fisiología , Animales , Fibrosis , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Neuropéptidos/biosíntesis , Quinolinas/farmacología , Receptores de Lisoesfingolípidos/análisis , Receptores de Esfingosina-1-Fosfato , Proteínas de Unión al GTP rac/biosíntesis , Proteína de Unión al GTP rac1 , Proteínas de Unión al GTP rho/biosíntesis , Proteína de Unión al GTP rhoA
18.
Am J Hypertens ; 22(2): 145-50, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19057515

RESUMEN

BACKGROUND: A combination of different types of antihypertensive drugs is widely used for the treatment of hypertension. We examined the inhibitory effects of a combination of an AT(1) receptor blocker (ARB), olmesartan, with various calcium channel blockers (CCBs) on inflammatory vascular remodeling. METHODS: Inflammatory vascular remodeling was induced by polyethylene-cuff placement around the femoral artery of C57BL/6J mice at 10 weeks of age. Olmesartan (0.5 mg/kg/day) was administered intraperitoneally using an osmotic minipump. CCBs (nifedipine 1.0 mg/kg/day, amlodipine 0.1 mg/kg/day, azelnidipine 0.1 mg/kg/day), and hydrochlorothiazide (HCTZ 0.5 mg/kg/day) were administered orally. RESULTS: In the injured artery, superoxide anion production and expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits p47(phox) and Rac-1 were markedly increased, together with expression of monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor (TNF)-alpha. Administration of a single drug alone at each concentration did not significantly inhibit these changes in the injured artery. However, a combination of olmesartan with various CCBs inhibited neointimal formation as well as oxidative stress and inflammatory markers in the injured artery. Moreover, among these CCBs, inhibition of these markers by olmesartan with azelnidipine was stronger than that caused by a combination with other CCBs. On the other hand, a combination of subeffective doses of olmesartan and HCTZ did not significantly affect vascular changes after cuff placement. CONCLUSIONS: These results suggest that the combination of ARB with CCB synergistically inhibits vascular remodeling and that the inhibitory actions of ARB on vascular remodeling may vary depending on the combined CCB.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Arteria Femoral/efectos de los fármacos , Imidazoles/farmacología , Tetrazoles/farmacología , Amlodipino/farmacología , Animales , Ácido Azetidinocarboxílico/análogos & derivados , Ácido Azetidinocarboxílico/farmacología , Quimiocina CCL2/biosíntesis , Dihidropiridinas/farmacología , Sinergismo Farmacológico , Arteria Femoral/lesiones , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/biosíntesis , Neuropéptidos/biosíntesis , Nifedipino/farmacología , Superóxidos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Túnica Íntima/efectos de los fármacos , Proteínas de Unión al GTP rac/biosíntesis , Proteína de Unión al GTP rac1
19.
Eye (Lond) ; 23(2): 461-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18849916

RESUMEN

PURPOSE: Allograft rejection is the main cause of graft failure in human corneal transplantation, for which underlying pathomechanism is not yet clear. We compared gene expression in the peripheral blood of patients who after undergoing corneal transplantation experienced graft rejection with those patients who accepted grafts. METHODS: Sixty-six patients who underwent corneal transplantation were studied including 18 patients who suffered subsequent graft rejection. cDNA array technology was used to survey and quantify transcript expression. A semiquantitative reverse transcriptase-PCR (RT-PCR) was used to confirm the gene expression pattern measured by a cDNA array of selected genes. RESULTS: Among 265 genes present on the array, eight genes were found to be differentially expressed. Four genes (Rac 2, RhoA, paxillin, and CD18) were further analysed by semiquantitative RT-PCR, and significant differences in mRNA expression levels in the rejection group were confirmed. CONCLUSIONS: Our study demonstrated that the expression of Rac2 mRNA was upregulated in the peripheral blood of patients experiencing corneal transplantation rejection compared to those patients who had no rejection episodes. In addition, three genes, RhoA, paxillin, and CD18, showed decreased expression in rejecting patients. cDNA array technology provides a potentially useful approach to identify novel genes that might participate in pathogenic pathways during corneal graft rejection.


Asunto(s)
Trasplante de Córnea , Rechazo de Injerto/diagnóstico , Proteínas de Unión al GTP rac/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Perfilación de la Expresión Génica/métodos , Rechazo de Injerto/inmunología , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Pronóstico , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Regulación hacia Arriba , Adulto Joven , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
20.
Biol Pharm Bull ; 31(5): 1003-7, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18451535

RESUMEN

In addition to humoral angiogenic factors, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), integrin-mediated adhesion of vascular endothelial cells to the extracellular matrix plays an important role in neovascularization. We recently found that TNIIIA2, a peptide derived from tenascin-C, induces functional activation of beta1 integrins. Here we investigated the effect of TNIIIA2 on vascular endothelial cell migration and proliferation, key processes for angiogenesis. TNIIIA2 was shown to activate beta1-integrins on human dermal microvascular endothelial cells (HDMEC). HDMEC adhered to fibronectin mainly via integrin alpha5beta1 and their haptotactic migration on that substrate was inhibited by TNIIIA2, in concomitant with a marked inhibition of Rac activation. TNIIIA2-treatment unaffected autophosphorylation of focal adhesion kinase (FAK), but induced its physical association with phospho-paxillin (Tyr118), suggesting the FAK/paxillin-dependent negative regulation of Rac activation. HDMEC proliferation on the fibronectin substrate was also inhibited by TNIIIA2-treatment, and this was accompanied either by an increase in the population of G 0/G1 cells and, conversely, a decrease in the population of S and G2/M cells or by dephosphorylation/inactivation of MAP-kinase (ERK1/2). Inhibited HDMEC migration and proliferation were both restored by pretreating the cells with a fibronectin peptide, FNIII14, which is capable of inactivating beta1-integrins. The chorioallantoic membrane assay demonstrated an antiangiogenic effect of TNIIIA2 in vivo. Thus, TNIIIA2 appears to negatively regulate angiogenesis by inhibiting migration and proliferation of endothelial cells. The ability to activate beta1-integrins may be responsible for the antiangiogenic effect of TNIIIA2, although it cannot be excluded the possibility that an additional mechanism(s) may play a role.


Asunto(s)
Inhibidores de la Angiogénesis , Integrina beta1/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Tenascina/farmacología , Animales , Biotransformación/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Citometría de Flujo , Humanos , Inmunoprecipitación , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Heridas y Lesiones/patología , Proteínas de Unión al GTP rac/biosíntesis , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...