Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360791

RESUMEN

Rhabdomyosarcoma (RMS) is a malignant soft tissue cancer that develops mostly in children and young adults. With regard to histopathology, four rhabdomyosarcoma types are distinguishable: embryonal, alveolar, pleomorphic and spindle/sclerosing. Currently, increased amounts of evidence indicate that not only gene mutations, but also epigenetic modifications may be involved in the development of RMS. Epigenomic changes regulate the chromatin architecture and affect the interaction between DNA strands, histones and chromatin binding proteins, thus, are able to control gene expression. The main aim of the study was to assess the role of protein arginine methyltransferases (PRMT) in the cellular biology of rhabdomyosarcoma. In the study we used two pan-inhibitors of PRMT, called AMI-1 and SAH, and evaluated their effects on proliferation and apoptosis of RMS cells. We observed that AMI-1 and SAH reduce the invasive phenotype of rhabdomyosarcoma cells by decreasing their proliferation rate, cell viability and ability to form cell colonies. In addition, microarray analysis revealed that these inhibitors attenuate the activity of the PI3K-Akt signaling pathway and affect expression of genes related to it.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Naftalenosulfonatos/farmacología , Proteína-Arginina N-Metiltransferasas , Rabdomiosarcoma , Transducción de Señal/efectos de los fármacos , Urea/análogos & derivados , Línea Celular Tumoral , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Urea/farmacología
2.
Pediatr Blood Cancer ; 67(6): e28222, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32207565

RESUMEN

BACKGROUND: Regorafenib is a small molecule multikinase inhibitor that inhibits multiple kinases including BRAF, KIT, PDGFRB, RAF, RET, and VEGFR1-3. PROCEDURES: The in vivo anticancer effects of regorafenib were assessed in a panel of six osteosarcoma models, three rhabdomyosarcoma models, and one Ewing sarcoma model. RESULTS: Regorafenib induced modest inhibition of tumor growth in the models evaluated. CONCLUSION: The overall pattern of response to regorafenib appears similar to that of the kinase inhibitor sorafenib, with pronounced slowing of tumor growth in some models, limited to the period of agent administration, being the primary treatment effect.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Sarcoma de Ewing/tratamiento farmacológico , Animales , Apoptosis , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Proliferación Celular , Niño , Femenino , Humanos , Ratones , Ratones SCID , Osteosarcoma/enzimología , Osteosarcoma/patología , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Anticancer Drugs ; 28(10): 1118-1125, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29045271

RESUMEN

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is often overexpressed in rhabdomyosarcoma (RMS). However, its oncogenic and functional role in RMS remains unclear. Therefore, we investigated the antitumor activity of LDK378 (ceritinib), a new second-generation ALK inhibitor approved for patients with ALK-positive non-small-cell lung cancers. Here, we report that LDK378 reduces cell viability and induces cell death in RMS cell lines at low micromolar IC50 concentrations irrespective of ALK expression levels or phosphorylation status. Compared with Karpas 299 non-Hodgkin's lymphoma cells carrying the NPM-ALK fusion gene, RMS cell lines proved to be far less sensitive to LDK378. The broad-range caspase inhibitor zVAD.fmk significantly protects RMS cells from LDK378-mediated cell death, indicating that LDK378 induces caspase-dependent apoptotic cell death. Before the onset of apoptosis, LDK378 reduces phosphorylation of AKT, S6 ribosomal protein, STAT3 and - to a lesser extent - phosphorylation of ERK, showing that it suppresses key survival pathways. Importantly, we identify a synergistic induction of cell death by combining subtoxic concentrations of LDK378 with the multitargeting kinase inhibitor sorafenib. Calculation of the combination index confirmed that this interaction is synergistic. Also, LDK378 cooperates with sorafenib to significantly reduce colony formation of RMS cells, showing that this combination affects long-term clonogenic growth. In conclusion, LDK378 induces caspase-dependent apoptotic cell death in RMS cells independent of their ALK status and synergizes at subtoxic concentrations with sorafenib to induce cell death. These findings have important implications for the use of LDK378 in RMS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/enzimología , Sulfonas/farmacología , Quinasa de Linfoma Anaplásico , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Niacinamida/administración & dosificación , Niacinamida/farmacología , Compuestos de Fenilurea/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Rabdomiosarcoma/patología , Sorafenib , Sulfonas/administración & dosificación
4.
Pediatr Blood Cancer ; 62(9): 1562-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26207356

RESUMEN

BACKGROUND: Sorafenib is an oral small molecule inhibitor of multiple kinases controlling tumor growth and angiogenesis. The purpose of the phase 2 study was to determine the response rate of sorafenib and gain further information on the associated toxicities, pharmacokinetics, and pharmacodynamics of sorafenib in children and young adults with relapsed or refractory tumors including rhabdomyosarcoma, Wilms tumor, hepatocellular carcinoma (HCC), and papillary thyroid carcinoma (PTC). PROCEDURE: Sorafenib, 200 mg/m(2) /dose, was administered every 12 hr continuously for 28 day cycles using a two-stage design in two primary strata (rhabdomyosarcoma and Wilms tumor) and two secondary strata (HCC and PTC). Correlative studies in consenting patients included determination of sorafenib steady state trough concentrations and assessments of VEGF and sVEGFR2. RESULTS: Twenty patients (median age of 11 years; range, 5-21) enrolled. No objective responses (RECIST) were observed in the 10 evaluable patients enrolled in each of the two primary disease strata of rhabdomyosarcoma and Wilms tumor. No patients with HCC or PTC were enrolled. Sorafenib was not associated with an excessive rate of dose-limiting toxicity (DLT). The mean ± SD steady state concentration during cycle 1 day 15 was 6.5 ± 3.9 µg/ml (n = 10). CONCLUSIONS: Sorafenib was well tolerated in children at 200 mg/m(2) /dose twice daily on a continuous regimen with toxicity profile and steady state drug concentrations similar to those previously reported. Single agent sorafenib was inactive in children with recurrent or refractory rhabdomyosarcoma or Wilms tumor.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Rabdomiosarcoma/tratamiento farmacológico , Terapia Recuperativa , Tumor de Wilms/tratamiento farmacológico , Adolescente , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/farmacocinética , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Niño , Preescolar , Femenino , Humanos , Neoplasias Renales/sangre , Neoplasias Renales/enzimología , Masculino , Proteínas de Neoplasias/sangre , Niacinamida/efectos adversos , Niacinamida/farmacocinética , Niacinamida/uso terapéutico , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/farmacocinética , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Rabdomiosarcoma/sangre , Rabdomiosarcoma/enzimología , Sorafenib , Insuficiencia del Tratamiento , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre , Tumor de Wilms/sangre , Tumor de Wilms/enzimología , Adulto Joven
5.
Ann Diagn Pathol ; 19(4): 239-42, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26005112

RESUMEN

The focus of this study was to investigate anaplastic lymphoma kinase (ALK) expression by immunohistochemistry using a highly specific antibody. Distribution and frequency of ALK expression may provide a clue for ALK inhibitor use in small round cell tumors of childhood. The study group involved 76 small round cell tumors of childhood, which composed of 11 rhabdomyosarcomas, 13 Wilms tumors, 7 Ewing sarcoma/primitive neuroectodermal tumors, 34 peripheral neuroblastic tumors, and 11 acute lymphoblastic lymphoma. Anaplastic lymphoma kinase protein expression in small round cell tumors of childhood is poorly described in the literature. The findings of our study highlight a potential and possible role of targeting ALK in pediatric solid tumors by using ALK immunohistochemistry. Anaplastic lymphoma kinase may also have an oncogenic role in rhabdomyosarcomas and peripheral neuroblastic tumors, and they may possibly be treated with ALK inhibitors. Anaplastic lymphoma kinase expression in Wilms tumors is not reported in the literature, previously. Our study evaluated ALK expression in Wilms tumor samples.


Asunto(s)
Tumor Desmoplásico de Células Pequeñas Redondas/enzimología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Quinasa de Linfoma Anaplásico , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Carcinoma de Células Pequeñas/enzimología , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Niño , Preescolar , Tumor Desmoplásico de Células Pequeñas Redondas/genética , Tumor Desmoplásico de Células Pequeñas Redondas/patología , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Neuroblastoma/enzimología , Neuroblastoma/genética , Neuroblastoma/patología , Tumores Neuroectodérmicos Primitivos/enzimología , Tumores Neuroectodérmicos Primitivos/genética , Tumores Neuroectodérmicos Primitivos/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Tirosina Quinasas Receptoras/genética , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Tumor de Wilms/enzimología , Tumor de Wilms/genética , Tumor de Wilms/patología
6.
Br J Cancer ; 109(12): 3084-91, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24149177

RESUMEN

BACKGROUND: Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase aberrantly expressed in cancer, but its clinical and functional importance remain controversial. Mutation or amplification of ALK, as well as its expression levels assessed by conventional immunohistochemistry methods, has been linked to prognosis in cancer, although with potential bias because of the semi-quantitative approaches. Herein, we measured ALK mRNA expression in rhabdomyosarcoma (RMS) and determined its clinical impact on patients' stratification and outcome. METHODS: Specimens were obtained from RMS patients and cell lines, and ALK expression was analysed by quantitative RT-PCR, western blotting, IHC, and copy number analysis. RESULTS: High ALK mRNA expression was detected in the vast majority of PAX3/7-FOXO1-positive tumours, whereas PAX3/7-FOXO1-negative RMS displayed considerably lower amounts of both mRNA and protein. Notably, ALK mRNA distinguished unfavourable PAX3/7-FOXO1-positive tumours from PAX3/7-FOXO1-negative RMS (P<0.0001), and also correlated with larger tumour size (P<0.05) and advanced clinical stage (P<0.01), independently of fusion gene status. High ALK mRNA levels were of prognostic relevance by Cox univariate regression analysis and correlated with increased risk of relapse (P=0.001) and survival (P=0.01), whereas by multivariate analysis elevated ALK mRNA expression resulted a negative prognostic marker when clinical stage was not included. CONCLUSION: Quantitative assessment of ALK mRNA expression helps to improve risk stratification of RMS patients and identifies tumours with adverse biological characteristics and aggressive behaviour.


Asunto(s)
ARN Mensajero/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Rabdomiosarcoma/enzimología , Quinasa de Linfoma Anaplásico , Línea Celular Tumoral , Niño , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Pronóstico , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Análisis de Supervivencia
7.
J Transl Med ; 9: 64, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21575221

RESUMEN

BACKGROUND: c-Met is a receptor tyrosine kinase (RTK) that is over-expressed in a variety of cancers and involved in cell growth, invasion, metastasis and angiogenesis. In this study, we investigated the role of c-Met in rhabdomyosarcoma (RMS) using its small molecule inhibitor SU11274, which has been hypothesized to be a potential therapeutic target for RMS. METHODS: The expression level of phosphorylated c-Met in RMS cell lines (RD, CW9019 and RH30) and tumor tissues was assessed by phospho-RTK array and immunohistochemistry, respectively. The inhibition effects of SU11274 on RMS cells were studied with regard to intracellular signaling, cell proliferation, cell cycle and cell migration. RESULTS: A high level of phosphorylated c-Met was detected in 2 alveolar RMS cell lines (CW9019 and RH30) and 14 out of 24 RMS tissue samples, whereas relatively low levels of phospho-c-Met were observed in the embryonic RMS cell line (RD). The small molecule SU11274 could significantly reduce the phosphorylation of c-Met, resulting in inhibition of cell proliferation, G1 phase arrest of cell cycle and blocking of cell migration in CW9019 and RH30 cell lines. CONCLUSION: These results might support the role of c-Met in the development and progression of RMS. Furthermore, the inhibitor of c-Met, SU11274, could be an effective targeting therapy reagent for RMS, especially alveolar RMS.


Asunto(s)
Indoles/farmacología , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Rabdomiosarcoma/enzimología , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/farmacología , Apoptosis/efectos de los fármacos , Recuento de Células , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/metabolismo , Rabdomiosarcoma/patología , Transducción de Señal/efectos de los fármacos
8.
J Clin Invest ; 131(15)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34166228

RESUMEN

The 12q13-q14 chromosomal region is recurrently amplified in 25% of fusion-positive (FP) rhabdomyosarcoma (RMS) cases and is associated with a poor prognosis. To identify amplified oncogenes in FP RMS, we compared the size, gene composition, and expression of 12q13-q14 amplicons in FP RMS with those of other cancer categories (glioblastoma multiforme, lung adenocarcinoma, and liposarcoma) in which 12q13-q14 amplification frequently occurs. We uncovered a 0.2 Mb region that is commonly amplified across these cancers and includes CDK4 and 6 other genes that are overexpressed in amplicon-positive samples. Additionally, we identified a 0.5 Mb segment that is only recurrently amplified in FP RMS and includes 4 genes that are overexpressed in amplicon-positive RMS. Among these genes, only serine hydroxymethyltransferase 2 (SHMT2) was overexpressed at the protein level in an amplicon-positive RMS cell line. SHMT2 knockdown in amplicon-positive RMS cells suppressed growth, transformation, and tumorigenesis, whereas overexpression in amplicon-negative RMS cells promoted these phenotypes. High SHMT2 expression reduced sensitivity of FP RMS cells to SHIN1, a direct SHMT2 inhibitor, but sensitized cells to pemetrexed, an inhibitor of the folate cycle. In conclusion, our study demonstrates that SHMT2 contributes to tumorigenesis in FP RMS and that SHMT2 amplification predicts differential response to drugs targeting this metabolic pathway.


Asunto(s)
Carcinogénesis , Cromosomas Humanos Par 12 , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glicina Hidroximetiltransferasa , Proteínas de Neoplasias , Rabdomiosarcoma , Carcinogénesis/genética , Carcinogénesis/metabolismo , Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 12/metabolismo , Femenino , Glicina Hidroximetiltransferasa/biosíntesis , Glicina Hidroximetiltransferasa/genética , Humanos , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/genética
9.
J Cell Biochem ; 110(1): 162-70, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20213728

RESUMEN

CARM1/PRMT4 is a member of the protein arginine methyltransferase (PRMT) family. CARM1 as a transcriptional coactivator plays an active role on mammalian genes. Here, we show that CARM1 can be recruited to the promoter of myogenin gene to enhance its transcriptional activation via PCAF at the early stage of TPA-induced RD cell differentiation. By adding adenosine dialdehyde, AdOx, to inhibit the PRMT in RD cells, the TPA-induced recruiting of p300, PCAF and the Brg1 at the myogenin promoter is abolished and myogenic differentiation is blocked. More specifically, the expression of PCAF and its nucleation are prohibited when CARM1 is knockdown by its specific siRNA. We suggest that the physical interaction of CARM1 and PCAF is likely pivotal for the activation of PCAF in the downstream of CARM1 pathway for inducing myogenin under TPA-induced differentiation. The findings shed lights on novel therapeutic targets in the treatment of rhabdomyosarcoma patients.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Miogenina/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Rabdomiosarcoma/enzimología , Acetato de Tetradecanoilforbol/farmacología , Activación Transcripcional/efectos de los fármacos , Factores de Transcripción p300-CBP/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Diferenciación Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Miogenina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Factores de Transcripción p300-CBP/genética
10.
Biol Pharm Bull ; 33(9): 1568-73, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20823576

RESUMEN

Skeletal muscle is the major producer of lactic acid in the body, but its oxidative fibers also use lactic acid as a respiratory fuel. Monocarboxylate transporter (MCT) 1 has been suggested to play a major role in influx of L-lactic acid for oxidation. The regulation mechanism of MCT1 was characterized utilizing rhabdomyosarcoma cells as an in vitro skeletal muscle model. The uptake of L-lactic acid via MCT1 was studied in the presence of various intracellular regulatory pathways, including pathways mediated by protein kinases A, C and G (PKA, PKC and PKG), protein tyrosine kinase (PTK), and Ca2+/calmodulin modulators. The results showed that PKG-, PTK-, and Ca2+/calmodulin-mediated regulatory pathways play no role in the regulation of L-lactic acid uptake, but a role for PKC- and PKA-mediated pathways was apparent. Uptake of L-lactic acid appeared to be stimulated by phorbol 12-myristate 13-acetate (PMA, a PKC activator) via an increase in Vmax of transport processes with no alteration in Km. In parallel, PMA treatment also resulted in an increase in the level of MCT1 expression. On the other hand, exposure to 8-Br-cAMP, a cAMP analog, and to forskolin, an adenylyl cyclase activator, resulted in a significant decrease in L-lactic acid uptake. Additionally, 8-Br-cAMP reduced Vmax but not Km values. Parallel to the decrease in Vmax of L-lactic acid uptake, the level of MCT1 expression was decreased in response to incubation with 8-Br-cAMP. These results indicate the possible involvement of a PKC- and PKA-mediated pathway associated with expression of MCT1 and lactate transport.


Asunto(s)
Líquido Intracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Simportadores/metabolismo , Transporte Biológico Activo/fisiología , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Humanos , Líquido Intracelular/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Transportadores de Ácidos Monocarboxílicos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Proteína Quinasa C/fisiología , Transporte de Proteínas/fisiología , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Simportadores/biosíntesis , Simportadores/fisiología
11.
Cell Biol Int ; 33(9): 1020-5, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19524683

RESUMEN

Sialidase Neu2 is a glycohydrolytic enzyme whose tissue distribution has been detected principally in differentiated skeletal muscle. In this study we show that Neu2 expression is absent in different embryonal and alveolar human tumor rhabdomyosarcoma (RMS) cells, which are genetically committed myoblasts characterized by delayed differentiation. Forced myogenic differentiation of an embryonal RMS cell line, as obtained via pharmacological and genetic p38 activation or via follistatin overexpression, was characterized by Neu2 loss of expression despite the significant rise of different muscle-specific markers, suggesting therefore that the defective myogenic program of RMS cells is accompanied by Neu2 suppression.


Asunto(s)
Desarrollo de Músculos , Mioblastos Esqueléticos/enzimología , Neuraminidasa/metabolismo , Rabdomiosarcoma/enzimología , Caveolina 3/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Folistatina/metabolismo , Humanos , MAP Quinasa Quinasa 6/metabolismo , Mioblastos Esqueléticos/patología , Cadenas Pesadas de Miosina/metabolismo , Neuraminidasa/genética , Rabdomiosarcoma/patología , Acetato de Tetradecanoilforbol/farmacología , Transfección
12.
Cancer Res ; 67(6): 2800-8, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363602

RESUMEN

Sarcomas are rare malignant mesenchymal tumors for which there are limited treatment options. One potential molecular target for sarcoma treatment is the Src tyrosine kinase. Dasatinib (BMS-354825), a small-molecule inhibitor of Src kinase activity, is a promising cancer therapeutic agent with p.o. bioavailability. Dasatinib exhibits antitumor effects in cultured human cell lines derived from epithelial tumors, including prostate and lung carcinomas. However, the action of dasatinib in mesenchymally derived tumors has yet to be shown. Based on our previous findings of Src activation in human sarcomas, we evaluated the effects of dasatinib in 12 cultured human sarcoma cell lines derived from bone and soft tissue sarcomas. Dasatinib inhibited Src kinase activity at nanomolar concentrations in these sarcoma cell lines. Downstream components of Src signaling, including focal adhesion kinase and Crk-associated substrate (p130(CAS)), were also inhibited at similar concentrations. This inhibition of Src signaling was accompanied by blockade of cell migration and invasion. Moreover, apoptosis was induced in the osteosarcoma and Ewing's subset of bone sarcomas at nanomolar concentrations of dasatinib. Inhibition of Src protein expression by small interfering RNA also induced apoptosis, indicating that these bone sarcoma cell lines are dependent on Src activity for survival. These results show that dasatinib inhibits migration and invasion of diverse sarcoma cell types and selectively blocks the survival of bone sarcoma cells. Therefore, dasatinib may provide therapeutic benefit by preventing the growth and metastasis of sarcomas in patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Apoptosis/fisiología , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Línea Celular Tumoral , Dasatinib , Activación Enzimática , Humanos , Invasividad Neoplásica , Osteosarcoma/enzimología , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/patología , Transducción de Señal/efectos de los fármacos
13.
Cancer Lett ; 442: 341-350, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30447254

RESUMEN

Overactivation of the Hedgehog (HH) signaling pathway is implicated in many cancers. In this study, we demonstrate that the small molecule RITA, a p53 activator, effectively downregulates HH signaling in human medulloblastoma and rhabdomyosarcoma cells irrespective of p53. This is mediated by a ROS-independent activation of the MAP kinase JNK. We also show that in vitro RITA sensitized cells to the GLI antagonist GANT61, as co-administration of the two drugs had more pronounced effects on cell proliferation and apoptosis. In vivo administration of RITA or GANT61 suppressed rhabdomyosarcoma xenograft growth in nude mice; however, co-administration did not further enhance tumor suppression, even though cell proliferation was decreased. RITA was more potent than GANT61 in downregulating HH target gene expression; surprisingly, this suppressive effect was almost completely eliminated when the two drugs were administered together. Notably, RNA-seq demonstrated a broader response of pathways involved in cancer cell growth in the combination treatment, providing a plausible interpretation for tumor reduction in the absence of HH signaling downregulation.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Furanos/farmacología , Proteínas Hedgehog/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Meduloblastoma/tratamiento farmacológico , Rabdomiosarcoma/tratamiento farmacológico , Proteína p53 Supresora de Tumor/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/enzimología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Femenino , Proteínas Hedgehog/genética , Humanos , Meduloblastoma/enzimología , Meduloblastoma/genética , Meduloblastoma/patología , Ratones Desnudos , Piridinas/farmacología , Pirimidinas/farmacología , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína con Dedos de Zinc GLI1/análisis , Proteína con Dedos de Zinc GLI1/genética
14.
J Neuropathol Exp Neurol ; 77(10): 964-972, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184235

RESUMEN

The human rhabdomyosarcoma cell line TE671 has been used extensively to study different aspects of muscle biology. However, its ability to differentiate and form myotubes has not been explored. Here, we examined muscle differentiation when we specifically stopped proliferation of human TE671 (WT-TE671) cells by using 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene (U0126), an MAPK inhibitor. Our data show that treated cells initiated fusion, and myotube formation and that expression levels of dysferlin and myogenin were increased, whereas those of pax7 were decreased. Treatment of WT-TE671 cells with vitamin D3 alone and cotreatment with U0126 also promoted dysferlin expression. In addition, we knocked out the DYSF gene, which is involved in muscle differentiation, using CRISPR/Cas9 technology in WT-TE671 cells (Dysf-KO TE671). No dysferlin expression was observed before and after U0126 treatment. Although myogenin expression was absent in vehicle-treated Dysf-KO TE671 cells, after addition of U0126, myogenin reached levels similar to WT-TE671. This widely available source of human cells appropriately treated with U0126 may represent a useful model to study human muscle physiology in vitro. This dysferlin-deficient cell line should allow the study of pathophysiological pathways involved in dysferlin-deficient muscle and constitute a tool for high-throughput screening of therapeutic compounds for patients with dysferlinopathy and other muscle diseases.


Asunto(s)
Butadienos/farmacología , Proteína 9 Asociada a CRISPR/metabolismo , Diferenciación Celular/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Enfermedades Musculares/enzimología , Nitrilos/farmacología , Rabdomiosarcoma/enzimología , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/fisiología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología
15.
Cancer Chemother Pharmacol ; 82(2): 251-263, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29855693

RESUMEN

PURPOSE: The anaplastic lymphoma kinase (ALK) has been demonstrated to be a valid clinical target in diseases such as anaplastic large cell lymphoma and non-small cell lung cancer. Recent studies have indicated that ALK is overexpressed in pediatric rhabdomyosarcoma (RMS) and hence we hypothesized that this kinase may be a suitable candidate for therapeutic intervention in this tumor. METHODS: We evaluated the expression of ALK in a panel of pediatric RMS cell lines and patient-derived xenografts (PDX), and sensitivity to ALK inhibitors was assessed both in vitro and in vivo. RESULTS: Essentially, all RMS lines were sensitive to crizotinib, NVP-TAE684 or LDK-378 in vitro, and molecular analyses demonstrated inhibition of RMS cell proliferation following siRNA-mediated reduction of ALK expression. However, in vivo PDX studies using ALK kinase inhibitors demonstrated no antitumor activity when used as single agents or when combined with standard of care therapy (vincristine, actinomycin D and cyclophosphamide). More alarmingly, however, crizotinib actually accelerated the growth of these tumors in vivo. CONCLUSIONS: While ALK appears to be a relevant target in RMS in vitro, targeting this kinase in vivo yields no therapeutic efficacy, warranting extreme caution when considering the use of these agents in pediatric RMS patients.


Asunto(s)
Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico/biosíntesis , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/enzimología , Quinasa de Linfoma Anaplásico/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Crizotinib/administración & dosificación , Crizotinib/farmacología , Ciclofosfamida/administración & dosificación , Dactinomicina/administración & dosificación , Interacciones Farmacológicas , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Pirimidinas/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Rabdomiosarcoma/genética , Transfección , Vincristina/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Sci Transl Med ; 10(448)2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973406

RESUMEN

The RAS isoforms are frequently mutated in many types of human cancers, including PAX3/PAX7 fusion-negative rhabdomyosarcoma. Pediatric RMS arises from skeletal muscle progenitor cells that have failed to differentiate normally. The role of mutant RAS in this differentiation blockade is incompletely understood. We demonstrate that oncogenic RAS, acting through the RAF-MEK [mitogen-activated protein kinase (MAPK) kinase]-ERK (extracellular signal-regulated kinase) MAPK effector pathway, inhibits myogenic differentiation in rhabdomyosarcoma by repressing the expression of the prodifferentiation myogenic transcription factor, MYOG. This repression is mediated by ERK2-dependent promoter-proximal stalling of RNA polymerase II at the MYOG locus. Small-molecule screening with a library of mechanistically defined inhibitors showed that RAS-driven RMS is vulnerable to MEK inhibition. MEK inhibition with trametinib leads to the loss of ERK2 at the MYOG promoter and releases the transcriptional stalling of MYOG expression. MYOG subsequently opens chromatin and establishes super-enhancers at genes required for late myogenic differentiation. Furthermore, trametinib, in combination with an inhibitor of IGF1R, potently decreases rhabdomyosarcoma cell viability and slows tumor growth in xenograft models. Therefore, this combination represents a potential therapeutic for RAS-mutated rhabdomyosarcoma.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Genes ras , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Miogenina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Rabdomiosarcoma/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Mioblastos/metabolismo , Mioblastos/patología , Proteínas de Fusión Oncogénica/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Receptor IGF Tipo 1/metabolismo , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Transcripción Genética/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Clin Invest ; 59(5): 889-99, 1977 May.
Artículo en Inglés | MEDLINE | ID: mdl-265945

RESUMEN

In the present study, terminal deoxynucleotidyltransferase was examined in the peripheral blood and (or) bone marrow of 115 children with a variety of neoplastic, hematologic, and other unrelated disorders. Terminal deoxynucleotidyltransferase activity was present at 4.08+/-0.74 U/108 cells in 23 morphologicall normal bone marrow samples from childhood controls. Terminal transferase was present at greater than 23 U/108 nucleated cells and at greater than31 U/108 blasts in the bone marrow of all children with acute lymphoblastic leukemia studied at initial diagnosis and at disease relapse. Terminal deoxynucleotidyltransferase was detectable at low levels, less than 7.5 U/108 cells, in all remission marrow smaples. Bone marrow terminal transferase activity was markedly elevated in all untreated acute lymphoblastic leukemia patients, whereas low levels which were difficult to interpret were present in the peripheral blood samples of two patients at diagnosis and six patients at relapse who had low absolute lymphoblast counts. Because of greater variation in the lymphoblast content of peripheral blood, bone marrow assays are more reliable in detecting disease activity. Marrow terminal deoxynucleotidyltransferase values obtained during the active phase of acute lymphoblastic leukemia were significantly greater than those found in other types of leukemia, bone marrow malignancies, and hematologic disorders. Terminal transferase determinations in blast cells of two patients with leukemic conversion of non-Hodgkin's lymphoma and in tumor cells from one patient with Burkitt's lymphoma were within the control range. These dat further define the usefulness of terminal deoxynucleotidyltrnasferase assay in the differentiation and classication of hematologic malignancies.


Asunto(s)
Células de la Médula Ósea , Médula Ósea/enzimología , ADN Nucleotidiltransferasas/análisis , Leucemia Linfoide/enzimología , Linfocitos/enzimología , Neoplasias/enzimología , Adolescente , Anemia Aplásica/enzimología , Niño , Preescolar , ADN Nucleotidiltransferasas/sangre , Humanos , Leucemia Mieloide Aguda/enzimología , Linfoma/enzimología , Neuroblastoma/enzimología , Oligonucleótidos , Púrpura Trombocitopénica/enzimología , Recurrencia , Remisión Espontánea , Rabdomiosarcoma/enzimología
18.
Cancer Res ; 65(9): 3868-76, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867386

RESUMEN

Identification of new drugs is strongly needed for sarcomas. Insulin-like growth factor-I receptor (IGF-IR) was found to provide a major contribution to the malignant behavior of these tumors, therefore representing a very promising therapeutic target. In this study, we analyzed the therapeutic potential of a novel kinase inhibitor of IGF-IR, NVP-AEW541, in Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma, the three most frequent solid tumors in children and adolescents. NVP-AEW541 inhibits IGF-I-mediated receptor activation and downstream signaling. Ewing's sarcoma cells were generally found to be more sensitive to the effects of this drug compared with rhabdomyosarcoma and osteosarcoma, in agreement with the high dependency of this neoplasm to IGF-IR signaling. NVP-AEW541 induced a G1 cell cycle block in all cells tested, whereas apoptosis was observed only in those cells that show a high level of sensitivity. Concurrent exposure of cells to NVP-AEW541 and other chemotherapeutic agents resulted in positive interactions with vincristine, actinomycin D, and ifosfamide and subadditive effects with doxorubicin and cisplatin. Accordingly, combined treatment with NVP-AEW541 and vincristine significantly inhibited tumor growth of Ewing's sarcoma xenografts in nude mice. Therefore, results encourage inclusion of this drug especially in the treatment of patients with Ewing's sarcoma. For the broadest applicability and best efficacy in sarcomas, NVP-AEW541 may be combined with vincristine, actinomycin D, and ifosfamide, three major drugs in the treatment of sarcomas.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Sarcoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/enzimología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/enzimología , Receptor IGF Tipo 1/fisiología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/enzimología , Sarcoma/enzimología , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/enzimología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vincristina/administración & dosificación
19.
Mol Cancer Ther ; 5(5): 1299-308, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16731763

RESUMEN

Myoblast cell cycle exit and differentiation are mediated in part by down-regulation of cyclin D1 and associated cyclin-dependent kinase (Cdk) activity. Because rhabdomyosarcoma may represent a malignant tumor composed of myoblast-like cells failing to exit the cell cycle and differentiate, we considered whether excess Cdk activity might contribute to this biology. Cyclin D-dependent Cdk4 and Cdk6 were expressed in most of a panel of six human rhabdomyosarcoma-derived cell lines. Cdk4 was expressed in 73% of alveolar and embryonal rhabdomyosarcoma tumors evaluated using a human tissue microarray. When challenged to differentiate by mitogen deprivation in vitro, mouse C2C12 myoblasts arrested in G(1) phase of the cell cycle, whereas four in the panel of rhabdomyosarcoma cell lines failed to do so. C2C12 myoblasts maintained in mitogen-rich media and exposed to a Cdk4/Cdk6 inhibitor PD 0332991 accumulated in G(1) cell cycle phase. Similar treatment of rhabdomyosarcoma cell lines caused G(1) arrest and prevented cell accumulation in vitro, and it delayed growth of rhabdomyosarcoma xenografts in vivo. Consistent with a role for Cdk4/Cdk6 activity as a regulator of myogenic differentiation, we observed that PD 0332991 exposure promoted morphologic changes and enhanced the expression of muscle-specific proteins in cultured myoblasts and in the Rh30 cell line. Our findings support the concept that pharmacologic inhibition of Cdk4/Cdk6 may represent a useful therapeutic strategy to control cell proliferation and possibly promote myogenic differentiation in rhabdomyosarcoma.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Mioblastos Esqueléticos/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Rabdomiosarcoma/enzimología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Fase G1/efectos de los fármacos , Humanos , Ratones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/enzimología , Piperazinas/metabolismo , Piridinas/metabolismo , Células Tumorales Cultivadas
20.
Anticancer Res ; 26(1A): 351-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16475717

RESUMEN

The prognosis for patients with chemo-refractory rhabdomyosarcoma remains poor. The tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a hopeful candidate for new strategies in chemotherapy. The effects of TRAIL and melphalan (Mel) in the rhabdomyosarcoma cell line TE-671 were investigated by colorimetric caspase assays and flow cytometry. TRAIL induced the activation of caspases-2, -3 and -8, but not the activation of caspase-9, in the Mel-resistant TE-671 cells. Inhibition of caspase-2 with the caspase-2 inhibitor z-VDVAD-fmk significantly down-regulated the TRAIL-induced caspase-3 activation, as well as the TRAIL-induced cytotoxicity. When TE-671 cells were treated with a combination of Mel and TRAIL, a significant synergism of drug-induced cytotoxicity was obtained. The inhibition of caspase-2 could completely abolish caspase-3 activation, suggesting that TRAIL sensitises TE-671 cells for Mel-induced cytotoxicity via a caspase-2- and -3-dependent mechanism. In conclusion, it was shown, for the first time, that TRAIL could sensitise Mel-resistant tumour cells to melphalan.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Reguladoras de la Apoptosis/farmacología , Caspasas/metabolismo , Melfalán/farmacología , Glicoproteínas de Membrana/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Reguladoras de la Apoptosis/administración & dosificación , Caspasa 2 , Inhibidores de Caspasas , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Activación Enzimática , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Melfalán/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA