Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Genet ; 23(2): 222-7, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10508522

RESUMEN

Chromosomal translocations resulting in the expression of chimaeric transcription factors are frequently observed in tumour cells, and have been suggested to be a common mechanism in human carcinogenesis. Ewing sarcoma and related peripheral primitive neuroectodermal tumours share recurrent translocations that fuse the gene EWSR1 (formerly EWS) from 22q-12 to FLI1 and genes encoding other ETS transcription factors (which bind DNA through the conserved ETS domain). It has been shown that transduction of the gene EWSR1-FLI1 (encoding EWS-FLI1 protein) can transform NIH3T3 cells, and that mutants containing a deletion in either the EWS domain or the DNA-binding domain in FLI1 lose this ability. This indicates that the EWS-FLI1 fusion protein may act as an aberrant transcription factor, but the exact mechanism of oncogenesis remains unknown. Because ETS transcription factors regulate expression of TGFBR2 (encoding the TGF-beta type II receptor, TGF-beta RII; Refs 9,14), a putative tumour suppressor gene, we hypothesized that TGFBR2 may be a target of the EWS-FLI1 fusion protein. We show here that Ewing sarcoma [corrected] (ES) cell lines with the EWSR1-FLI1 fusion have reduced TGF-beta sensitivity, and that fusion-positive ES cells and primary tumours both express low or undetectable levels of TGFBR2 mRNA and protein product. Co-transfection of FLI1 and the TGFBR2 promoter induces promoter activity, whereas EWSR1-FLI1 leads to suppression of TGFBR2 promoter activity and FLI1-induced promoter activity. Introduction of EWSR1-FLI1 into cells lacking the EWSR1-FLI1 fusion suppresses TGF-beta RII expression, whereas antisense to EWSR1-FLI1 in ES cell lines positive for this gene fusion restores TGF-beta RII expression. Furthermore, introduction of normal TGF-beta RII into ES cell lines restores TGF-beta sensitivity and blocks tumorigenicity. Our results implicate TGF-beta RII as a direct target of EWS-FLI1.


Asunto(s)
Proteínas de Fusión Oncogénica/fisiología , Proteínas Proto-Oncogénicas , Receptores de Factores de Crecimiento Transformadores beta/genética , Factores de Transcripción/fisiología , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas , Proteína Proto-Oncogénica c-fli-1 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína EWS de Unión a ARN , Receptor Tipo II de Factor de Crecimiento Transformador beta , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Eliminación de Secuencia , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
2.
Neuron ; 11(2): 321-31, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8394722

RESUMEN

Retinoic acid (RA) induces the neuronal differentiation of many human neuroblastoma cell lines. In this study, we show that RA treatment of neuroblastoma cells induces the expression of TrkB, the receptor for the neurotrophins BDNF, NT-3, and NT-4/5. BDNF addition to RA-treated SH-SY5Y neuroblastoma cells stimulated the tyrosine phosphorylation of TrkB and neuronal differentiation. RA treatment of KCNR neuroblastoma cells, which constitutively express BDNF mRNA, resulted in the expression of TrkB and differentiation in the absence of added BDNF. Finally, in 15N neuroblastoma cells, which express BDNF mRNA but do not differentiate in response to RA, RA induced only a truncated form of TrkB. 15N cells transfected with full-length TrkB differentiated in the absence of RA. These results indicate that RA induces the neuronal differentiation of neuroblastoma cells by modulating the expression of neurotrophin receptors.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Neuroblastoma/patología , Proteínas Proto-Oncogénicas/genética , Tretinoina/farmacología , Diferenciación Celular/efectos de los fármacos , Humanos , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/metabolismo , Receptor trkA , Receptores de Superficie Celular/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/patología , Tirosina/metabolismo
3.
Cell Death Differ ; 14(2): 318-26, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16778834

RESUMEN

Chemoresistance and increased expression of TrkB and brain-derived neurotrophic factor (BDNF) are biomarkers of poor prognosis in tumors from patients with neuroblastoma (NB). Previously, we found BDNF activation of TrkB through PI3K/Akt protects NB from etoposide/cisplatin-induced cell death. In this study, the role of Bim, a proapoptotic protein, was investigated. Bim was involved in paclitaxel but not etoposide or cisplatin-induced cell death in NB cells. Pharmacological and genetic studies showed that BDNF-induced decreases in Bim were regulated by MAPK and not PI3K/Akt pathway. Both MAPK and PI3K pathways were involved in BDNF protection of NB cells from paclitaxel-induced cell death, while PI3K predominantly mediated BDNF protection of NB cells from etoposide or cisplatin-induced cell death. These data indicate that different chemotherapeutic drugs induce distinct death pathways and growth factors utilize different signal transduction pathways to modulate the effects of chemotherapy on cells.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Neuroblastoma/enzimología , Neuroblastoma/patología , Proteínas Proto-Oncogénicas/metabolismo , Receptor trkB/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Muerte Celular/efectos de los fármacos , Cisplatino/farmacología , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Proteínas de la Membrana/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Imitación Molecular/efectos de los fármacos , Neuroblastoma/genética , Paclitaxel/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor trkB/genética
4.
J Clin Invest ; 80(3): 804-11, 1987 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2887586

RESUMEN

We have found highly predictable patterns of protooncogene expression in cell lines and tumor tissue of neuroblastoma (NB), a tumor of the peripheral nervous system (PNS). These patterns make it possible to recognize two different genetically definable subgroups among histopathologically indistinguishable tumors. Additionally, we have identified a difference in neurotransmitter biosynthetic enzyme activity in these two subgroups of NB. The patterns of protooncogene expression and neurotransmitter biosynthetic enzymes suggests that these tumors arise in different cells of the PNS.


Asunto(s)
Neuroblastoma/genética , Tumores Neuroectodérmicos Periféricos Primitivos/genética , Oncogenes , Neoplasias del Sistema Nervioso Periférico/genética , Línea Celular , Humanos , Neuroblastoma/enzimología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Tumores Neuroectodérmicos Periféricos Primitivos/enzimología , Tumores Neuroectodérmicos Periféricos Primitivos/metabolismo , Tumores Neuroectodérmicos Periféricos Primitivos/patología , Neurotransmisores/biosíntesis , Neoplasias del Sistema Nervioso Periférico/enzimología , Neoplasias del Sistema Nervioso Periférico/metabolismo , Neoplasias del Sistema Nervioso Periférico/patología
5.
Mol Cell Biol ; 8(4): 1677-83, 1988 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-3380093

RESUMEN

We detected expression of the c-myb proto-oncogene, which was initially thought to be expressed in a tissue-specific manner in cells of hematopoietic lineage, in human tissues of neuronal origin. Since the level of c-myb expression declined during fetal development, we studied the regulation of its expression in human neuroblastoma cell lines induced to differentiate by retinoic acid. The expression of c-myb declined during the maturation of neuroblastoma cells, and this change was mediated by a decrease in c-myb transcription.


Asunto(s)
Regulación de la Expresión Génica , Proto-Oncogenes/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Humanos , Neuroblastoma , Proto-Oncogenes Mas , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación
6.
Oncogene ; 36(1): 97-109, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27270431

RESUMEN

As a transcription factor, localization to the nucleus and the recruitment of cofactors to regulate gene transcription is essential. Nuclear localization and nucleosome remodeling and histone deacetylase (NuRD) complex binding are required for the zinc-finger transcription factor CASZ1 to function as a neuroblastoma (NB) tumor suppressor. However, the critical amino acids (AAs) that are required for CASZ1 interaction with NuRD complex and the regulation of CASZ1 subcellular localization have not been characterized. Through alanine scanning, immunofluorescence cell staining and co-immunoprecipitation, we define a critical region at the CASZ1 N terminus (AAs 23-40) that mediates the CASZ1b nuclear localization and NuRD interaction. Furthermore, we identified a nuclear export signal (NES) at the N terminus (AAs 176-192) that contributes to CASZ1 nuclear-cytoplasmic shuttling in a chromosomal maintenance 1-dependent manner. An analysis of CASZ1 protein expression in a primary NB tissue microarray shows that high nuclear CASZ1 staining is detected in tumor samples from NB patients with good prognosis. In contrast, cytoplasmic-restricted CASZ1 staining or low nuclear CASZ1 staining is found in tumor samples from patients with poor prognosis. These findings provide insight into mechanisms by which CASZ1 regulates transcription, and suggests that regulation of CASZ1 subcellular localization may impact its function in normal development and pathologic conditions such as NB tumorigenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neuroblastoma/metabolismo , Señales de Exportación Nuclear , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Expresión Génica , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Mutación , Neuroblastoma/genética , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Transcripción Genética , Transcriptoma , Proteínas Supresoras de Tumor/química
7.
Cancer Res ; 52(16): 4402-7, 1992 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-1322787

RESUMEN

In this report we provide evidence for the activation of distinct differentiation pathways during treatment of the neuroblastoma cell line SMS-KCNR with 1 mM dibutyryl cyclic AMP (dbcAMP) and/or 5 microM retinoic acid (RA). Our results show that the adrenal gland specific gene pG2 is induced only during dbcAMP treatment, while RA induces a neuronal phenotype and expression of all neural related genes while decreasing the expression of many chromaffin related genes. Furthermore dbcAMP does not affect the DNA content distribution of SMS-KCNR [G1 = 61.8 +/- 4.1% (SD); S = 20.3 +/- 6.3%; G2-M = 18 +/- 5.4%] despite morphological and molecular signs of cellular differentiation. Conversely, RA arrests cell growth causing a decrease in cells in the growth fraction (S + G2 + M = 15.6 +/- 6.1%) and an increase in cells in G1 (G1 = 84.3 +/- 5%). Using cyclic AMP and RA in combination, we found that RA inhibited expression of adrenal gland specific gene pG2 and induced a neuronal phenotype. Since dbcAMP does not cause a significant G1 block in SMS-KCNR cells we propose that this agent may be able to induce SMS-KCNR only to an intermediate stage of chromaffin differentiation in which cells retain their proliferative potential.


Asunto(s)
Diferenciación Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes de Retinoblastoma/efectos de los fármacos , Neuroblastoma/genética , Bucladesina/farmacología , Diferenciación Celular/efectos de los fármacos , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Genes de Retinoblastoma/genética , Marcadores Genéticos , Humanos , Neuroblastoma/patología , Fenotipo , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Fase S/efectos de los fármacos , Factores de Tiempo , Transcripción Genética , Tretinoina/farmacología , Células Tumorales Cultivadas , Péptido Intestinal Vasoactivo/farmacología
8.
Cancer Res ; 55(8): 1798-806, 1995 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-7712490

RESUMEN

A large number of poor prognosis neuroblastoma (NB) tumors constitutively express brain-derived neurotrophic factor (BDNF) and variably express the gene for its tyrosine kinase (Trk) receptor TrkB. Good prognosis NB tumors typically express high levels of TrkA mRNA, which encodes the signal transducing receptor for nerve growth factor, p140TrkA. These neurotrophins are necessary for neural cell survival and differentiation. This study evaluates the effects of activation of the BDNF-TrkB signal transduction pathway on the growth, survival, morphology, and invasive capacity of NB cells. We find that the addition of BDNF to SY5Y cells induced to express p145TrkB by retinoic acid treatment does not significantly affect cell proliferation yet will support cell survival. Activation of the BDNF-TrkB signal transduction pathway stimulates disaggregation of cells and extension of neuritic processes which can be blocked by a BDNF-neutralizing antibody. Treatment of cells with K252a, an inhibitor of Trk, reverses the cellular disaggregation. An evaluation of the effects of BDNF and nerve growth factor on the ability of NB cells to penetrate basement membrane proteins indicated that BDNF stimulated a 2-fold increase while nerve growth factor inhibited RA-SY5Y cell invasion. Thus, activation of the p145TrkB signal transduction pathway stimulates NB cell survival, disaggregation, and invasion; all characteristics of metastatic cells. Furthermore, these studies indicate that activation of different Trk signal transduction pathways in NB cells results in distinct differences in tumor cell biology and these may be relevant to the clinical course of the patients.


Asunto(s)
Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Neuroblastoma/metabolismo , Neuroblastoma/patología , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Factor Neurotrófico Derivado del Encéfalo , Diferenciación Celular , Línea Celular , Supervivencia Celular , ADN de Neoplasias/biosíntesis , Humanos , Cinética , Invasividad Neoplásica , Factores de Crecimiento Nervioso/biosíntesis , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Receptor de Factor Neurotrófico Ciliar , Timidina/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas
9.
Cancer Res ; 46(8): 4139-42, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3731081

RESUMEN

Many human neuroblastoma tumors and cells in culture contain amplified N-myc DNA and this is associated with tumor stage. We have analyzed pairs of neuroblastoma cell cultures derived from two patients at the time of diagnosis and after tumor progression following the initiation of therapy. Cell cultures derived after progression have increased expression of N-myc RNA. In one pair this increase is associated with increased N-myc DNA amplification; in the other, amplification decreases and activation of N-myc is most likely the result of a regulatory change. Analysis of the pattern of DNA amplification in these cell cultures demonstrates additional changes that might be associated with tumor progression.


Asunto(s)
Neuroblastoma/genética , Proto-Oncogenes , Secuencia de Bases , Línea Celular , ADN de Neoplasias/análisis , Amplificación de Genes , Humanos , Neuroblastoma/patología , ARN Mensajero/análisis
10.
Cancer Res ; 48(15): 4307-11, 1988 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-3390826

RESUMEN

Genetic characterization of human tumors promises new insights of biological importance and clinical relevance. We have found that two solid tumors, peripheral neuroepithelioma and Ewing's sarcoma of bone, which share a common cytogenetic rearrangement, are characterized by an indistinguishable and highly reproducible pattern of protooncogene expression. c-myc, N-myc, c-myb, and c-mil/raf-1 are all expressed at similar levels in these tumors. c-fes and c-sis expression was not detected in any specimens of either tumor. In contrast, the protooncogene c-ets-1, located near the breakpoint of the chromosomal translocation in these tumors, is variable in its expression. We also detected high levels of choline acetyltransferase in these tumors, which suggests a common neural origin. Since it is likely that the clinical behavior and therapeutic responsiveness of tumors relate closely to their biological and genetic features, the pattern of protooncogene expression of individual tumors may provide a novel basis for their characterization.


Asunto(s)
Regulación de la Expresión Génica , Tumores Neuroectodérmicos Periféricos Primitivos/genética , Proto-Oncogenes , Sarcoma de Ewing/genética , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Colina O-Acetiltransferasa/análisis , Humanos , Tumores Neuroectodérmicos Periféricos Primitivos/enzimología , Sarcoma de Ewing/enzimología , Translocación Genética
11.
Cancer Res ; 55(11): 2380-6, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7757990

RESUMEN

Recent work on a variety of normal and malignant cell lines has shown that induction and secretion of biologically active TGF-beta may occur after exposure to all-trans-retinoic acid (RA), coincident with decreased growth rate and/or differentiation. This study evaluates the expression and regulation of transforming growth factor beta (TGF-beta) and its receptors during RA-induced cell growth arrest and induction of differentiation in the RA-sensitive human neuroblastoma cell line SMS-KCNR and the RA-resistant neuroblastoma cell line SK-N-AS. RA treatment of SMS-KCNR cells results in a 40-fold increase in TGF-beta 1 mRNA after 4 days of RA, a dose-dependent increase in TGF-beta 1 secretion, an increase in types I (TBRI) and III (TBRIII) TGF-beta receptor proteins, and an increase in type II TGF-beta receptor (TBRII) mRNA coincident with RA-responsiveness of the cells. However, in the RA-resistant line SK-N-AS, TGF-beta 1 is constitutively secreted at levels that are unchanged after RA treatment, and although TBRI and TBRIII mRNA is expressed in untreated SK-N-AS cells, levels of TBRI and TBRIII protein and TBRII mRNA decrease after RA treatment. Thus, in RA-sensitive neuroblastoma cells, RA treatment may result in the induction of a negative autocrine TGF-beta 1 growth regulatory loop. These results suggest the hypothesis that: (a) induction of a TGF-beta 1 negative autocrine growth loop may be a necessary component for RA-responsiveness of neuroblastoma cells in vivo; and (b) the inability to induce or maintain this TGF-beta 1 negative autocrine growth loop may be a mechanism of RA resistance in neuroblastoma.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Tretinoina/farmacología , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Estudios de Evaluación como Asunto , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Neuroblastoma/patología , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
12.
Cancer Res ; 56(16): 3737-42, 1996 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8706017

RESUMEN

Brain-derived neurotrophic factor (BDNF) and its receptors are necessary for the survival and development of many neuronal cells. Because BDNF and TrkB are expressed in many poor-prognosis neuroblastoma (NB) tumors, we evaluated the role of BDNF in affecting sensitivity to chemotherapeutic agents. We investigated the effects of activation of the BDNF-TrkB signal transduction pathway in two NB cell lines, 15N and SY5Y. 15N cells lack the high-affinity receptor p145TrkB and express BDNF; 15N cells were used along with 15N-TrkB cells, a subline transfected with a TrkB expression vector. In cytotoxicity assays, 15N-TrkB cells were consistently 1.4-2 fold more resistant to vinblastine than 15N cells. Drug accumulation assays showed a 50% reduction in[3H]vinblastine accumulation in 15N-TrkB cells compared with control 15N cells. Addition of 30 ng/ml BDNF resulted in a reduction to 46% of control in 15N cells and a reduction to 28% of control in 15N-TrkB cells. SY5Y cells were chosen as a second model because they lack both endogenous BDNF and TrkB expression. p145TrkB expression is induced by 1 nM retinoic acid. Vinblastine accumulation was not significantly affected by 1 nM retinoic acid in SY5Y cells. Addition of 30 ng/ml BDNF decreased [3H]vinblastine accumulation to 58% of control in SY5Y cells and decreased [3H]vinblastine accumulation to 62% of control in TrkB-expressing SY5Y cells. Although an increase in BDNF expression in seen in multidrug-resistant sublines of SY5Y and BE(2)-C NB cells, the protective effect of BDNF in vinblastine toxicity may be unrelated to mdr-1, because the activity of other agents transported by P-glycoprotein was not affected. There was no increase in mdr-1 expression in 1 nM RA SY5Y cells and 15N-TrkB cells, as assessed by Northern blot analysis. In addition to the effects of BDNF on vinblastine cytotoxicity and accumulation, there was an inhibition in the ability of vinblastine to depolymerize tubulin in BDNF-treated cells. Thus, BDNF and TrkB may partially rescue NB cells from vinblastine toxicity and thereby may contribute to a more chemoresistant phenotype.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas del Tejido Nervioso/farmacología , Neuroblastoma/tratamiento farmacológico , Vinblastina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Factor Neurotrófico Derivado del Encéfalo , Resistencia a Medicamentos , Humanos , Proteínas del Tejido Nervioso/genética , ARN Mensajero/análisis , Tretinoina/farmacología , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas , Vinblastina/farmacocinética
13.
Cancer Res ; 50(18): 6055-61, 1990 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-2393870

RESUMEN

Neuropeptide Y (NPY) expression is limited to tissues of the central and peripheral nervous system. In the adrenal gland, NPY is found in a subset of cells of the adrenal medulla. Using in situ hybridization analysis, NPY mRNA expression was characterized during human fetal adrenal medullary development. We found a biphasic pattern of NPY mRNA expression during the development of the human adrenal medulla. NPY mRNA is detectable at the earliest evaluable time point (7.5 weeks of gestational age) through 18 weeks of gestational age, and is then not detectable until 8 months after birth. We also analyzed NPY mRNA expression in neuroblastoma tumors, which often arise in the adrenal medulla. Thirty-eight neuroblastoma tumors were analyzed for NPY mRNA expression using in situ hybridization. We found NPY mRNA expression in 30 of 38 tumors; 15 of 15 Stage IVS tumors from children under 1 year of age at diagnosis expressed NPY mRNA, whereas 0 of 4 Stage IV tumors from children less than 1 year of age at diagnosis expressed NPY mRNA. These data suggest that in children under 1 year of age at diagnosis, Stage IVS and Stage IV neuroblastoma may be marked by the presence or absence, respectively, of NPY mRNA expression. Moreover, since NPY is expressed for only a short period of time during embryogenesis, these tumors may arise from different neuroblast populations occurring during the course of adrenal medullary development.


Asunto(s)
Glándulas Suprarrenales/análisis , Neuroblastoma/análisis , Neuropéptido Y/genética , Glándulas Suprarrenales/embriología , Femenino , Expresión Génica , Humanos , Estadificación de Neoplasias , Neuropéptido Y/análisis , Embarazo , ARN Mensajero/análisis
14.
Cancer Res ; 54(1): 124-33, 1994 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-8261433

RESUMEN

To investigate the presence of biologically active somatostatin (SS) receptors in neural crest-derived tumors, radioligand binding studies, cyclic AMP accumulation, intracellular calcium, and growth assays were performed in eight human neuroblastoma (NB) cell lines. Mathematical modeling of binding experiments strongly indicates the presence of heterogeneity of sites. The first site (SSR1) is present in 40% of the NB cell lines and binds with low capacity (0.5 pmol/mg protein) and high affinity (0.1-1 nM) SS14, SS28, and analogues. The second site (SSR2) is a high capacity site (200 pmol/mg protein), widely distributed in all of the cell lines investigated, that shows relative selectivity yet low affinity (100 nM) for SS14, SS28, and [D-Trp8]SS14 without any apparent biological activity. SSR1 is coupled to a pertussis toxin-sensitive G protein, inhibits forskolin- or VIP-stimulated adenylate cyclase activity, decreases intracellular free calcium, and mediates inhibition (30%) of both DNA synthesis and cell growth. Analysis of cell cycle distribution in aphidicolin-synchronized SSR1-positive NB cells indicated that this inhibitory effect is partially mediated by a transient accumulation in G0-G1. Our data indicate high affinity binding sites for SS14, and analogues are present and biologically active in a subset of NB cells.


Asunto(s)
Neuroblastoma/metabolismo , Receptores de Somatostatina/metabolismo , Sitios de Unión , Calcio/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Colforsina/farmacología , Simulación por Computador , AMP Cíclico/metabolismo , Humanos , Neuroblastoma/química , Neuroblastoma/patología , Octreótido/farmacología , Receptores de Somatostatina/análisis , Receptores de Somatostatina/fisiología , Células Tumorales Cultivadas , Péptido Intestinal Vasoactivo/farmacología
15.
Oncogene ; 16(25): 3337-43, 1998 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-9681834

RESUMEN

Retinoic acid (RA) treatment of SMS-KCNR neuroblastoma (NB) cells leads to G1 growth arrest and neuronal differentiation. To investigate the molecular mechanisms by which RA alters cell growth, we analysed the expression and activity of components of the cell cycle machinery after culture in RA. Within 2 days of RA treatment and prior to the arrest of NB cells in the G1 phase of the cell cycle, there is a complete downregulation of G1 cyclin/Cdk activities. Protein levels for the G1 cyclin/Cdks were essentially unchanged during this time although there was a decrease in the steady-state levels of p67N-Myc and hyperphosphorylated Rb proteins. The Cdk inhibitors, p21Cip1 and p27Kip1 were constitutively expressed in KCNR while p15INK4B and p16INK4A were not detected. RA induced an increase in the expression of p27Kip1 but not p21Cip1. Furthermore, coincident with the decrease in kinase activity there was an increase in G1 cyclin/Cdk bound p27Kip1. These results indicate that changes in the level of p27Kip1 and its binding to G1 cyclin/Cdks may play a key role in RA induced growth arrest of NB cells.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Asociadas a Microtúbulos , Proteínas Supresoras de Tumor , Antineoplásicos/farmacología , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Fase G1/efectos de los fármacos , Fase G1/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Genes Supresores de Tumor , Humanos , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Neuroblastoma/genética , Neuroblastoma/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Tretinoina/farmacología , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
16.
Oncogene ; 6(11): 2149-54, 1991 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1945417

RESUMEN

The genes of the ets family are thought to code for a novel class of transcriptional factors. These proteins have a specific DNA-binding domain different from the basic domain of both the helix-loop-helix and leucine zipper families of DNA-binding proteins. The ets-1 gene product has been shown to bind to the enhancer region of the human T-cell receptor alpha gene during thymocyte ontogeny. This finding explains the high expression of ets-1 observed in T cells and the correlation between ets-1 expression and the expression of the T-cell receptor gene during fetal development. The ets-1 gene is also possibly biologically active in neural cells. By using RNA in situ hybridization analysis, we demonstrate the presence of ets-1 transcripts in cells of peripheral embryonal neuroectodermal tumors, specifically neuroepithelioma and neuroblastoma. In addition, the gene is found transcribed in Ewing's sarcoma, postulated to be ontogenetically related to tumors derived from the neural crest.


Asunto(s)
Tejido Nervioso/metabolismo , Neoplasias del Sistema Nervioso Periférico/metabolismo , Proteínas Oncogénicas de Retroviridae/biosíntesis , Expresión Génica , Humanos , Linfocitos/metabolismo , Microscopía Fluorescente , Neuroblastoma/metabolismo , Tumores Neuroectodérmicos Periféricos Primitivos/metabolismo , Hibridación de Ácido Nucleico , ARN Mensajero/análisis , Sarcoma de Ewing/metabolismo
17.
Oncogene ; 3(3): 281-8, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3060792

RESUMEN

Human neuroblastoma (NB) is a highly malignant tumor arising in cells that originate in the embryonal neural crest. Several lines of investigation suggest that both NB and other tumors of developing tissues are blocked in their ability to differentiate and achieve growth arrest. Since in vivo differentiation of NB has been frequently observed and may be of clinical importance (Fox et al., 1959; Evans et al., 1976), we have utilized the in vitro induction of NB differentiation by retinoic acid (RA) to study the molecular events associated with NB differentiation. We have focused our studies on changes that occur in the expression of various proto-oncogenes during NB tumors cell differentiation because proto-oncogenes are likely to be of central importance in mediating processes critical for cellular growth and maturation. In these studies, we have found that the expression of no fewer than five proto-oncogenes including c-Ha-ras, c-ets-1, and c-fos change during the differentiation of NB cells, while the expression of c-erb-B changes in association with the arrest of growth that occurs during NB differentiation. In some cases the altered expression of a proto-oncogene was transcriptionally regulated, while in others post-transcriptional mechanisms were important.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Neuroblastoma/patología , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Tretinoina/farmacología , Humanos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/biosíntesis , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/patología
18.
Cell Death Differ ; 1(1): 49-58, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17180006

RESUMEN

Retinoic Acid (RA) has been shown to control growth and induce differentiation in a number of human neuroblastoma (NB) cell lines. However, a number of NB cell lines may be termed resistant to RA as they fail to growth arrest and differentiate. In studying the mechanism mediating RA-resistance, we noted that invariably RA-resistant NB cell lines constitutively express Insulin-like Growth Factor 2 (IGF2) (Gaetano, 1991b). The NB cell line LAN-1-15N (15N) represented an interesting model in which to study the development of RA-resistance as initially 15N cells are growth arrested by RA, however with prolonged culture (8-10 days) cells begin to proliferate. Coincidentally, RA induces IGF2 mRNA and protein secretion in 15N NB cells (Matsumoto, 1992). In this study we isolated RA-resistant 15N cell lines and analyzed their growth properties and changes in cell cycle related (cdc2, cdk2, cyclins A, B, D and E) and early response (fos and jun) gene expression to evaluate the role IGF2 may play in mediating RA resistance. We found that exogenous IGF2 stimulates growth in 15N and is capable of altering RA induced inhibition of NB cell growth. Finally we show that by blocking the Insulin-like Growth Factor Receptor (IGF1(R)) with a monoclonal antibody (alpha-IR3) in the presence of RA the growth of RAR cell lines could be completely blocked. These data are consistent with the concept that signals by IGF2 and transduced via the IGF1(R) can mediate resistance to the growth inhibiting properties of RA.

19.
Cell Death Differ ; 10(2): 230-9, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12700651

RESUMEN

Poor prognosis neuroblastoma (NB) tumors are marked by amplification and overexpression of N-myc. Retinoic acid (RA) decreases N-myc levels and induces cell cycle arrest in vitro and increases event-free survival in advanced stage NB patients. In this study, we investigated the mechanism(s) by which RA regulates cell cycle and how N-myc affects NB cell cycle progression. Constitutive N-myc overexpression stimulates increases in cyclin E-dependent kinase activity and decreases in p27 resulting in increased DNA synthesis. N-myc regulates p27 levels through an increase in targeting of p27 to the proteasome via cyclin E kinase-dependent phosphorylation of p27 and its ubiquitination. N-myc also stimulates an increase in proteasome activity. In RA-treated cells in which N-myc levels decline as p27 levels increase, degradation of p27 is also decreased. However, RA does not affect the activity of proteasome. The decrease in the degradation of p27 in RA-treated cells is due in part to a decrease in the N-myc stimulated phosphorylation of p27. However, RA also decreases Skp2 levels thus impairing the ability of p27 to be ubiquitinated. Thus, RA induces both N-myc-dependent and -independent mechanisms to minimize the degradation of p27 and arrest NB cell growth.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Tretinoina/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina E , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Cisteína Endopeptidasas/metabolismo , Humanos , Complejos Multienzimáticos/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Fosforilación , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Quinasas Asociadas a Fase-S/efectos de los fármacos , Ubiquitinas/metabolismo
20.
Endocrinology ; 130(6): 3669-76, 1992 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1375906

RESUMEN

Insulin-like growth factors (IGF-I and IGF-II) are mitogenic polypeptides that play an important role in normal growth and development. IGF-II has been shown to stimulate the growth of neuroblastoma tumors in an autocrine and paracrine fashion. Critical in determining the role of IGF-II in tumorigenesis is the necessity to delineate factors affecting the transcription of IGF-II in normal and tumor tissues. To date such factors are poorly characterized. In this study we find that retinoic acid (RA), a naturally occurring morphogen, that has been shown to be indispensable in the development of the chick limb bud, stimulates an increase in IGF-II messenger RNA (mRNA) in the Lan-1-15N neuroblastoma cell line. This increase in IGF-II is coincident with RA mediated inhibition of DNA synthesis. An increase in the steady state levels of IGF-II mRNA is detectable within 2 h of RA treatment and maximal by 24 h. In RA-treated Lan-1-15N cells, IGF-II mRNA levels are regulated at the level of new gene transcription and result in an increase in IGF-II protein in the culture supernatant. These studies suggest one mechanism affecting the production of IGF-II in vivo may be mediated by RA and detail a model system by which transcriptional regulation of IGF-II mRNA can be analyzed.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/genética , ARN Mensajero/metabolismo , Tretinoina/farmacología , División Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/fisiología , Replicación del ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Neuroblastoma , Poli A/genética , Poli A/aislamiento & purificación , Poli A/metabolismo , ARN/genética , ARN/aislamiento & purificación , ARN Mensajero/genética , Timidina/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA