Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Apoptosis ; 22(3): 449-462, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27864650

RESUMEN

The mechanisms by which oxidative stress induces spinal cord neuron death has not been completely understood. Investigation on the molecular signal pathways involved in oxidative stress-mediated neuronal death is important for development of new therapeutics for oxidative stress-associated spinal cord disorders. In current study we examined the role of heme oxygenase-1 (HO-1) in the modulation of MLK3/MKK7/JNK3 signaling, which is a pro-apoptotic pathway, after treating primary spinal cord neurons with H2O2. We found that MLK3/MKK7/JNK3 signaling was substantially activated by H2O2 in a time-dependent manner, demonstrated by increase of activating phosphorylation of MLK3, MKK7 and JNK3. H2O2 also induced expression of HO-1. Transduction of neurons with HO-1-expressing adeno-associated virus before H2O2 treatment introduced expression of exogenous HO-1 in neurons. Exogenous HO-1 reduced phosphorylation of MLK3, MKK7 and JNK3. Consistent with its inhibitory effect on MLK3/MKK7/JNK3 signaling, exogenous HO-1 decreased H2O2-induced neuronal apoptosis and necrosis. Furthermore, we found that exogenous HO-1 inhibited expression of Cdc42, which is crucial for MLK3 activation. In addition, HO-1-induced down-regulation of MLK3/MKK7/JNK3 signaling might be related to up-regulation of microRNA-137 (mir-137). A mir-137 inhibitor alleviated the inhibitory effect of HO-1 on JNK3 activation. This inhibitor also increased neuronal death even when exogenous HO-1 was expressed. Therefore, our study suggests a novel mechanism by which HO-1 exerted its neuroprotective efficacy on oxidative stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/fisiología , Peróxido de Hidrógeno/antagonistas & inhibidores , MAP Quinasa Quinasa 7/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Proteína Quinasa 10 Activada por Mitógenos/fisiología , Neuronas/patología , Transducción de Señal/efectos de los fármacos , Médula Espinal/citología , Proteína de Unión al GTP cdc42/fisiología , Animales , Apoptosis/fisiología , Células Cultivadas , Inducción Enzimática , Hemo Oxigenasa (Desciclizante)/genética , Peróxido de Hidrógeno/farmacología , MicroARNs/biosíntesis , MicroARNs/genética , MicroARNs/fisiología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Fosforilación , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Transducción Genética , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/genética , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
2.
J Cell Sci ; 128(15): 2866-80, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092936

RESUMEN

Dendritic cells are potent antigen-presenting cells endowed with the unique ability to initiate adaptive immune responses upon inflammation. Inflammatory processes are often associated with an increased production of serotonin, which operates by activating specific receptors. However, the functional role of serotonin receptors in regulation of dendritic cell functions is poorly understood. Here, we demonstrate that expression of serotonin receptor 5-HT7 (5-HT7R) as well as its downstream effector Cdc42 is upregulated in dendritic cells upon maturation. Although dendritic cell maturation was independent of 5-HT7R, receptor stimulation affected dendritic cell morphology through Cdc42-mediated signaling. In addition, basal activity of 5-HT7R was required for the proper expression of the chemokine receptor CCR7, which is a key factor that controls dendritic cell migration. Consistent with this, we observed that 5-HT7R enhances chemotactic motility of dendritic cells in vitro by modulating their directionality and migration velocity. Accordingly, migration of dendritic cells in murine colon explants was abolished after pharmacological receptor inhibition. Our results indicate that there is a crucial role for 5-HT7R-Cdc42-mediated signaling in the regulation of dendritic cell morphology and motility, suggesting that 5-HT7R could be a new target for treatment of a variety of inflammatory and immune disorders.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Receptores de Serotonina/metabolismo , Transducción de Señal/inmunología , Proteína de Unión al GTP cdc42/biosíntesis , Células 3T3 , Animales , Línea Celular , Quimiocina CCL19/metabolismo , Colon/citología , Colon/inmunología , Células Dendríticas/citología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/biosíntesis , Receptores CCR7/biosíntesis , Receptores de Serotonina/biosíntesis , Receptores de Serotonina/genética , Regulación hacia Arriba
3.
Neurochem Res ; 42(5): 1317-1324, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28097464

RESUMEN

Schwann cells (SCs) are unique glial cells in the peripheral nerve and may secrete multiple neurotrophic factors, adhesion molecules, extracellular matrix molecules to form the microenvironment of peripheral nerve regeneration, guiding and supporting nerve proliferation and migration. Cdc42 plays an important regulatory role in dynamic changes of the cytoskeleton. However, there is a little study referred to regulation and mechanism of Cdc42 on glial cells after peripheral nerve injury. The present study investigated the role of Cdc42 in the proliferation and migration of SCs after sciatic nerve injury. Cdc42 expression was tested, showing that the mRNA and protein expression levels of Cdc42 were significantly up-regulated after sciatic nerve injury. Then, we isolated and purified SCs from injuried sciatic nerve at day 7. The purified SCs were transfected with Cdc42 siRNA and pcDNA3.1-Cdc42, and the cell proliferation, cell cycle and migration were assessed. The results implied that Cdc42 siRNA remarkably inhibited Schwann cell proliferation and migration, and resulted in S phase arrest. While pcDNA3.1-Cdc42 showed a contrary effect. Besides, we also observed that Cdc42 siRNA down-regulated the protein expression of ß-catenin, Cyclin D1, c-myc and p-p38, which were up-regulated by pcDNA3.1-Cdc42. Meanwhile, the inhibitor of Wnt/ß-catenin and p38 MAPK signaling pathway IWP-2 and SB203580 significantly inhibited the effect of pcDNA3.1-Cdc42 on cell proliferation and migration. Overall, our data indicate that Cdc42 regulates Schwann cell proliferation and migration through Wnt/ß-catenin and p38 MAPK signaling pathway after sciatic nerve injury, which provides further insights into the therapy of the sciatic nerve injury.


Asunto(s)
Células de Schwann/fisiología , Neuropatía Ciática/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Células de Schwann/efectos de los fármacos , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética , Proteína de Unión al GTP cdc42/administración & dosificación , Proteína de Unión al GTP cdc42/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
4.
Med Mycol ; 55(4): 445-452, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-27664992

RESUMEN

Aspergillus fumigatus is the most common airborne pathogen causing fatal mycoses in immunocompromised patients. During the first 8 hours of development A. fumigatus conidia break dormancy, expand isotopically, establish an axis of polarity, and begin to extend germ tubes in a polar manner. The transition from isotropic to polar growth is critical for tissue invasion and pathogenesis. In the current work, we used two-color microarrays to examine the A. fumigatus transcriptome during early development, focusing on the isotropic to polar switch. The most highly regulated transcripts in the isotropic to polar switch did not include known polarity genes. Transcripts encoding the Cdc42 module, polarisome components, and septins, known to be critical players in polarity, showed relatively steady levels during the isotropic to polar switch. Indeed, these transcripts were present in dormant conidia, and their levels changed little from dormancy through germ tube emergence. Not only did the isotropic to polar switch show little change in the expression of key polarity genes of the Cdc42 module, polarisome, and septins, it also showed the lowest overall levels of both up- and downregulation in early development.


Asunto(s)
Aspergillus fumigatus/crecimiento & desarrollo , Aspergillus fumigatus/genética , Perfilación de la Expresión Génica , Complejos Multienzimáticos/análisis , Septinas/biosíntesis , Proteína de Unión al GTP cdc42/biosíntesis , Análisis por Micromatrices , Complejos Multienzimáticos/genética , Septinas/genética
5.
Dig Dis Sci ; 62(4): 958-967, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28181096

RESUMEN

BACKGROUND: Cell division cycle 42 (CDC42), an important member of the Rho family, is overexpressed in various human cancers. However, its expression and role in pancreatic cancer (PC) are not well understood. AIM: The present study was designed to investigate the expression patterns and underlying cellular mechanisms of CDC42 in PC. METHODS: First, immunohistochemical analysis, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to detect CDC42 expression in clinical pancreatic carcinoma and adjacent tissues. Second, differential expression of CDC42 between PC cells and normal cells was evaluated by qRT-PCR and Western blotting. Third, the correlation between CDC42 expression as well as clinicopathological characteristics and patient survival was analyzed. Finally, CDC42 was knocked down to examine its role both in vivo and in vitro. RESULTS: The results showed significantly increased CDC42 expression in pancreatic tumor tissues compared with adjacent normal tissues, as revealed by qRT-PCR, Western blotting and immunostaining. Compared to PanC-1 cells, CDC42 expression was downregulated in HPDE6-C7 cells as shown by qRT-PCR and Western blotting. High CDC42 expression was observed in 69.2% (83/120) of pancreatic adenocarcinoma patients and was significantly associated with tumor differentiation (p = 0.013), median tumor size (p = 0.005), tumor infiltration (pT stage, p = 0.04), lymph nodal status (pN stage, p = 0.044) and TNM staging (p = 0.003). Multivariate Cox regression analysis revealed CDC42 expression to be an independent predictor of survival of PC patients (HR 3.0, 95% CI 1.60-5.61, p = 0.001). Finally, we found that CDC42 promoted the proliferation of PanC-1 cells both in vivo and in vitro. CONCLUSIONS: Our findings reveal that CDC42 might play an important role in promoting PC development, and the findings suggest that CDC42 might serve as a potential prognostic indicator of PC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma Ductal Pancreático/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Anciano , Animales , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Transformada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Valor Predictivo de las Pruebas , Distribución Aleatoria , Resultado del Tratamiento , Proteína de Unión al GTP cdc42/genética
6.
Tumour Biol ; 36(2): 569-75, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25273174

RESUMEN

δ-Catenin is a member of the p120 catenin family. Similar to p120ctn, δ-catenin contains nine central Armadillo repeats and binds to the juxtamembrane domain (JMD) of E-cadherin. We used immunohistochemistry to detect δ-catenin expression in breast carcinoma (128 cases), and δ-catenin mRNA and protein expression was detected by reverse transcription-polymerase chain reaction and Western blotting (45 cases). The effects of δ-catenin on the activity of small GTPases and the biological behavior of breast cancer cells were explored by pulldown, flow cytometry, methyl thiazolyl tetrazolium, and Matrigel invasion assays. The results showed that δ-catenin expression increased in breast cancer tissues and was associated with a higher degree of malignancy (invasive lobular breast cancer, high tumor-node-metastasis stage, lymph node metastasis, and C-erbB-2+) and poor prognosis. Postoperative survival was shorter in patients with δ-catenin-positive expression than in patients with negative expression. δ-Catenin may regulate Cdc42/Rac1 activity, promote proliferation and invasion of breast cancer cells, and alter cell cycle progression. We conclude that δ-catenin tends to overexpress in breast carcinoma and promotes the malignant phenotype.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Cateninas/biosíntesis , Pronóstico , Adulto , Anciano , Neoplasias de la Mama/patología , Cateninas/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática/genética , Metástasis Linfática/patología , Células MCF-7 , Persona de Mediana Edad , Proteínas de Unión al GTP Monoméricas/biosíntesis , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis , Catenina delta
7.
Carcinogenesis ; 35(11): 2425-35, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24970760

RESUMEN

Metastatic disease is the leading cause of cancer mortality. Identifying biomarkers and regulatory mechanisms is important toward developing diagnostic and therapeutic tools against metastatic cancer. In this study, we demonstrated that podocalyxin-like 1 (PODXL) is overexpressed in breast tumor cells and increased in lymph node metastatic cancer. Mechanistically, we found that the expression of PODXL was associated with cell motility and invasiveness. Suppression of PODXL in MDA-MB-231 cells reduced lamellipodia formation and focal adhesion kinase (FAK) and paxillin phosphorylation. PODXL knockdown reduced the formation of invadopodia, such as inhibiting the colocalization of F-actin with cortactin and suppressing phosphorylation of cortactin and neural Wiskott-Aldrich syndrome protein. Conversely, overexpression of PODXL in MCF7 cells induced F-actin/cortactin colocalization and enhanced invadopodia formation and activation. Invadopodia activity and tumor invasion in PODXL-knockdown cells are similar to that in cortactin-knockdown cells. We further found that the DTHL motif in PODXL is crucial for regulating cortactin phosphorylation and Rac1/Cdc42 activation. Inhibition of Rac1/Cdc42 impeded PODXL-mediated cortactin activation and FAK and paxillin phosphorylation. Moreover, inhibition of PODXL in MDA-MB-231 cells significantly suppressed tumor colonization in the lungs and distant metastases, similar to those in cortactin-knockdown cells. These findings show that overexpression of PODXL enhanced invadopodia formation and tumor metastasis by inducing Rac1/Cdc42/cortactin signaling network.


Asunto(s)
Neoplasias de la Mama/genética , Cortactina/biosíntesis , Sialoglicoproteínas/biosíntesis , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis , Neoplasias de la Mama/patología , Cortactina/genética , Matriz Extracelular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Seudópodos/genética , Sialoglicoproteínas/genética , Transducción de Señal/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética
8.
Cell Physiol Biochem ; 33(3): 758-68, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24662459

RESUMEN

BACKGROUND/AIMS: Demonstrating the molecular mechanisms of human adipose tissue-derived mesenchymal stem cells (hADSCs) differentiation and proliferation could develop hADSCs-based cell therapy. METHODS: The microRNA-137 (miR-137) and cell division control protein 42 homolog (CDC42) levels were regulated by oligonucleotides transfection. The adipogenic differentiation was induced for 10 days in an adipogenic medium and assessed by using an Oil Red O stain. The regulation of miR-137 on CDC42 expression was determined by western blot, real-time PCR and luciferase reporter assay. RESULTS: We confirmed the roles of miR-137 on hADSCs proliferation and adipogenic differentiation. We showed that overexpression of miR-137 inhibited both hADSCs proliferation and adipogenic differentiation. Overexpression of miR-137 also downregulated protein and mRNA levels of CDC42, a predicted target of miR-137. In contrast, inhibition of miR-137 with 2'-O-methyl antisense RNA increased proliferation and adipogenic differentiation in hADSCs. Luciferase reporter activity in the miR-137 target site within the CDC42 3'UTR was lower in miR-137-transfected hADSCs than in control miRNA-transfected hADSCs. RNA interference-mediated downregulation of CDC42 in hADSCs inhibited their proliferation and adipogenic differentiation. CONCLUSION: Our results indicate that miR-137 regulates hADSCs adipogenic differentiation and proliferation by directly targeting CDC42. These findings improve our knowledge of the molecular mechanisms governing hADSCs differentiation and proliferation.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , MicroARNs/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Tejido Adiposo/citología , Humanos , Células del Estroma/citología , Células del Estroma/metabolismo
9.
PLoS Pathog ; 8(5): e1002690, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22589722

RESUMEN

Epstein-Barr virus (EBV) is closely associated with nasopharyngeal carcinoma (NPC), a human malignancy notorious for its highly metastatic nature. Among EBV-encoded genes, latent membrane protein 1 (LMP1) is expressed in most NPC tissues and exerts oncogenicity by engaging multiple signaling pathways in a ligand-independent manner. LMP1 expression also results in actin cytoskeleton reorganization, which modulates cell morphology and cell motility- cellular process regulated by RhoGTPases, such as Cdc42. Despite the prominent association of Cdc42 activation with tumorigenesis, the molecular basis of Cdc42 activation by LMP1 in NPC cells remains to be elucidated. Here using GST-CBD (active Cdc42-binding domain) as bait in GST pull-down assays to precipitate active Cdc42 from cell lysates, we demonstrated that LMP1 acts through its transmembrane domains to preferentially induce Cdc42 activation in various types of epithelial cells, including NPC cells. Using RNA interference combined with re-introduction experiments, we identified FGD4 (FYVE, RhoGEF and PH domain containing 4) as the GEF (guanine nucleotide exchange factor) responsible for the activation of Cdc42 by LMP1. Serial deletion experiments and co-immunoprecipitation assays further revealed that ectopically expressed FGD4 modulated LMP1-mediated Cdc42 activation by interacting with LMP1. Moreover, LMP1, through its transmembrane domains, directly bound FGD4 and enhanced FGD4 activity toward Cdc42, leading to actin cytoskeleton rearrangement and increased motility of NPC cells. Depletion of FGD4 or Cdc42 significantly reduced (∼50%) the LMP1-stimulated cell motility, an effect that was partially reversed by expression of a constitutively active mutant of Cdc42. Finally, quantitative RT-PCR and immunohistochemistry analyses showed that FGD4 and LMP1 were expressed in NPC tissues, supporting the potential physiologically relevance of this mechanism in NPC. Collectively, our results not only uncover a novel mechanism underlying LMP1-mediated Cdc42 activation, namely LMP1 interaction with FGD4, but also functionally link FGD4 to NPC tumorigenesis.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Neoplasias Nasofaríngeas/patología , Proteínas de la Matriz Viral/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Citoesqueleto de Actina/fisiología , Carcinoma , Línea Celular Tumoral , Movimiento Celular , Infecciones por Virus de Epstein-Barr/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Interleucina-1alfa/metabolismo , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Carcinoma Nasofaríngeo , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
10.
J Neurosci ; 31(2): 492-500, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21228159

RESUMEN

Duchenne muscular dystrophy is caused by mutations in the Dystrophin gene and is characterized by muscle degeneration and the occurrence of mental deficits in a significant number of patients. Although Dystrophin and its closely related ortholog Utrophin are present at a variety of synapses, little is known about their roles in the nervous system. Previously, we reported that absence of postsynaptic Dystrophin from the Drosophila neuromuscular junction (NMJ) disrupts synaptic homeostasis, resulting in increased stimulus-evoked neurotransmitter release. Here, we show that RhoGAP crossveinless-c (cv-c), a negative regulator of Rho GTPase signaling pathways, genetically interacts with Dystrophin. Electrophysiological characterization of the cv-c-deficient NMJ and the use of presynaptic- and postsynaptic-specific transgenic rescue versus RNA interference reveal that the absence of postsynaptic cv-c results in elevated evoked neurotransmitter release. The cv-c mutant NMJ exhibits an increased number of presynaptic neurotransmitter release sites and higher probability of vesicle release without apparent changes in postsynaptic glutamate receptor numbers or function. Moreover, we find that decreasing expression of the Rho GTPase Cdc42 suppresses the high neurotransmitter release in the cv-c and Dystrophin mutants, suggesting that Cdc42 is a substrate of Cv-c. These results indicate that Dystrophin and the Rho GTPase signaling pathway likely interact at the postsynaptic side of the NMJ to maintain synaptic homeostasis. The absence of this postsynaptic pathway results in presynaptic structural and functional alterations, suggesting that retrograde signaling mechanisms are affected.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Distrofina/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Unión Neuromuscular/fisiología , Sinapsis/fisiología , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Distrofina/genética , Proteínas Activadoras de GTPasa/genética , Homeostasis , Larva , Potenciales Postsinápticos Miniatura , Mutación , Neurotransmisores/metabolismo , Interferencia de ARN , Receptores de Glutamato/metabolismo , Transducción de Señal , Alas de Animales/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis
11.
Biochem Biophys Res Commun ; 418(1): 93-7, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22244890

RESUMEN

Max binding protein (MNT) is a member of the Myc/Max/Mad network that plays a role in cell proliferation, differentiation and apoptosis. We previously observed that MNT was differentially expressed in hepatocellular carcinoma (HCC) and interacted with Nck1 by 2-DE. Nck family adaptor proteins function to couple tyrosine phosphorylation signals, regulate actin cytoskeletal reorganization and lead to cell motility. In order to investigate the regulatory role of MNT in HCC migration, we used transient transfection with a MNT expressing vector to overexpress MNT protein in SMMC7721 cells, and MNT siRNA to knockdown MNT expression. Rho Family Small GTPase activation assay, Western blots and transwell assay were used to determine the migration potential of cells. We found that knockdown of MNT expression might promote SMMC7721 cell migration, while the overexpressed MNT could significantly inhibit cell migration. It further emphasized the role of MNT in inhibition of cell migration that might be a promising target for HCC chemotherapy.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Carcinoma Hepatocelular/patología , Movimiento Celular , Neoplasias Hepáticas/patología , Proteínas Represoras/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/metabolismo , Proteínas Represoras/genética , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis
12.
Mol Reprod Dev ; 79(5): 356-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22407942

RESUMEN

Mammalian target of rapamycin (mTOR) is central to the control of cell proliferation, growth, and survival in mammalian cells. Prolonged treatment with rapamycin inhibits mTOR complex 2 (mTORC2) activity, and both the mTORC1-mediated S6K1 and 4E-BP1/eIF4E pathways are essential for TORC2-mediated RhoA, Cdc42, and Rac1 expression during cell motility and F-actin reorganization. The functions of mTOR in the mouse oocyte remain unclear, however. The present study shows that rapamycin affects mTOR expression and cytoskeleton reorganization during meiotic maturation of mouse oocytes. mTOR mRNA was expressed in germinal vesicles (GV) until metaphase I (MI), and increased during metaphase II (MII). Immunostaining showed that mTOR localized around the spindle and in the cytoplasm of oocytes. Treatment of oocytes with rapamycin decreased mTOR at the RNA and protein level, and altered asymmetric division. Formation of the actin cap and the cortical granule-free domain were also disrupted after rapamycin treatment, indicating the failure of spindle migration. Injection of an anti-mTOR antibody yielded results consistent with those obtained for rapamycin treatment, further confirming the involvement of mTOR in oocyte polarity. Furthermore, rapamycin treatment reduced the mRNA expression of small GTPases (RhoA, Cdc42, and Rac1), which are crucial regulatory factors for cytoskeleton reorganization. Taken together, these results suggest that rapamycin inhibits spindle migration and asymmetric division during mouse oocyte maturation via mTOR-mediated small GTPase signaling pathways.


Asunto(s)
Citocinesis , Proteínas de Unión al GTP Monoméricas/metabolismo , Oocitos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Capping de la Actina/biosíntesis , Animales , Proliferación Celular , Citocinesis/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Metafase/fisiología , Ratones , ARN Mensajero/biosíntesis , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis , Proteína de Unión al GTP rhoA/biosíntesis
13.
Nat Cell Biol ; 2(9): 637-44, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10980705

RESUMEN

RhoA organizes actin stress fibres and is necessary for cell transformation by oncogenes such as src and ras. Moreover, RhoA is implicated in cadherin clustering during the formation of adherens junctions. The catenin p120 has also been implicated in cadherin clustering through an unknown mechanism. Here we show that p120 selectively inhibits RhoA activity in vitro and in vivo. RhoA inhibition and the interaction of p120 with cadherins are mutually exclusive, suggesting a mechanism for regulating the recruitment and exchange of RhoA at nascent cell-cell contacts. By affecting RhoA activation, p120 could modulate cadherin functions, including suppression of invasion, neurite extension and junction formation.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fosfoproteínas/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Células 3T3 , Animales , Cadherinas/metabolismo , Cateninas , Moléculas de Adhesión Celular/genética , Guanosina Difosfato/metabolismo , Humanos , Lisofosfolípidos/farmacología , Ratones , Fenotipo , Fosfoproteínas/genética , Células Tumorales Cultivadas , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/biosíntesis , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Catenina delta
14.
Dig Dis Sci ; 56(7): 2009-16, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21221794

RESUMEN

INTRODUCTION: MicroRNAs (miRNAs) are a class of small (19-25 nucleotides) noncoding RNAs that regulate the expressions of a wide variety of genes, including some involved in cancer development. Some recent studies show that DNA methylation contributes to down-regulation of microRNA-137 (miR-137) during tumorigenesis. Whether down-regulation of miR-137 also exists in gastric cancer is unknown. AIM: Our aim was to test the hypothesis that down-regulation of miR-137 also exists in gastric cancer. METHODS: Expression of levels of miR-137 were examined using real-time PCR on paired gastric cancer and adjacent non-cancerous tissues. The methylation status is detected by MSP. RESULTS: Results show that miR-137 is downregulated by hypermethylation of the promoter in gastric cancer tissues. Epigenetic silencing of miR-137 induced an up-regulation of its targets, Cdc42. Restoration of the miR-137 expression in gastric cancer cell lines downregulated the Cdc42 expression. Restoration of the miR-137 and inactivation of Cdc42 induce apoptosis and cell cycle G1 arrest in gastric cancer cells. Furthermore, the miR-137 expression was found to be inversely correlated with CDC42 expression in gastric caner. CONCLUSIONS: miR-137 is frequently down-regulated in gastric cancer and is a negative regulator of Cdc42.


Asunto(s)
Carcinoma/metabolismo , MicroARNs/biosíntesis , Neoplasias Gástricas/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Metilación de ADN , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Datos de Secuencia Molecular
15.
Mycopathologia ; 172(5): 347-55, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21796487

RESUMEN

Aspergillus fumigatus is an opportunistic pathogen that may cause severe invasive disease in immunocompromised patients. The filamentous fungi undergo polarized growth, searching for nutrients in the environment and causing invasive growth in tissue. Sho1 is a sensor of the high osmolarity glycerol pathway, and the sho1 mutant showed a decrease in growth rate. We found that sho1 is involved in the polarized growth of A. fumigatus. The sho1 mutation resulted in extended isotropic growth of germinating conidia followed by multiple germ tubes and wide hyphae with short intercalary cells by calcofluor white staining. The mechanism by which sho1 gene affected polarized growth is investigated. A reduced number of apical vesicles with greater dispersion were observed by transmission electron microscopy in the Spitzenkörper body of the sho1 mutant. Actin patches were distributed randomly at low density at early stages of mutant strain fungal development and reaggregated to the hyphal tip of later stages when long filamentous fungi formed. Actin patches located at the tip of polarized wild-type cells. RNA levels of polarized growth-related genes Rho GTPases were detected by real-time PCR. The sho1 gene did not affect the RNA expression when strains were cultured at 37°C for 6 h. At 17 h, the RNA expression of rho1, rho3 and CDC42 in the sho1 mutant were 0.18-, 0.18- and 0.33-fold of that in the wild type. The sho1 gene affected the polarized growth through affecting the expression of Rho GTPases, the distribution of actin cytoskeleton, vesicle quantity and distribution.


Asunto(s)
Aspergillus fumigatus/crecimiento & desarrollo , Proteínas Fúngicas/genética , Hifa/crecimiento & desarrollo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Esporas Fúngicas/crecimiento & desarrollo , Citoesqueleto de Actina/metabolismo , Aspergillus fumigatus/citología , Aspergillus fumigatus/genética , Aspergillus fumigatus/metabolismo , Polaridad Celular/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Hifa/genética , Hifa/metabolismo , Mutación , ARN Mensajero/biosíntesis , Esporas Fúngicas/genética , Esporas Fúngicas/metabolismo , Proteína de Unión al GTP cdc42/biosíntesis , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
16.
Breast Cancer Res ; 12(5): R73, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20860838

RESUMEN

INTRODUCTION: Rho GTPases are overexpressed and hyperactivated in human breast cancers. Deficiency of p190B RhoGAP, a major inhibitor of the Rho GTPases, inhibits mouse mammary tumor virus long terminal repeat (MMTV)-Neu/ErbB2 mammary tumor formation and progression in part through effects within the stromal environment, suggesting that p190B function is pro-tumorigenic. To further investigate the potential pro-tumorigenic actions of p190B, we examined the effects of exogenous p190B expression within the mammary epithelium on MMTV-Neu tumor formation and progression. METHODS: Tetracycline-regulatable p190B transgenic mice were bred to MMTV-Neu mice, and the effects of exogenous p190B expression on tumor latency, multiplicity, growth rates, angiogenesis, and metastasis were examined. The effects of exogenous p190B expression on cell-matrix adhesion and invasion were tested using non-transformed primary mammary epithelial cells (MECs). Rho GTPase activity, oxidative stress as an indicator of reactive oxygen species (ROS) production, and downstream signaling pathways were analyzed. RESULTS: Altered p190B expression resulted in a 2-fold increase in tumor multiplicity and a 3-fold increase in metastases compared to control mice indicating that exogenous p190B expression in the mammary epithelium promotes MMTV-Neu mammary tumor formation and progression. Interestingly, non-transformed primary MECs expressing exogenous p190B displayed increased adhesion to laminin and type IV collagen and formed invasive structures in a three-dimensional culture assay. Ras related C3 botulinum toxin 1 (Rac1)-GTP levels were elevated in p190B transgenic tumors whereas Ras homologous A (RhoA) and cell division cycle 42 (Cdc42)-GTP levels were not significantly altered. Rac1 activity affects production of ROS, which regulate transformation, metastasis, and oxidative stress. Protein carbonylation, which is indicative of oxidative stress, was elevated 1.75-fold in p190B transgenic tumors as compared to control tumors suggesting that exogenous p190B expression may affect Rac1-dependent ROS production. CONCLUSIONS: These studies indicate that paradoxically, p190B RhoGAP, a major inhibitor of the Rho GTPases in vitro, has pro-tumorigenic functions that enhance MMTV-Neu induced mammary tumor formation and metastasis. Furthermore, exogenous p190B expression enhances cell adhesion and invasion, which may facilitate metastasis. Rac1 activity and oxidative stress are elevated in tumors expressing exogenous p190B suggesting that p190B may promote tumorigenesis through a Rac1/ROS dependent mechanism.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Neoplasias Mamarias Animales/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Animales , Adhesión Celular , Transformación Celular Neoplásica/genética , Uniones Célula-Matriz , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Proteínas Activadoras de GTPasa/genética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Virus del Tumor Mamario del Ratón/genética , Ratones , Metástasis de la Neoplasia , Neovascularización Patológica , Estrés Oxidativo , Especies Reactivas de Oxígeno , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis , Proteína de Unión al GTP rhoA/biosíntesis , Proteína de Unión al GTP rhoA/metabolismo
17.
J Virol ; 83(19): 9759-72, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19640983

RESUMEN

Keratinocytes of the skin or mucosa are the primary entry portals for herpes simplex virus type 1 (HSV-1) in vivo. We hypothesized that dynamics of cell motility and adhesion contribute to the initial steps of HSV-1 infection of epithelial cells, and thus, we investigated the impact of Rac1 and Cdc42, which serve as key regulators of actin dynamics. Measurement of endogenous Rac1 and Cdc42 in the human keratinocyte cell line HaCaT indicated temporary changes in activity levels of Rac1/Cdc42 upon HSV-1 infection. Overexpression of Rac1/Cdc42 mutants in HaCaT cells demonstrated a decrease of infection efficiency with constitutively active Rac1 or Cdc42, while dominant-negative Rac1 had no effect. Accordingly, we addressed whether the absence of Rac1 and/or Cdc42 influenced infection, and we performed RNA interference studies. Both in HaCaT cells and in primary human keratinocytes, reduction of Rac1 and/or Cdc42 did not suppress infection. When mouse epidermis was infected ex vivo, we observed early HSV-1 infection in basal keratinocytes. Similar results were obtained upon infection of mouse epidermis with a keratinocyte-restricted deletion of the rac1 gene, indicating no inhibitory effect on HSV-1 infection in the absence of Rac1. Our results suggest that HSV-1 infection of keratinocytes does not depend on pathways involving Rac1 and Cdc42 and that constitutively active Rac1 and Cdc42 have the potential to interfere with HSV-1 infectivity.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Queratinocitos/virología , Transducción de Señal , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP rac1/biosíntesis , Animales , Epidermis/virología , Genes Dominantes , Humanos , Ratones , Ratones Noqueados , Proteínas de Unión al GTP rho/biosíntesis , Proteína de Unión al GTP rhoA/biosíntesis
18.
Am J Pathol ; 175(2): 903-15, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19574426

RESUMEN

Platelet endothelial cell adhesion molecule (PECAM)-1 has been previously implicated in endothelial cell migration; additionally, anti-PECAM-1 antibodies have been shown to inhibit in vivo angiogenesis. Studies were therefore performed with PECAM-1-null mice to further define the involvement of PECAM-1 in blood vessel formation. Vascularization of subcutaneous Matrigel implants as well as tumor angiogenesis were both inhibited in PECAM-1-null mice. Reciprocal bone marrow transplants that involved both wild-type and PECAM-1-deficient mice revealed that the impaired angiogenic response resulted from a loss of endothelial, but not leukocyte, PECAM-1. In vitro wound migration and single-cell motility by PECAM-1-null endothelial cells were also compromised. In addition, filopodia formation, a feature of motile cells, was inhibited in PECAM-1-null endothelial cells as well as in human endothelial cells treated with either anti-PECAM-1 antibody or PECAM-1 siRNA. Furthermore, the expression of PECAM-1 promoted filopodia formation and increased the protein expression levels of Cdc42, a Rho GTPase that is known to promote the formation of filopodia. In the developing retinal vasculature, numerous, long filamentous filopodia, emanating from endothelial cells at the tips of angiogenic sprouts, were observed in wild-type animals, but to a lesser extent in the PECAM-1-null mice. Together, these data further establish the involvement of endothelial PECAM-1 in angiogenesis and suggest that, in vivo, PECAM-1 may stimulate endothelial cell motility by promoting the formation of filopodia.


Asunto(s)
Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Animales , Movimiento Celular/genética , Células Cultivadas , Colágeno , Combinación de Medicamentos , Humanos , Laminina , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Fisiológica/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proteoglicanos , Seudópodos/fisiología , Retina/crecimiento & desarrollo , Vasos Retinianos , Proteína de Unión al GTP cdc42/biosíntesis
19.
Parasitol Res ; 107(1): 153-62, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20369253

RESUMEN

A full-length cDNA encoding a Rho-family small GTPase gene cdc42 of Trichinella spiralis was expressed in E. coli. The recombinant protein of TsCDC42 was purified and used to raise the polyclonal antibodies. The expression of TsCDC42 in different stages of parasite was investigated. The western blot showed that TsCDC42 was expressed in all stages of T. spiralis, including newborn larvae, muscle larvae and adult worms. The immuno-localization revealed that TsCDC42 was ubiquitously distributed in the newborn larvae, muscle larvae and adult worm. Cross-species RNAi was done by knockdown Tscdc42 RNAi in C. elegans. The results revealed that endogenous expression level of CDC42 was decreased by knockdown Tscdc42 RNAi in C. elegans, and this knockdown reduced the progeny of C. elegans. It suggested that Tscdc42 might play the same roles in the early development of T. spiralis.


Asunto(s)
Caenorhabditis elegans/crecimiento & desarrollo , Regulación de la Expresión Génica , Proteínas del Helminto/biosíntesis , Trichinella spiralis/enzimología , Proteína de Unión al GTP cdc42/biosíntesis , Animales , Anticuerpos Antihelmínticos/inmunología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Escherichia coli/genética , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas del Helminto/genética , Proteínas del Helminto/aislamiento & purificación , Humanos , Larva/química , Larva/enzimología , Ratones , Microscopía Fluorescente , Interferencia de ARN , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Trichinella spiralis/química , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/aislamiento & purificación
20.
Cell Death Dis ; 11(10): 907, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33097698

RESUMEN

The eukaryotic cell cycle involves a highly orchestrated series of events in which the cellular genome is replicated during a synthesis (S) phase and each of the two resulting copies are segregated properly during mitosis (M). Host cell factor-1 (HCF-1) is a transcriptional co-regulator that is essential for and has been implicated in basic cellular processes, such as transcriptional regulation and cell cycle progression. Although a series of HCF-1 transcriptional targets have been identified, few functional clues have been provided, especially for chromosome segregation. Our results showed that HCF-1 activated CDC42 expression by binding to the -881 to -575 region upstream of the CDC42 transcription start site, and the regulation of CDC42 expression by HCF-1 was correlated with cell cycle progression. The overexpression of a spontaneously cycling and constitutively active CDC42 mutant (CDC42F28L) rescued G1 phase delay and multinucleate defects in mitosis upon the loss of HCF-1. Therefore, these results establish that HCF-1 ensures proper cell cycle progression by regulating the expression of CDC42, which indicates a possible mechanism of cell cycle coordination and the regulation mode of typical Rho GTPases.


Asunto(s)
Factor C1 de la Célula Huésped/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Ciclo Celular/fisiología , Segregación Cromosómica , Ciclina A/biosíntesis , Ciclina A/genética , Progresión de la Enfermedad , Puntos de Control de la Fase G1 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Factor C1 de la Célula Huésped/genética , Humanos , Mitosis , Regiones Promotoras Genéticas , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA