Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Biochem J ; 479(3): 401-424, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35147166

RESUMEN

The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade promotes cardiomyocyte hypertrophy and is cardioprotective, with the three RAF kinases forming a node for signal integration. Our aims were to determine if BRAF is relevant for human heart failure, whether BRAF promotes cardiomyocyte hypertrophy, and if Type 1 RAF inhibitors developed for cancer (that paradoxically activate ERK1/2 at low concentrations: the 'RAF paradox') may have the same effect. BRAF was up-regulated in heart samples from patients with heart failure compared with normal controls. We assessed the effects of activated BRAF in the heart using mice with tamoxifen-activated Cre for cardiomyocyte-specific knock-in of the activating V600E mutation into the endogenous gene. We used echocardiography to measure cardiac dimensions/function. Cardiomyocyte BRAFV600E induced cardiac hypertrophy within 10 d, resulting in increased ejection fraction and fractional shortening over 6 weeks. This was associated with increased cardiomyocyte size without significant fibrosis, consistent with compensated hypertrophy. The experimental Type 1 RAF inhibitor, SB590885, and/or encorafenib (a RAF inhibitor used clinically) increased ERK1/2 phosphorylation in cardiomyocytes, and promoted hypertrophy, consistent with a 'RAF paradox' effect. Both promoted cardiac hypertrophy in mouse hearts in vivo, with increased cardiomyocyte size and no overt fibrosis. In conclusion, BRAF potentially plays an important role in human failing hearts, activation of BRAF is sufficient to induce hypertrophy, and Type 1 RAF inhibitors promote hypertrophy via the 'RAF paradox'. Cardiac hypertrophy resulting from these interventions was not associated with pathological features, suggesting that Type 1 RAF inhibitors may be useful to boost cardiomyocyte function.


Asunto(s)
Cardiomegalia/patología , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas B-raf/fisiología , Animales , Carbamatos/farmacología , Carbamatos/toxicidad , Cardiomegalia/metabolismo , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Dimerización , Técnicas de Sustitución del Gen , Insuficiencia Cardíaca/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Mutación Puntual , Conformación Proteica/efectos de los fármacos , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Sulfonamidas/toxicidad
2.
Curr Eye Res ; 47(1): 135-142, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34133251

RESUMEN

PURPOSE: This study aimed to investigate the effect of Raf-1 kinase inhibitory protein (RKIP) on diabetic retinal neurodegeneration in streptozotocin-treated rat model and high glucose-treated rat Müller cells. METHODS: Control and streptozotocin-treated rats were intravitreally injected with saline, RKIP gene overexpression lentivirus (oeRKIP) or negative control lentivirus (RKIP-vector). Normal or high glucose-treated Müller cells were transfected with saline, RKIP gene overexpression lentivirus or negative control lentivirus. Western blotting and immunofluorescence assay were utilized to evaluate the function of RKIP on the expression of RKIP, p38 mitogen-activated protein kinase (p38-MAPK), glutamate/aspartate transporter (GLAST), glutamine synthetase (GS), glial fibrillar acidic protein (GFAP) and cysteine-aspartic acid protease-3 (caspase-3). A glutamate assay kit was adopted to detect glutamate level in retina samples. Apoptosis of Müller cells was determined by Annexin-V/PI staining and flow cytometry. RESULTS: High glucose-treated Müller cells exhibited promoted apoptosis, while RKIP overexpression in high glucose-treated Müller cells down-regulated the enhanced apoptosis. Compared with rats injected with saline, streptozotocin-treated hyperglycemic rats displayed enhancement in the immunoreactivities of p38-MAPK and GFAP as well as in the protein expression of p38-MAPK and caspase-3. Strikingly, intravitreal injection of RKIP gene overexpression lentivirus in the hyperglycemic rats reversed the augmented immunoreactivities and protein expression mentioned above. Meanwhile, RKIP overexpression in the hyperglycemic rats improved the immunoreactivities and protein expression of RKIP, GS and GLAST. Besides, RKIP down-regulated the increased level of retinal glutamate in the hyperglycemic rats. CONCLUSIONS: Intravitreal injection of RKIP gene overexpression lentivirus functioned in preventing diabetic retinal neurodegeneration in a rat model of diabetes presumably by inhibiting p38-MAPK pathway.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental , Retinopatía Diabética/genética , Células Ependimogliales/patología , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas/genética , Proteínas Proto-Oncogénicas c-raf/genética , Animales , Retinopatía Diabética/diagnóstico , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Ratas , Ratas Sprague-Dawley
3.
Exp Dermatol ; 28(9): 1079-1082, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31338879

RESUMEN

Since Drosophila melanogaster has proven to be a useful model system to study phenotypes of oncogenic mutations and to identify new anti-cancer drugs, we generated human BRAFV600E homologous dRaf mutant (dRafA572E ) Drosophila melanogaster strains to use these for characterisation of mutant phenotypes and exploit these phenotypes for drug testing. For mutant gene expression, the GAL4/UAS expression system was used. dRafA572E was expressed tissue-specific in the eye, epidermis, heart, wings, secretory glands and in the whole animal. Expression of dRaf A572E under the control of an eye-specific driver led to semi-lethality and a rough eye phenotype. The vast majority of other tissue-specific and ubiquitous drivers led to a lethal phenotype only. The rough eye phenotype was used to test BRAF inhibitor vemurafenib and MEK1/2 inhibitor cobimetinib. There was no phenotype rescue by this treatment. However, a significant rescue of the lethal phenotype was observed under a gut-specific driver. Here, MEK1/2 inhibitor cobimetinib rescued Drosophila larvae to reach pupal stage in 37% of cases as compared to 1% in control experiments. Taken together, the BRAFV600E homolog dRaf A572E exerts mostly lethal effects in Drosophila. Gut-specific dRaf A572E expression might in future be developed further for drug testing.


Asunto(s)
Azetidinas/farmacología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-raf/genética , Animales , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/fisiología , Evaluación Preclínica de Medicamentos , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Intestinos/enzimología , Larva , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especificidad de Órganos , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/deficiencia , Proteínas Proto-Oncogénicas c-raf/fisiología , Vemurafenib/farmacología
4.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L241-L247, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29722566

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal fibrotic lung disease associated with aberrant activation and differentiation of fibroblasts, leading to abnormal extracellular matrix production. Currently, it is still an untreatable disease (except for lung transplantation). Here, we demonstrate that the Raf1 inhibitor GW5074 ameliorates lung fibrosis in bleomycin-induced pulmonary fibrosis. Posttreatment with GW5074 reduced fibronectin (FN) expression, collagen deposition, and inflammatory cell infiltration in bleomycin-challenged mice, suggesting an antifibrotic property of GW5074. To determine the molecular mechanisms by which inhibition of Raf1 ameliorates lung fibrosis, we investigated the role of Raf1 in TGF-ß1 signaling in human lung fibroblasts. GW5074 or downregulation of Raf1 by siRNAs significantly attenuated TGF-ß1-induced smooth muscle actin, FN, and collagen I expression, whereas overexpression of Raf1 promoted the effects of TGF-ß1 in lung fibroblasts. Furthermore, we found that Raf1-promoted TGF-ß1 signaling was through the Raf1/ERK/Smad pathway and contributed to the cell proliferation and migration in human lung fibroblasts. This study provides preclinical and mechanistic evidence for development of Raf1 inhibitors as potential antifibrotic drugs for the treatment of IPF.


Asunto(s)
Bleomicina/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Fibrosis Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bleomicina/farmacología , Humanos , Indoles/farmacología , Pulmón/metabolismo , Pulmón/patología , Ratones , Fenoles/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología
5.
BMC Cancer ; 16: 273, 2016 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-27085492

RESUMEN

BACKGROUND: Despite recent progress, investigating the impact of targeted therapies on Head and Neck Squamous Cell Carcinoma (HNSCC) remains a challenge. We investigated whether short-term culture of tumour fragments would permit the evaluation of tumour sensitivity to targeted therapies at the individual level. METHODS: We cultivated tumour slices prepared from 18 HNSCC tumour samples obtained during surgical resection. The samples were treated for 48 h with a panel of 8 targeted therapies directed against selected oncogenic transduction pathways. We analysed the cell proliferation index (CPI) of tumour cells using Ki67 labelling and the activation status of the RAF-MEK-ERK cascade through ERK phosphorylation analysis. RESULTS: Fourteen tumours were successfully analysed after short-term culture of tumour samples, revealing a striking individual heterogeneity of HNSCC in terms of tumour cell sensitivity to targeted therapies. Using 50% inhibition of CPI as threshold, sorafenib was shown to be active in 5/14 tumours. Cetuximab, the only approved targeted drug against HNSCC, was active in only 2/14 tumours. A more than 50% inhibition was observed with at least one drug out of the eight tested in 10/14 tumours. Cluster analysis was carried out in order to examine the effect of the drugs on cell proliferation and the RAF-MEK-ERK cascade. CONCLUSIONS: In vitro culture of tumour fragments allows for the evaluation of the effects of targeted therapies on freshly resected human tumours, and might be of value as a possible guide for the design of clinical trials and for the personalization of the medical treatment of HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Terapia Molecular Dirigida , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Cetuximab/administración & dosificación , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Antígeno Ki-67/biosíntesis , MAP Quinasa Quinasa 1/biosíntesis , MAP Quinasa Quinasa Quinasa 3/biosíntesis , Fosforilación , Medicina de Precisión , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Carcinoma de Células Escamosas de Cabeza y Cuello
6.
Int J Clin Exp Pathol ; 8(1): 608-14, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25755752

RESUMEN

Chordoma is a rare and low-malignant neoplasm which is considered to arise from notochord remnants. Due to its large resistance to chemotherapy and radiotherapy, surgical resection so far is the prior treatment for chordoma. However, the recurrence rate is high even after complete surgical resection. Recently, targeted cancer therapy has been demonstrated to be effective in several other tumors, while the related research on chordoma is rare. Mitogen-activated protein kinase signaling pathway is acknowledged to participate in tumor development, in which Raf-1 and extracellular regulated protein kinase 1/2 (ERK1/2) play vital roles. In this study, we evaluated the expression of Raf-1 and ERK1/2 by immunohistochemical staining in 42 chordoma tissue and 16 distant normal tissue. Moreover, we also investigated the correlations of Raf-1 and ERK1/2 expression with clinical features in sacral chordoma. Expression of Raf-1 and ERK1/2 was both significantly higher in sacral chordoma tissue than distant normal tissue (P = 0.008, P = 0.019). Raf-1 positive expression was related to surrounding muscle invasion (P = 0.032) and chordoma recurrence (P = 0.002), but the results did not indicate any association with patients' age, gender, tumor size and location. ERK1/2 was associated with tumor size (P = 0.044) instead of other clinical factors (P > 0.05). Spearman correlation test showed close relation between ERK1/2 and Raf-1 (P = 0.001, r = 0.518). Kaplan-Meier survival Curve and log-rank test showed that Raf-1 positive expression was associated with shorter continuous disease-free survival time (CDFS) (P = 0.001), while ERK1/2 had no relation to CDFS (P = 0.961). Conclusively, Raf-1 may be an important biomarker in predicting the prognosis of chordoma patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Cordoma/patología , Recurrencia Local de Neoplasia/patología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Sacro/patología , Neoplasias de la Columna Vertebral/patología , Adolescente , Adulto , Anciano , Cordoma/enzimología , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Recurrencia Local de Neoplasia/enzimología , Recurrencia Local de Neoplasia/mortalidad , Pronóstico , Sacro/enzimología , Neoplasias de la Columna Vertebral/enzimología , Neoplasias de la Columna Vertebral/mortalidad , Regulación hacia Arriba , Adulto Joven
7.
Oncol Rep ; 33(3): 1560-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25607597

RESUMEN

Hepatocellular carcinoma (HCC) is the sixth most common cancer and the second most lethal cancer worldwide. Evidence has shown HCC cell resistance to TRAIL­mediated apoptosis. In a previous study, we verified that silencing SNAIL downregulated the growth of HCC cells. In addition, the mechanism of resistance to TRAIL in HCC cells was connected with the activation of nuclear factor-κB (NF-κB). Thus, it was hypothesized that the downregultaion of SNAIL sensitizes HCC cells to TRAIL-induced apoptosis by regulating the NF-κB pathway. In the present study, the most effective lentiviral vectors carrying shRNA against SNAIL were selected and adenoviral vectors harboring TRAIL were constructed. The expression of SNAIL and TRAIL was detected by quantitative PCR and western blotting. HCC cell viability and apoptosis were assessed using an MTT assay and the Hoechst test. To determine how to sensitize HCC cells to TRAIL-induced apoptosis after silencing SNAIL, p53 was assessed by western blot analysis. We also investigated the expression of Bcl-xL, cIAP2, survivin and Raf-1 protein using western blot analysis and the apoptotic degree of HuH-7 cells was detected using the Hoechst test following the suppression of each gene, which was a possible molecular mechanism to sensitive TRAIL-induced apoptosis through the downregulation of SNAIL in HCC cells. Silencing SNAIL resulted in increased apoptosis by enhancing sensitization to TRAIL in all the HCC cells. Additionally, p53 protein was upregulated in HuH-7 cells. Expression of Bcl-xL, cIAP2, survivin and Raf-1 was downregulated following silencing of SNAIL, while down-regulation of any of the proteins contributed to SNAIL suppression enhancing HCC cell sensitivity to TRAIL­induced apoptosis, with the exception of cIAP2. The results demonstrated that silencing SNAIL can sensitize TRAIL-induced apoptosis in HCC cells by upregulating p53 protein and by regulating related genes of the NF-κB pathway such as Bcl-xL, survivin and Raf-1.


Asunto(s)
Apoptosis/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factores de Transcripción/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Células HEK293 , Células Hep G2 , Humanos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Neoplasias Hepáticas/genética , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción de la Familia Snail , Survivin , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Factores de Transcripción/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Proteína bcl-X/biosíntesis
8.
Int Immunol ; 27(4): 183-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25381666

RESUMEN

Primary Sjögren's syndrome (pSS) is a chronic autoimmune disorder of the exocrine glands with associated lymphocytic infiltrates in the affected glands. Dryness of the mouth and eyes results from involvement of the salivary and lacrimal glands. The efficacy of Rituximab (RTX) in pSS is still open to debate. This study delineates the signaling pathway involved in RTX-mediated down-regulation of pro-inflammatory factors in a co-culture system of pSS salivary gland epithelial cells (SGEC) with syngeneic pSS B-lymphocytes. In addition, the effects of RTX on the activation of the Raf-1/ERK1/2 pathway in pSS SGEC co-cultured with syngeneic pSS T-lymphocytes were also investigated. This study demonstrated that RTX may interfere with the ERK1/2 pathway in a syngeneic co-culture of pSS SGEC with pSS B-lymphocytes, leading to decreased cytokine production by SGEC. These novel findings reveal that syngeneic co-culture of pSS SGEC with pSS B-lymphocytes leads to a down-regulation of Raf-1 in epithelial cells that adversely regulates the activity of the ERK1/2 pathway and determines a subsequent reduction of the release of pro-inflammatory factors.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Rituximab/farmacología , Glándulas Salivales/inmunología , Síndrome de Sjögren/patología , Linfocitos B/citología , Linfocitos B/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Flavonoides/farmacología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , ARN Mensajero/biosíntesis , Glándulas Salivales/citología , Síndrome de Sjögren/tratamiento farmacológico , Linfocitos T/citología , Linfocitos T/inmunología
9.
Tumour Biol ; 36(2): 1313-21, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25355599

RESUMEN

Colorectal cancer (CRC, also known as colon cancer, rectal cancer, or bowel cancer) is the second leading cause of cancer mortality in the Western world. MicroRNAs (miRNAs) are a class of small (18-25 nucleotides long) noncoding RNAs with important posttranscriptional regulatory functions. miRNAs play important roles in various physiological and pathological processes including carcinogenesis in various solid cancers including CRC. In order to investigate the roles that miRNAs played in CRC, the expression of human miRNAs (in 20 normal adjacent tissue samples and 20 colon cancer samples) was examined in this study. miR-455, miR-484, and miR-101 were significantly downregulated in colon cancer samples. And overexpression of miR-455 significantly inhibited the proliferation and the invasion of SW480, but had no effect on apoptosis. PCR and Western blot showed that overexpression of miR-455 decreased protein expression of RAF proto-oncogene serine/threonine-protein kinase (RAF1) but had no effect on mRNA level. Luciferase assay indicated that miR-455 regulated RAF1 expression directly. Moreover, overexpression of RAF1 partially reversed the inhibitory effect of miR-455 on the growth and the invasion of SW480. The data indicated that miR-455 regulates the proliferation and invasion of colorectal cancer cells, at least in part, by downregulating RAF1, a direct target of miR-455. Collectively, our study demonstrated that miR-455-RAF1 may represent a new potential therapeutic target for colorectal carcinoma treatment.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/genética , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Apoptosis/genética , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Invasividad Neoplásica/genética , Proto-Oncogenes Mas
10.
Anticancer Res ; 34(12): 6945-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25503120

RESUMEN

Squamous cell carcinomas (SCC) make up 96% of all oral cancers. Most laboratory SCC studies grow cells as a monolayer, which does not accurately represent the disease in vivo. We used a more relevant multicellular spheroid (MCS) model to study this disease. The SCC9ß6KDFyn cell line, which expresses full-length ß6 and a kinase dead Fyn formed the largest MCS. Cell adhesive properties are dynamic and N-cadherin was increased in the largest MCS. c-Raf mediates the survival of tumor cells and was consistently expressed both in monolayers and in the MCS by SCC9ß6D1 cells which lack the ß6 cytoplasmic tail and, do not activate Fyn. SCC9ß6KDFyn cells also express high levels of c-Raf when grown as spheroids in which Fyn suppression stimulates MCS formation. Tumor microenvironment and growth patterns modulate cell behavior and suppression of Fyn kinase may promote MCS growth.


Asunto(s)
Carcinoma de Células Escamosas/patología , Cadenas beta de Integrinas/biosíntesis , Neoplasias de la Boca/patología , Proteínas Proto-Oncogénicas c-fyn/biosíntesis , Esferoides Celulares/patología , Cadherinas/biosíntesis , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Humanos , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Transducción de Señal , Células Tumorales Cultivadas , Microambiente Tumoral
11.
Proc Natl Acad Sci U S A ; 111(52): 18661-6, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512530

RESUMEN

Lung adenocarcinomas harboring activating mutations in the epidermal growth factor receptor (EGFR) represent a common molecular subset of non-small cell lung cancer (NSCLC) cases. EGFR mutations predict sensitivity to EGFR tyrosine kinase inhibitors (TKIs) and thus represent a dependency in NSCLCs harboring these alterations, but the genetic basis of EGFR dependence is not fully understood. Here, we applied an unbiased, ORF-based screen to identify genetic modifiers of EGFR dependence in EGFR-mutant NSCLC cells. This approach identified 18 kinase and kinase-related genes whose overexpression can substitute for EGFR in EGFR-dependent PC9 cells, and these genes include seven of nine Src family kinase genes, FGFR1, FGFR2, ITK, NTRK1, NTRK2, MOS, MST1R, and RAF1. A subset of these genes can complement loss of EGFR activity across multiple EGFR-dependent models. Unbiased gene-expression profiling of cells overexpressing EGFR bypass genes, together with targeted validation studies, reveals EGFR-independent activation of the MEK-ERK and phosphoinositide 3-kinase (PI3K)-AKT pathways. Combined inhibition of PI3K-mTOR and MEK restores EGFR dependence in cells expressing each of the 18 EGFR bypass genes. Together, these data uncover a broad spectrum of kinases capable of overcoming dependence on EGFR and underscore their convergence on the PI3K-AKT and MEK-ERK signaling axes in sustaining EGFR-independent survival.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Receptores ErbB/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/enzimología , Sistema de Señalización de MAP Quinasas , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-mos/biosíntesis , Proteínas Proto-Oncogénicas c-mos/genética , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor trkA/biosíntesis , Receptor trkA/genética , Receptor trkB
12.
Ross Fiziol Zh Im I M Sechenova ; 100(12): 1431-42, 2014 Dec.
Artículo en Ruso | MEDLINE | ID: mdl-25936182

RESUMEN

p53 is a transcription factor and then induced by cellular stress regulates ageing, cell cycle arrest and apoptosis. Published data also demonstrated that p53 participates in the regulation of neuronal differentiation. However, the data concerning the mechanisms of neuronal differentiation by p53 is very limited. In the present work we have studied a role and mechanisms of p53 activated by Nutlin-3 in the differentiation of PC12 cells. In our experiments Nutlin-3 stimulated the cell differentiation by significantly increasing of the neuritis upgrowth and upregulation of tyrosine hydroxylase (TH) expression, the catecholamine main rate-limited ferment. We have also analyzed MAPK signaling cascade that tightly participate in the neuronal differentiation. Obtained data demonstrated that activation of p53 by Nutlin-3 injections increased the activity of cRaf and ERK1/2, as well as transcription factor CREB, which is one of the targets for ERK pathway and regulates transcription of TH. Thus, we demonstrated that p53 activation lead to increased differentiation of PC12 cells and this effect is mediated by cRaf/ERK/CREB pathway.


Asunto(s)
Apoptosis/genética , Diferenciación Celular/genética , Proteína p53 Supresora de Tumor/genética , Tirosina 3-Monooxigenasa/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Catecolaminas/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Imidazoles/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/genética , Neuronas/metabolismo , Células PC12 , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/genética , Ratas , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
13.
Ann Pathol ; 33(6): 375-85, 2013 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24331719

RESUMEN

Cutaneous melanoma is a malignant tumor with a high metastatic potential. If an early treatment is associated with a favorable outcome, the prognosis of metastatic melanoma remains poor. Advances in molecular characterization of cancers, notably the discovery of BRAF gene mutations in metastatic melanoma, allowed to the recent development of targeted therapies against mutated BRAF protein. Despite high tumor response rates observed in clinical trials, these new drugs are associated with frequent secondary tumor resistance occurrence and paradoxical carcinogenic side effects. The cellular and molecular mechanisms of these carcinogenic side effects and secondary resistance are not yet fully elucidated and are actually intensely studied. This review of the literature focus on the mechanisms of these carcinogenic side effects and on the tumor resistance associated with anti-BRAF targeted therapies.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Indoles/efectos adversos , Indoles/farmacología , Leucemia/inducido químicamente , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/secundario , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Primarias Secundarias/inducido químicamente , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/inducido químicamente , Sulfonamidas/efectos adversos , Sulfonamidas/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/inducido químicamente , Resistencia a Antineoplásicos/genética , Activación Enzimática/efectos de los fármacos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes ras , Humanos , Indoles/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Queratoacantoma/inducido químicamente , Melanoma/inducido químicamente , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Modelos Biológicos , Terapia Molecular Dirigida , Mutación Missense , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/enzimología , Nevo Pigmentado/enzimología , Nevo Pigmentado/patología , Mutación Puntual , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/fisiología , Sulfonamidas/uso terapéutico , Microambiente Tumoral , Vemurafenib
14.
Oncol Rep ; 30(2): 877-89, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23760062

RESUMEN

Chemotherapy is an important option for the treatment of advanced breast cancer, but multidrug resistance is one of the major obstacles in the clinical control of breast cancer. The present study investigated the effects of the miR­195-led gene pathway in the sensitization of breast cancer cells to treatment with the chemotherapeutic drug Adriamycin. Breast cancer cell lines and tissue specimens (obtained from chemotherapy-sensitive or resistant patients) as well as a normal breast cell line were used to assess expression of miR-195, Raf-1, Bcl-2 and P-glycoprotein mRNA and/or mRNA. miR-195 mimics, inhibitor and Raf-1 siRNA were used to transfect breast cancer MCF-7 and MCF-7/ADR cells (an Adriamycin-resistant MCF-7 subline) for cell viability, apoptosis and gene expression analysis. The data showed that miR-195 expression was low in breast cancer cells and multidrug-resistant breast cancer tissues, which was associated with reduced Raf-1 expression in vitro and ex vivo. Induction of miR-195 expression promoted tumor cell apoptosis and inhibited breast cancer cell viability, but induced the sensitivity of breast cancer cells to Adriamycin treatment and was associated with inhibition of Raf-1 expression in breast cancer cells. Moreover, knockdown of Raf-1 expression had similar effects of miR-195 mimics on breast cancer cells, both of which were able to suppress Bcl-2 and P-glycoprotein expression in breast cancer cells. The data from the current study demonstrated that expression of miR-195 was inversely associated with Raf-1 expression in breast cancer cell lines and tissue specimens, and that Raf-1 is the target gene of miR-195. Thus, expression of miR-195 or knockdown of Raf-1 can similarly reduce tumor cell survival but increase apoptosis through downregulation of Raf-1 and Bcl-2 and P-glycoprotein expression. In conclusion, this gene pathway mediated the sensitivity of breast cancer cells to Adriamycin treatment.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Doxorrubicina/farmacología , MicroARNs/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , MicroARNs/biosíntesis , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/deficiencia , Proteínas Proto-Oncogénicas c-raf/genética , ARN Mensajero/genética , Regulación hacia Arriba
15.
Int J Biochem Cell Biol ; 45(2): 384-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23127800

RESUMEN

Ras proteins function as a point of convergence for different signalling pathways in eukaryotes and are involved in many cellular responses; their different subcellular locations could regulate distinct functions. To investigate the localization of active Ras in vivo in Saccharomyces cerevisiae, we expressed a probe consisting of a GFP fusion with a trimeric Ras binding domain of Raf1 (eGFP-RBD3), which binds Ras-GTP with a much higher affinity than Ras-GDP. Our results show that in wild type cells active Ras accumulates mainly at the plasma membrane and in the nucleus during growth on medium containing glucose, while it accumulates mainly in mitochondria in wild type glucose-starved cells and relocalizes to the plasma membrane and to the nucleus upon addition of this sugar. A similar pattern is observed in a strain deleted in the CYR1 gene indicating that the absence of adenylate cyclase does not impair the localization of Ras-GTP. Remarkably, in a gpa2Δ, but not in a gpr1Δ mutant, active Ras accumulates in internal membranes and mitochondria, both when cells are growing on glucose medium or are starved, indicating that Gpa2, but not Gpr1 is required for the recruitment of Ras-GTP at the plasma membrane and in the nucleus. Moreover, deletion of both HXK1 and HXK2 also causes a mitochondrial localization of the probe, which relocalizes to the plasma membrane and to the nucleus upon expression of HXK2 on a centromeric plasmid, suggesting that this kinase is involved in the proper localization of active Ras.


Asunto(s)
Membrana Celular/enzimología , Núcleo Celular/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas ras/metabolismo , Activación Enzimática , Fermentación , Subunidades alfa de la Proteína de Unión al GTP , Proteínas Fluorescentes Verdes/biosíntesis , Hexoquinasa/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/crecimiento & desarrollo , Análisis de la Célula Individual
16.
Cancer Sci ; 103(10): 1774-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22738312

RESUMEN

Overexpression of Raf-1 has commonly been observed in solid tumors including non-small cell lung cancer (NSCLC). The objective of this study was to investigate whether overexpression of Raf-1, phosphorylated-Raf-1 (p-Raf-1) or both correlates with poor survival rate in NSCLC patients and to explore associations between expression of these proteins and NSCLC cell fate both in vitro and in vivo. Expression of Raf-1 and p-Raf-1 were detected by immunohistochemistry in tumor specimens from 152 NSCLC patients and associations between their expression and the clinicopathological characteristics were assessed. Five-year median survival rate of patients were analyzed by Kaplan-Meier method, log-rank test and Cox regression. Cell fate was compared between normal tumor cells and those with Raf-1 silencing, in both the adenocarcinoma cell line A549 and xenografted mice that were infected with the A549 cell line. The incidence of overexpression of both Raf-1 and p-Raf-1 in NSCLC was much higher than normal control (P < 0.05), and the survival rate of patients with positive expression of Raf-1, p-Raf-1 or both was found to be significantly lower than the negative group (P < 0.05). Both univariate and multivariate analyses showed Raf-1 (P = 0.000, P = 0.010), p-Raf-1 (P = 0.004, P = 0.046), or both (P = 0.001, P = 0.016) was good prognostic markers for poor survival rate in NSCLC patients. Suppression of Raf-1 inhibited tumorigenesis by inducing apoptosis both in vitro and in vivo. These findings demonstrate that overexpression of Raf-1, p-Raf-1 or both could be considered as a new independent prognostic biomarker for poor survival rates for NSCLC patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/patología , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Fosforilación , Pronóstico , Modelos de Riesgos Proporcionales , Tasa de Supervivencia , Trasplante Heterólogo
17.
Biochem Biophys Res Commun ; 420(3): 611-5, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22445893

RESUMEN

The growth of thyroid cells is tightly regulated by thyroid stimulating hormone (TSH) through the cyclic adenosine 3', 5'-monophosphate (cAMP) signaling pathway by potentiating the mitogenic activity of insulin and insulin-like growth factors (IGFs). However, we recently reported that thyroglobulin (Tg), a major product of the thyroid, also induces the growth of thyroid cells cultured in 0.2% serum in the absence of TSH and insulin. In this report, we demonstrate that Tg induced phosphorylation of molecules of the c-Raf/MEK/ERK pathway of the mitogen-activated protein kinase (MAPK). The MEK-1/2 inhibitor PD98059 suppressed Tg-induced phosphorylation of ERK1/2 and reduced bromodeoxyuridine (BrdU) incorporation. Tg also induced expression of the essential transcriptional factors c-Myc, c-Fos and c-Jun and phosphorylation of the retinoblastoma (Rb) protein. The present results, together with the previous report, suggest that Tg utilizes multiple signaling cascades to induce thyroid cell growth independent of TSH/cAMP stimulation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Quinasas Quinasa Quinasa PAM/biosíntesis , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Tiroglobulina/farmacología , Glándula Tiroides/efectos de los fármacos , Animales , Línea Celular , Medio de Cultivo Libre de Suero/farmacología , Replicación del ADN/efectos de los fármacos , Activación Enzimática , Flavonoides/farmacología , Expresión Génica/efectos de los fármacos , Insulina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Glándula Tiroides/citología , Glándula Tiroides/enzimología , Tirotropina/farmacología
18.
Neurotoxicol Teratol ; 33(5): 530-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21784148

RESUMEN

Changes within the glucocorticoid receptor (GR) cellular signaling pathway were evaluated in adolescent mice exposed to 50 ppb arsenic during gestation. Previously, we reported increased basal plasma corticosterone levels, decreased hippocampal GR levels and deficits in learning and memory performance in perinatal arsenic-exposed mice. The biosynthesis of members of the mitogen-activated protein kinase (MAPK) signaling pathway, known to be involved in learning and memory, is modulated by the binding of GR to glucocorticoid response elements (GREs) in the gene promoters. Two genes of the MAPK pathway, Ras and Raf, contain GREs which are activated upon binding of GRs. We evaluated the activity of GRs at Ras and Raf promoters using chromatin immunoprecipitation and real-time PCR and report decreased binding of the GR at these promoters. An ELISA-based GR binding assay was used to explore whether this decreased binding was restricted to in vivo promoters and revealed no differences in binding of native GR to synthetic GREs. The decreased in vivo GR binding coincides with significantly decreased mRNA levels and slight reductions of protein of both H-Ras and Raf-1 in perinatally arsenic-exposed mice. Nuclear activated extracellular-signal regulated kinase (ERK), a downstream target of Ras and Raf, whose transcriptional targets also play an important role in learning and memory, was decreased in the hippocampus of arsenic-exposed animals when compared to controls. GR-mediated transcriptional deficits in the MAPK/ERK pathway could be an underlying cause of previously reported learning deficits and provide the link to arsenic-induced deficiencies in cognitive development.


Asunto(s)
Arsénico/toxicidad , Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/efectos de los fármacos , Animales , Western Blotting/métodos , Inmunoprecipitación de Cromatina/métodos , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Elementos de Respuesta/genética , Transducción de Señal/genética
19.
Cancer Lett ; 309(2): 199-208, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21705135

RESUMEN

RGS19 is a regulator of G protein signaling which is upregulated in ovarian cancers and its overexpression promotes cell proliferation in several mammalian cell types. Here we showed that cyclin D1/3 and Cdk6 were upregulated in HEK293 cells overexpressing RGS19, while INK4A and INK4B were reduced. Moreover, RGS19 augmented serum-stimulated PTEN/PDK/Akt and Rb phosphorylations in 293/RGS19 and Caco2/RGS19 cells. These changes were reversed upon the knockdown of RGS19. Consistent with an elevated Akt activity, increased levels of phosphorylated Bad and c-Raf and a diminished expression of TSC2 were detected, thus demonstrating that RGS19 can deregulate cell proliferation via multiple pathways.


Asunto(s)
Ciclo Celular , Proliferación Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas RGS/metabolismo , Ciclina D1/biosíntesis , Ciclina D1/genética , Ciclina D3/biosíntesis , Ciclina D3/genética , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Células HEK293 , Células HeLa , Humanos , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Proteínas RGS/biosíntesis , Proteínas RGS/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/biosíntesis , Proteína Letal Asociada a bcl/biosíntesis
20.
Anticancer Res ; 31(2): 549-53, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21378337

RESUMEN

AIM: Expression of mitogen-activated protein kinase (MAPK) signaling and its role in cell proliferation of the bone malignancies, osteosarcoma (OS) and malignant fibrous histiocytoma (MFH) were investigated. MATERIALS AND METHODS: Gene expression and protein levels of RAF1 and MEK1/2 in 6 human sarcoma cell lines and 7 surgically obtained OS specimens were assessed by RT-PCR and immunohistochemistry, respectively. MEK inhibitor, U0126 [1,4-diamino-2,3-dicyano-1,4-bis (2-aminophynyltio) butadiene], was used for cell proliferation assays. RESULTS: RAF1 and MEK 1/2 mRNA was detected in all cell lines and OS specimens. RAF1, MEK 1/2 and p-MEK protein was also expressed in the cells, as was MEK1/2 in OS specimens. Treatment with U0126 resulted in dose- and time-dependent inhibition of cell proliferation and suppression of p-ERK expression, opposite to promotion of p-MEK. CONCLUSION: U0126 blocks MAPK signaling and decreases cell proliferation in OS and MFH. Thus, selective MAPK inhibitors might be therapeutically advantageous in the treatment of bone and soft tissue sarcomas.


Asunto(s)
Neoplasias Óseas/enzimología , Histiocitoma Fibroso Maligno/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Osteosarcoma/enzimología , Western Blotting , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Butadienos/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Histiocitoma Fibroso Maligno/tratamiento farmacológico , Histiocitoma Fibroso Maligno/patología , Humanos , Inmunohistoquímica , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/biosíntesis , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/biosíntesis , MAP Quinasa Quinasa 2/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nitrilos/farmacología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...