Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(22)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34830244

RESUMEN

p130 Crk-associated substrate (p130Cas) is associated with poor prognosis and treatment resistance in breast and lung cancers. To elucidate p130Cas functional and clinical role in colorectal cancer (CRC) progression/therapy resistance, we performed cell culture experiments and bioinformatic/statistical analyses of clinical data sets. p130Cas expression was associated with poor survival in the cancer genome atlas (TCGA) data set. Knockdown/reconstitution experiments showed that p130Cas drives migration but, unexpectedly, inhibits proliferation in CRC cells. TCGA data analyses identified the growth factor epiregulin (EREG) as inversely correlated with p130Cas. p130Cas knockdown and simultaneous EREG treatment further enhanced proliferation. RNA interference and EREG treatment experiments suggested that p130Cas/EREG limit each other's expression/activity. Inverse p130Cas/EREG Spearman correlations were prominent in right-sided and earlier stage CRC. p130Cas was inducible by 5-fluorouracil (5-FU) and FOLFIRI (folinic acid, 5-FU, irinotecan), and p130Cas and EREG were upregulated in distant metastases (GSE121418). Positive p130Cas/EREG correlations were observed in metastases, preferentially in post-treatment samples (especially pulmonary metastases). p130Cas knockdown sensitized CRC cells to FOLFIRI independent of EREG treatment. RNA sequencing and gene ontology analyses revealed that p130Cas is involved in cytochrome P450 drug metabolism and epithelial-mesenchymal transition. p130Cas expression was associated with poor survival in right-sided, stage I/II, MSS (microsatellite stable), or BRAF-mutated CRC. In summary, p130Cas represents a prognostic factor and potential therapeutic target in CRC.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias Colorrectales/diagnóstico , Proteína Sustrato Asociada a CrK/genética , Epirregulina/genética , Transición Epitelial-Mesenquimal/genética , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Atlas como Asunto , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/metabolismo , Epirregulina/metabolismo , Femenino , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Leucovorina/uso terapéutico , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Mutación , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia
2.
PLoS One ; 11(9): e0162502, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27622508

RESUMEN

OBJECTIVE: Ezrin and p130Cas are structural proteins with an important role in signaling pathways and have been shown to promote cancer dissemination. We previously reported on overexpression of both ezrin and p130Cas in breast carcinoma effusions compared to primary carcinomas. Since ovarian and breast carcinomas share the ability to disseminate by forming malignant effusions, we sought to study the role of these molecules in ovarian carcinoma (OC). METHODS: OC cell lines were cultured in two different 3-dimensional conditions, on alginate scaffolds and as spheroids, which served as models for solid tumor and malignant effusions, respectively. shRNA was used to reduce protein expression in the cells. The malignant potential was evaluated by chemo-invasion assay, branching capacity on Matrigel and rate of proliferation. Subsequently, clinical specimens of high-grade serous carcinoma effusions, ovarian tumors and solid metastases were analyzed for ezrin and p130Cas expression. RESULTS: Higher ezrin expression was found in cells composing the spheroids compared to their counterparts cultured on alginate scaffold and in clinical samples of malignant effusions compared to solid tumors. In addition, reduced Ezrin expression impaired the invasion ability and the branching capacity of OC cells to a greater extent than reduced p130Cas expression. However, ezrin and p130Cas expression in effusions was unrelated to clinical outcome. CONCLUSIONS: The 3-dimensional cell cultures were found to mimic the different tumor sites and be applicable as a model. The in vitro results concur with the clinical specimen analysis, suggesting that in OC, the role of ezrin in disease progression is more pronounced than that of p130Cas.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neoplasias Ováricas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Líquido Ascítico/metabolismo , Líquido Ascítico/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/metabolismo , Derrame Pleural Maligno/patología , ARN Interferente Pequeño/genética , Esferoides Celulares
3.
Biochim Biophys Acta ; 1860(8): 1753-63, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27068854

RESUMEN

We previously demonstrated that focal adhesion kinase (FAK), p130Cas and paxillin are crucially involved in the enhanced malignant properties under expression of ganglioside GD3 in melanoma cells. Therefore, molecules existing in the GD3-mediated signaling pathway could be considered as suitable targets for therapeutic intervention in malignant melanoma. The aim of this study was to determine whether blockade of p130Cas and/or paxillin by RNAi suppresses melanoma growth. We found a suitable dose (40 µM siRNA, 25 µl/tumor) of the siRNA to suppress p130Cas in the xenografts generated in nu/nu mice. Based on these results, we performed intratumoral (i.t.) treatment with anti-p130Cas and/or anti-paxillin siRNAs mixed with atelocollagen as a drug delivery system in a xenograft tumor of a human melanoma cell line, SK-MEL-28. Mixture of atelocollagen (1.75%) and an siRNA (500 or 1000 pmol/tumor) was injected into the tumors every 3 days after the first injection. An siRNA against human p130Cas markedly suppressed tumor growth of the xenograft in a dose-dependent manner, whereas siRNA against human paxillin slightly inhibited the tumor growth. A control siRNA against firefly luciferase showed no effect. To our surprise, siRNA against human p130Cas (500 or 1000 pmol/tumor) combined with siRNA against human paxillin dramatically suppressed tumor growth. In agreement with the tumor suppression effects of the anti-p130Cas siRNA, reduction in Ki-67 positive cell number as well as in p130Cas expression was demonstrated by immunohistostaining. These results suggested that blockade of GD3-mediated growth signaling pathways by siRNAs might be a novel and promising therapeutic strategy against malignant melanomas, provided signaling molecules such as p130Cas and paxillin are significantly expressed in individual cases. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.


Asunto(s)
Proteína Sustrato Asociada a CrK , Gangliósidos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma , Proteínas de Neoplasias , Paxillin , ARN Interferente Pequeño , Animales , Línea Celular Tumoral , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/biosíntesis , Proteína Sustrato Asociada a CrK/genética , Gangliósidos/biosíntesis , Gangliósidos/genética , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Paxillin/antagonistas & inhibidores , Paxillin/biosíntesis , Paxillin/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Cell Mol Med ; 20(5): 962-72, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26945908

RESUMEN

Epidemiologic data show the incidence of gastric cancer in men is twofold higher than in women worldwide. Oestrogen is reported to have the capacity against gastric cancer development. Endogenous oestrogen reduces gastric cancer incidence in women. Cancer patients treated with oestrogens have a lower subsequent risk of gastric cancer. Accumulating studies report that bone marrow mesenchymal stem cells (BMMSCs) might contribute to the progression of gastric cancer through paracrine effect of soluble factors. Here, we further explore the effect of oestrogen on BMMSCs-mediated human gastric cancer invasive motility. We founded that HBMMSCs notably secrete interleukin-8 (IL-8) protein. Administration of IL-8 specific neutralizing antibody significantly inhibits HBMMSCs-mediated gastric cancer motility. Treatment of recombinant IL-8 soluble protein confirmed the role of IL-8 in mediating HBMMSCs-up-regulated cell motility. IL-8 up-regulates motility activity through Src signalling pathway in human gastric cancer. We further observed that 17ß -estradiol inhibit HBMMSCS-induced cell motility via suppressing activation of IL8-Src signalling in human gastric cancer cells. 17ß-estradiol inhibits IL8-up-regulated Src downstream target proteins including p-Cas, p-paxillin, p-ERK1/2, p-JNK1/2, MMP9, tPA and uPA. These results suggest that 17ß-estradiol significantly inhibits HBMMSCS-induced invasive motility through suppressing IL8-Src signalling axis in human gastric cancer cells.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica , Interleucina-8/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Mucosa Gástrica/metabolismo , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Paxillin/antagonistas & inhibidores , Paxillin/genética , Paxillin/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Estómago/patología
5.
Cancer Biol Ther ; 15(11): 1571-82, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25482942

RESUMEN

Blocking the migration of metastatic cancer cells is a major goal in the therapy of cancer. The receptor tyrosine kinase AXL is one of the main triggers for cancer cell migration in neoplasia of breast, colon, skin, thyroid and prostate. In our study we analyzed the effect of AXL inhibition on cell motility and viability in triple negative breast cancer cell lines overexpressing AXL. Thereby we reveal that the compound BMS777607, exhibiting the lowest IC50 values for inhibition of AXL kinase activity in the studied cell lines, attenuates cell motility to a lower extent than the kinase inhibitors MPCD84111 and SKI606. By analyzing the target kinases of MPCD84111 and SKI606 with kinase profiling assays we identified Lyn, a Src family kinase, as a target of both compounds. Knockdown of Lyn and the migration-related CRK-associated substrate (p130Cas), had a significant inhibitory effect on cell migration. Taken together, our findings highlight the importance of combinatorial or multikinase inhibition of non-receptor tyrosine kinases and AXL receptor tyrosine kinase in the therapy of triple negative breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Análisis por Conglomerados , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Tirosina Quinasa del Receptor Axl
6.
Nat Commun ; 3: 1291, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23250421

RESUMEN

MicroRNAs (miRNAs) are essential for regulating normal embryonic development and carcinogenesis. Here we report that miR-17-5p, an oncofoetal miRNA, is a key regulator of colorectal cancer progression. We show that miR-17-5p is an oncogenic miRNA that regulates tumorigenesis and progression by targeting the gene encoding P130 and subsequently activating the Wnt/ß-catenin pathway. Using specimens from two large cohorts of colorectal cancer patients, we found that patients whose tumours had high miR-17-5p expression had shorter overall survival rates but showed a better response to adjuvant chemotherapy than did patients whose tumours had low miRNA expression. We also observed a strong inverse correlation between miR-17-5p and P130 expression. The current findings suggest that miR-17-5p is a crucial determinant of colorectal cancer progression.


Asunto(s)
Neoplasias Colorrectales/genética , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , MicroARNs/fisiología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/fisiopatología , Proteína Sustrato Asociada a CrK/biosíntesis , Regulación hacia Abajo/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/biosíntesis , Neoplasias Experimentales/genética , Neoplasias Experimentales/fisiopatología , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Supervivencia , Vía de Señalización Wnt/fisiología
7.
Biochem J ; 445(3): 323-32, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22587391

RESUMEN

Actin dynamics are implicated in various cellular processes, not only through the regulation of cytoskeletal organization, but also via the control of gene expression. In the present study we show that the Src family kinase substrate p130Cas (Cas is Crk-associated substrate) influences actin remodelling and concomitant muscle-specific gene expression, thereby regulating myogenic differentiation. In C2C12 myoblasts, silencing of p130Cas expression by RNA interference impaired F-actin (filamentous actin) formation and nuclear localization of the SRF (serum-response factor) co-activator MAL (megakaryocytic acute leukaemia) following the induction of myogenic differentiation. Consequently, formation of multinucleated myotubes was abolished. Re-introduction of wild-type p130Cas, but not its phosphorylation-defective mutant, into p130Cas-knockdown myoblasts restored F-actin assembly, MAL nuclear localization and myotube formation. Depletion of the adhesion molecule integrin ß3, a key regulator of myogenic differentiation as well as actin cytoskeletal organization, attenuated p130Cas phosphorylation and MAL nuclear localization during C2C12 differentiation. Moreover, knockdown of p130Cas led to the activation of the F-actin-severing protein cofilin. The introduction of a dominant-negative mutant of cofilin into p130Cas-knockdown myoblasts restored muscle-specific gene expression and myotube formation. The results of the present study suggest that p130Cas phosphorylation, mediated by integrin ß3, facilitates cofilin inactivation and promotes myogenic differentiation through modulating actin cytoskeleton remodelling.


Asunto(s)
Actinas/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Desarrollo de Músculos/fisiología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Núcleo Celular/metabolismo , Cofilina 2/antagonistas & inhibidores , Cofilina 2/genética , Cofilina 2/metabolismo , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/genética , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Modelos Biológicos , Desarrollo de Músculos/genética , Mutagénesis , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN
8.
J Gen Virol ; 92(Pt 11): 2620-2627, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21813705

RESUMEN

Human papillomaviruses (HPVs) with tropism for mucosal epithelia are the major aetiological factors in cervical cancer. Most cancers are associated with so-called high-risk HPV types, in particular HPV16, and constitutive expression of the HPV16 E6 and E7 oncoproteins is critical for malignant transformation in infected keratinocytes. E6 and E7 bind to and inactivate the cellular tumour suppressors p53 and Rb, respectively, thus delaying differentiation and inducing proliferation in suprabasal keratinocytes to enable HPV replication. One member of the Rb family, p130, appears to be a particularly important target for E7 in promoting S-phase entry. Recent evidence indicates that p130 regulates cell-cycle progression as part of a large protein complex termed DREAM. The composition of DREAM is cell cycle-regulated, associating with E2F4 and p130 in G0/G1 and with the B-myb transcription factor in S/G2. In this study, we addressed whether p130-DREAM is disrupted in HPV16-transformed cervical cancer cells and whether this is a critical function for E6/E7. We found that p130-DREAM was greatly diminished in HPV16-transformed cervical carcinoma cells (CaSki and SiHa) compared with control cell lines; however, when E6/E7 expression was targeted by specific small hairpin RNAs, p130-DREAM was reformed and the cell cycle was arrested. We further demonstrated that the profound G1 arrest in E7-depleted CaSki cells was dependent on p130-DREAM reformation by also targeting the expression of the DREAM component Lin-54 and p130. The results show that continued HPV16 E6/E7 expression is necessary in cervical cancer cells to prevent cell-cycle arrest by a repressive p130-DREAM complex.


Asunto(s)
Ciclo Celular , Proteína Sustrato Asociada a CrK/metabolismo , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/patogenicidad , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Células Epiteliales/fisiología , Células Epiteliales/virología , Humanos , Proteínas de Interacción con los Canales Kv/antagonistas & inhibidores , Multimerización de Proteína , Proteínas Represoras/antagonistas & inhibidores
9.
Proc Natl Acad Sci U S A ; 107(5): 2201-6, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20133863

RESUMEN

Inhibitors of poly(ADP-ribose) polymerase (PARP) are in clinical trials for cancer therapy, on the basis of the role of PARP in recruitment of base excision repair (BER) factors to sites of DNA damage. Here we show that PARP inhibition to block BER is toxic to hypoxic cancer cells, in which homology-dependent repair (HDR) is known to be down-regulated. However, we also report the unexpected finding that disruption of PARP, itself, either via chemical PARP inhibitors or siRNAs targeted to PARP-1, can inhibit HDR by suppressing expression of BRCA1 and RAD51, key factors in HDR of DNA breaks. Mechanistically, PARP inhibition was found to cause increased occupancy of the BRCA1 and RAD51 promoters by repressive E2F4/p130 complexes, a pathway prevented by expression of HPV E7, which disrupts p130 activity, or by siRNAs to knock down p130 expression. Functionally, disruption of p130 by E7 expression or by siRNA knockdown also reverses the cytotoxicity and radiosensitivity associated with PARP inhibition, suggesting that the down-regulation of BRCA1 and RAD51 is central to these effects. Direct measurement of HDR using a GFP-based assay demonstrates reduced HDR in cells treated with PARP inhibitors. This work identifies a mechanism by which PARP regulates DNA repair and suggests new strategies for combination cancer therapies.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Factor de Transcripción E2F4/metabolismo , Genes BRCA1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Recombinasa Rad51/genética , Línea Celular Tumoral , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/fisiología , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes BRCA1/efectos de los fármacos , Humanos , Fenantrenos/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Fármacos Sensibilizantes a Radiaciones/farmacología
10.
Virology ; 397(1): 139-44, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19945133

RESUMEN

HPV type 58 (HPV-58) is the third most common HPV type in cervical cancer from Eastern Asia, yet little is known about how it promotes carcinogenesis. In this study, we demonstrate that HPV-58 E7 significantly promoted the proliferation and extended the lifespan of primary human keratinocytes (PHKs). HPV-58 E7 abrogated the G1 and the postmitotic checkpoints, although less efficiently than HPV-16 E7. Consistent with these observations, HPV-58 E7 down-regulated the cellular tumor suppressor pRb to a lesser extent than HPV-16 E7. Similar to HPV-16 E7 expressing PHKs, Cdk2 remained active in HPV-58 E7 expressing PHKs despite the presence of elevated levels of p53 and p21. Interestingly, HPV-58 E7 down-regulated p130 more efficiently than HPV-16 E7. Our study demonstrates a correlation between the ability of down-regulating pRb/p130 and abrogating cell cycle checkpoints by HPV-58 E7, which also correlates with the biological risks of cervical cancer progression associated with HPV-58 infection.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Papillomaviridae/patogenicidad , Proteínas E7 de Papillomavirus/metabolismo , Proliferación Celular , Células Cultivadas , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Humanos , Queratinocitos/virología , Proteínas Supresoras de Tumor/antagonistas & inhibidores
11.
Mol Cancer Ther ; 7(5): 1185-94, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18483306

RESUMEN

Src family kinase activity is elevated in many human tumors, including breast cancer, and is often associated with aggressive disease. We examined the effects of SKI-606 (bosutinib), a selective Src family kinase inhibitor, on human cancer cells derived from breast cancer patients to assess its potential for breast cancer treatment. Our results show that SKI-606 caused a decrease in cell motility and invasion of breast cancer cell lines with an IC50 of approximately 250 nmol/L, which was also the IC50 for inhibition of cellular Src kinase activity in intact tumor cells. These changes were accompanied by an increase in cell-to-cell adhesion and membrane localization of beta-catenin. By contrast, cell proliferation and survival were unaffected by SKI-606 at concentrations sufficient to block cell migration and invasion. Analysis of downstream effectors of Src revealed that SKI-606 inhibits the phosphorylation of focal adhesion kinase (FAK), proline-rich tyrosine kinase 2 (Pyk2), and Crk-associated substrate (p130Cas), with an IC50 similar to inhibition of cellular Src kinase. Our findings indicate that SKI-606 inhibits signaling pathways involved in controlling tumor cell motility and invasion, suggesting that SKI-606 is a promising therapeutic for breast cancer.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Movimiento Celular/efectos de los fármacos , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/metabolismo , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Modelos Biológicos , Fosforilación , Transducción de Señal , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo
12.
Cell Mol Life Sci ; 65(9): 1446-57, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18392556

RESUMEN

Physical forces can activate colon cancer cell adhesion, critical for metastasis. Paxillin is phosphorylated by FAK and required for pressure-stimulated adhesion. However, whether paxillin acts as an inert scaffolding protein or whether paxillin phosphorylation is required is unknown. Transfection with paxillin point-phosphorylation mutants demonstrated that phosphorylation at tyrosines 31 and 118 together is necessary for pressure-stimulated adhesion. We further evaluated potential paxillin partners. Reducing the adaptor protein Crk or the focal adhesion protein p130Cas blocked pressure-stimulated adhesion. Furthermore, Crk and p130Cas both displayed increased co-immunoprecipitation with paxillin in response to increased pressure, except in cells transfected with a Y31Y118 paxillin mutant. Inhibiting the small GTPase Rac1 also abolished pressure-stimulated adhesion, and reducing paxillin by siRNA blocked Rac1 phosphorylation by pressure. Thus, paxillin phosphorylation at tyrosines 31 and 118 together is necessary for pressure-induced adhesion. Paxillin, Crk and Cas form a trimeric complex that activates Rac1 and mediates this effect.


Asunto(s)
Neoplasias del Colon/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Paxillin/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Actinina/antagonistas & inhibidores , Adhesión Celular , Línea Celular Tumoral , Neoplasias del Colon/patología , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Humanos , Metástasis de la Neoplasia , Paxillin/antagonistas & inhibidores , Paxillin/química , Fosforilación , Presión , Proteínas Proto-Oncogénicas c-crk/antagonistas & inhibidores , Interferencia de ARN , Células Tumorales Cultivadas , Tirosina/metabolismo
13.
J Biol Chem ; 282(37): 27392-27401, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17623670

RESUMEN

The pattern recognition receptor CD36 initiates a signaling cascade that promotes microglial activation and recruitment to beta-amyloid deposits in the brain. In the present study we identify the focal adhesion-associated proteins p130Cas, Pyk2, and paxillin as novel members of the tyrosine kinase signaling pathway downstream of CD36 and show that assembly of this complex is essential for microglial migration. In primary microglia and macrophages exposed to beta-amyloid, the scaffolding protein p130Cas is rapidly tyrosine-phosphorylated and co-localizes with CD36 to membrane ruffles contemporaneous with F-actin polymerization. These beta-amyloid-stimulated events are not detected in CD36 null cells and are dependent on CD36 activation of Src family tyrosine kinases. Fyn, a Src kinase known to interact with CD36, co-precipitates with p130Cas and is an essential upstream intermediate in the signaling pathways leading to phosphorylation of the p130Cas substrate domain. Furthermore, the p130Cas-interacting kinase Pyk2 and the cytoskeletal adapter protein paxillin also demonstrate CD36-dependent phosphorylation, identifying these focal adhesion molecules as additional members of this beta-amyloid signaling cascade. Disruption of this p130Cas complex by small interfering RNA silencing inhibits p44/42 mitogen-activated protein kinase phosphorylation and microglial migration, illustrating the importance of this pathway in microglial activation and recruitment. Together, these data are the first to identify the signaling cascade that directly links CD36 to the actin cytoskeleton and, thus, implicates it in diverse processes such as cellular migration, adhesion, and phagocytosis.


Asunto(s)
Actinas/metabolismo , Antígenos CD36/fisiología , Proteína Sustrato Asociada a CrK/fisiología , Citoesqueleto/metabolismo , Microglía/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Movimiento Celular , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/genética , Quinasa 2 de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Paxillin/metabolismo , Fosforilación , Transducción de Señal , Receptores Toll-Like/fisiología , Tirosina/metabolismo
14.
Cancer Res ; 65(20): 9185-9, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16230377

RESUMEN

Src family kinases (SFK) are currently being investigated as targets for treatment strategies in various cancers. The novel SFK/Abl inhibitor, dasatinib (BMS-354825), is a promising therapeutic agent with oral bioavailability. Dasatinib has been shown to inhibit growth of Bcr-Abl-dependent chronic myeloid leukemia xenografts in nude mice. Dasatinib also has been shown to have activity against cultured human prostate and breast cancer cells. However, the molecular mechanism by which dasatinib acts on epithelial tumor cells remains unknown. In this study, we show that dasatinib blocks the kinase activities of the SFKs, Lyn, and Src, in human prostate cancer cells at low nanomolar concentrations. Moreover, focal adhesion kinase and Crk-associated substrate (p130(CAS)) signaling downstream of SFKs are also inhibited at similar concentrations of dasatinib. Consistent with inhibition of these signaling pathways, dasatinib suppresses cell adhesion, migration, and invasion of prostate cancer cells at low nanomolar concentrations. Therefore, dasatinib has potential as a therapeutic agent for metastatic prostate cancers harboring activated SFK and focal adhesion kinase signaling.


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/metabolismo , Dasatinib , Relación Dosis-Respuesta a Droga , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA