Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Oncogene ; 36(1): 97-109, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27270431

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neuroblastoma/metabolismo , Sinais de Exportação Nuclear , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Espaço Intracelular/metabolismo , Mutação , Neuroblastoma/genética , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica , Transcriptoma , Proteínas Supressoras de Tumor/química
2.
Cell Death Differ ; 18(7): 1174-83, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21252912

RESUMO

Neuroblastoma (NB) is a common childhood malignant tumor of the neural crest-derived sympathetic nervous system. In NB the frequent loss of heterozygosity (LOH) on chromosome 1p raises the possibility that this region contains tumor-suppressor genes whose inactivation contributes to tumorigenesis. The human homolog of the Drosophila neural fate determination gene CASZ1, a zinc-finger transcription factor, maps to chromosome 1p36.22, a region implicated in NB tumorigenesis. Quantitative real-time PCR analysis showed that low-CASZ1 expression is significantly correlated with increased age (≥18 months), Children's Oncology Group high-risk classification, 1p LOH and MYCN amplification (all P<0.0002) and decreased survival probability (P=0.0009). CASZ1 was more highly expressed in NB with a differentiated histopathology (P<0.0001). Retinoids and epigenetic modification agents associated with regulation of differentiation induced CASZ1 expression. Expression profiling analysis revealed that CASZ1 regulates the expression of genes involved in regulation of cell growth and developmental processes. Specific restoration of CASZ1 in NB cells induced cell differentiation, enhanced cell adhesion, inhibited migration and suppressed tumorigenicity. These data are consistent with CASZ1 being a critical modulator of neural cell development, and that somatically acquired disruption of normal CASZ1 expression contributes to the malignant phenotype of human NB.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes Supressores de Tumor , Neuroblastoma/genética , Fatores de Transcrição/metabolismo , Adesão Celular , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Reprogramação Celular , Cromossomos Humanos Par 1 , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Estimativa de Kaplan-Meier , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Retinoides/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
3.
Oncogene ; 27(39): 5204-13, 2008 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-18504438

RESUMO

Loss of 1p36 heterozygosity commonly occurs with MYCN amplification in neuroblastoma tumors, and both are associated with an aggressive phenotype. Database searches identified five microRNAs that map to the commonly deleted region of 1p36 and we hypothesized that the loss of one or more of these microRNAs contributes to the malignant phenotype of MYCN-amplified tumors. By bioinformatic analysis, we identified that three out of the five microRNAs target MYCN and of these miR-34a caused the most significant suppression of cell growth through increased apoptosis and decreased DNA synthesis in neuroblastoma cell lines with MYCN amplification. Quantitative RT-PCR showed that neuroblastoma tumors with 1p36 loss expressed lower level of miR-34a than those with normal copies of 1p36. Furthermore, we demonstrated that MYCN is a direct target of miR-34a. Finally, using a series of mRNA expression profiling experiments, we identified other potential direct targets of miR-34a, and pathway analysis demonstrated that miR-34a suppresses cell-cycle genes and induces several neural-related genes. This study demonstrates one important regulatory role of miR-34a in cell growth and MYCN suppression in neuroblastoma.


Assuntos
MicroRNAs/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Sequência de Bases , Deleção Cromossômica , Cromossomos Humanos Par 1 , Primers do DNA , Humanos , Perda de Heterozigosidade , Mutagênese Sítio-Dirigida , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/genética , Neuroblastoma/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Cell Death Differ ; 14(2): 318-26, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16778834

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Regulação para Baixo/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Proteínas Proto-Oncogênicas/metabolismo , Receptor trkB/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Morte Celular/efeitos dos fármacos , Cisplatino/farmacologia , Ativação Enzimática/efeitos dos fármacos , Etoposídeo/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Proteínas de Membrana/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mimetismo Molecular/efeitos dos fármacos , Neuroblastoma/genética , Paclitaxel/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor trkB/genética
5.
Br J Cancer ; 95(7): 879-88, 2006 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-16969347

RESUMO

Neuroblastoma (NB) is the most common extracranial solid tumour in infants. Unfortunately, most children present with advanced disease and have a poor prognosis. In the present study, we evaluated the role of the peroxisome proliferator-activated receptor gamma (PPARgamma) agonist rosiglitazone (RGZ) in two NB cell lines (SK-N-AS and SH-SY5Y), which express PPARgamma. Rosiglitazone decreased cell proliferation and viability to a greater extent in SK-N-AS than in SH-SY5Y. Furthermore, 20 microM RGZ significantly inhibited cell adhesion, invasiveness and apoptosis in SK-N-AS, but not in SH-SY5Y. Because of the different response of SK-N-AS and SH-SY5Y cells to RGZ, the function of PPARgamma as a transcriptional activator was assessed. Noticeably, transient transcription experiments with a PPARgamma responsive element showed that RGZ induced a three-fold increase of the reporter activity in SK-N-AS, whereas no effect was observed in SH-SY5Y. The different PPARgamma activity may be likely due to the markedly lower amount of phopshorylated (i.e. inactive) protein observed in SK-N-AS. To our knowledge, this is the first demonstration that the differential response of NB cells to RGZ may be related to differences in PPARgamma transactivation. This finding indicates that PPARgamma activity may be useful to select those patients, for whom PPARgamma agonists may have a beneficial therapeutic effect.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3 , Caspases/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Metaloproteinase 9 da Matriz/metabolismo , PPAR gama/genética , Rosiglitazona , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Ativação Transcricional , Transfecção
6.
Neuroscience ; 126(1): 149-62, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15145081

RESUMO

Developmental exposure to ethanol causes profound damage to the cerebellum, ranging from aberration in neuronal differentiation to cell loss. As a major neurotrophic factor, brain-derived neurotrophic factor (BDNF) and its receptor TrkB are expressed in the developing, as well as adult, cerebellum. Many neurotrophic effects of BDNF are mediated by gene transcription. We hypothesized that ethanol interfered with BDNF signaling and disrupted BDNF-regulated transcriptional activity. Using a transgenic mouse model expressing an activator protein-1 (AP-1) luciferase reporter construct, we demonstrated that BDNF stimulated AP-1 transactivation in cultured cerebellar granule neurons. This observation was validated by the study using a human neuronal cell line expressing inducible TrkB (TB8 neuroblastoma cells). BDNF induced AP-1 transactivation, as well as increased the binding activity of AP-1 protein complex to a DNA sequence containing AP-1 sites in TB8 cells. BDNF-mediated AP-1 activation was mediated by PI3K/Akt and JNK pathways; BDNF activated Akt and JNKs, and blocking these pathways significantly inhibited BDNF-stimulated AP-1 transactivation. More importantly, ethanol inhibited BDNF-mediated activation of PI3K/Akt and JNKs, and blocked BDNF-stimulated AP-1 activation. Since ethanol did not affect either the expression or autophosphorylation of TrkB, it could be concluded that the site of ethanol action was downstream of TrkB. The present study establishes that this AP-1 reporter transgenic mouse model is valuable for assessing AP-1 activity in the CNS neurons. Our results provide an insight into molecular mechanism(s) of ethanol action.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Cerebelo/citologia , Etanol/farmacologia , Neurônios/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Animais , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Interações Medicamentosas , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuroblastoma , Neurônios/citologia , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Receptor trkB/metabolismo
7.
Cell Death Differ ; 10(2): 230-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12700651

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Quinases Associadas a Fase S/metabolismo , Tretinoína/farmacologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina E , Quinase 6 Dependente de Ciclina , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Cisteína Endopeptidases/metabolismo , Humanos , Complexos Multienzimáticos/metabolismo , Neuroblastoma/genética , Neuroblastoma/patologia , Fosforilação , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Quinases Associadas a Fase S/efeitos dos fármacos , Ubiquitinas/metabolismo
8.
Med Pediatr Oncol ; 36(1): 97-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11464914

RESUMO

BACKGROUND AND PROCEDURE: To investigate the molecular mechanisms by which retinoic acid (RA) alters cell growth, the expression and activity of components of the cell cycle machinery were analyzed. RESULTS AND CONCLUSIONS: Within 2 days of RA treatment, and prior to the arrest of NB cells in the G1 phase of the cell cycle, there was a complete downregulation of GI cyclin/cdk activities. Protein levels for the G1 cyclin/cdk were essentially unchanged during this time, although there was a decrease in the steady state levels of hyperphosphorylated Rb and p60N-MYC proteins. The cdk inhibitors, p21Cip1 and p27Kip1 were constitutively expressed in KCNR, while p15 INK4B and p16 INK4A mRNA were undetected. Within 24 hr of RA treatment, there was a 4-fold increase in the expression of p27Kip1, although p27 mRNA levels were unchanged. Levels of p21Cip1 were unaltered. Coincident with the decrease in kinase activity there was an increase in p27 bound to G1 cyclin/cdk. The increase in p27 was not due to an increase in transcription. In other cell systems, increased expression of c-MYC has been shown to lead to a decrease in p27 levels that is regulated at the post-transcriptional level (sequestration). To determine whether increased levels of N-MYC could affect the level of p27, we evaluated the expression of p27 in a series of N-MYC transfected cells and found that constitutive overexpression of N-MYC led to a decrease in the steady-state levels of p27 and in p27 bound to G1 cyclin/cdk complexes. Using adenoviral vectors expressing p27, we found that infection leads to increased p27 expression, which causes a decrease in cdk activity and an accumulation of cells in G1.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Neuroblastoma/genética , Tretinoína/farmacologia , Proteínas Supressoras de Tumor , Adenoviridae/genética , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Fase G1/efeitos dos fármacos , Genes do Retinoblastoma , Genes myc , Vetores Genéticos/genética , Humanos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neuroblastoma/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Proteína do Retinoblastoma/metabolismo , Transfecção , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
9.
J Biol Chem ; 275(22): 16560-8, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10748015

RESUMO

The human unc-33-like phosphoprotein (hUlip/CRMP-4) is a member of a family of developmentally regulated genes that are highly expressed in the nervous system. Mutations in the C. elegans unc-33 gene lead to worms with abnormal movements. The hUlip gene encodes a 570-amino acid protein with 98% homology to its murine (Ulip) (Byk, T., Dobransky, T., Cifuentes-Diaz, C., and Sobel, A. (1996) J. Neurosci. 16, 688-701) and rat (CRMP-4) (Wang, L. H., and Strittmatter, S. M. (1996) J. Neurosci. 16, 6197-6207) counterparts (Gaetano, C., Matsuo, T., and Thiele, C. J. (1997) J. Biol. Chem. 272, 12195-12201). The hUlip gene was isolated from a human genomic library. It contains 15 exons, including an exon defined by an anaplastic oligodendroglioma expressed sequence tag, and spans at least 61.7 kilobases. hUlip lacks sequences corresponding to the first six exons found in unc-33. unc-33 exons correspond to homologous hUlip exons as follows: VII to 1 and 2, VIII to 3-9, IX to 10-12, and X to 13 and 14. Using the hUlip clone 1 phage, fluorescence in situ hybridization analysis indicates that the hybridization signal localizes to human chromosome 5q32. Deletion analysis of 5'-flanking sequences delineated the sequences sufficient to express a reporter gene in both neuroblastoma cells and myoblasts. A consensus MyoD/myogenin binding site is located in a region of the downstream promoter that is nearly identical to its mouse homologue. Mutagenesis shows that this conserved MyoD/myogenin site is necessary for full promoter activity in both myoblasts and neuroblastoma cells.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Helminto/química , Proteínas de Helminto/genética , Proteínas Musculares , Músculos/metabolismo , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/genética , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Evolução Biológica , Mapeamento Cromossômico , DNA Complementar , Éxons , Humanos , Íntrons , Camundongos , Dados de Sequência Molecular , Neuroblastoma/genética , Neuroblastoma/patologia , Ratos
11.
Am J Pathol ; 155(5): 1661-70, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10550322

RESUMO

The types of neurotrophin receptors that are expressed in neuroblastomas have different prognostic implications; trkA is a marker of good prognosis, whereas trkB expression is associated with poor prognosis. This suggests that either the signaling that is mediated via these receptors modulates the biological features of neuroblastoma cells differently, or that distinct lineages of sympathoadrenal precursors have been transformed. In this report, we evaluate the biological effects after activation of trkA or trkB by their major ligands in SH-SY5Y human neuroblastoma cells. Both trkA and trkB induce differentiation, inhibit growth, and promote the survival of cells under conditions of nutrient deprivation. However, the up-regulation of insulin-like growth factor-II (IGF-II) expression is a predominant feature of trkA activation by nerve growth factor (NGF). The growth inhibition induced by blocking the insulin-like growth factor-I receptor suggests that IGF-II is a component of the effector mechanism of trkA activation by NGF in trkA-transfected cells. Although trkA and trkB expression is associated with different prognoses in neuroblastoma, our study indicates that the effects mediated by these receptors in vivo may be quite similar for certain subsets of neuroblastomas.


Assuntos
Biomarcadores Tumorais , Neoplasias Encefálicas/metabolismo , Fator de Crescimento Insulin-Like II/biossíntese , Neuroblastoma/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Neoplasias Encefálicas/patologia , Técnicas de Transferência de Genes , Humanos , Neuroblastoma/patologia , Células Tumorais Cultivadas , Regulação para Cima
12.
Nat Genet ; 23(2): 222-7, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508522

RESUMO

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.


Assuntos
Proteínas de Fusão Oncogênica/fisiologia , Proteínas Proto-Oncogênicas , Receptores de Fatores de Crescimento Transformadores beta/genética , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Nus , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas de Fusão Oncogênica/genética , Regiões Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinases , Proteína Proto-Oncogênica c-fli-1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína EWS de Ligação a RNA , Receptor do Fator de Crescimento Transformador beta Tipo II , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Deleção de Sequência , Transativadores/genética , Fatores de Transcrição/genética , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
13.
Lab Invest ; 79(8): 1007-13, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10462038

RESUMO

Expression level of trkA tyrosine kinase receptor for nerve growth factor is a major prognostic determinant of neuroblastoma, suggesting that defective trkA-mediated signaling is responsible for the tumorigenesis of this childhood malignancy. We investigated the biologic effect of trkA, with special reference to its effect on insulin-like growth factor-II (IGF-II) expression, in SK-N-AS human neuroblastoma cells transfected with human trkA cDNA. Nerve growth factor treatment of trkA-transfected cells promoted growth and changed the morphologic phenotype into a substrate-adherent, flatter phenotype (S-type), and down-regulated the mRNA expression of IGF-II. The effects on both growth and the morphologic differentiation of SK-N-AS cells differed significantly from those of previous studies, and implied that trkA effects can be diverse, depending on the phenotype of the individual neuroblastoma cells. Immunohistochemical screening of trkA and IGF-II expression in adrenal neuroblastomas (n = 25) also favored the nonoverlapping pattern of trkA and IGF-II expression (p < 0.05). Because IGF-II is believed to play a significant role in the tumorigenesis of neuroblastoma, the inverse relationship between trkA and IGF-II strongly suggests that a low level of trkA can be a feature of the pathogenetic mechanism of IGF-II expressing adrenal neuroblastomas.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator de Crescimento Insulin-Like II/genética , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Neoplasias das Glândulas Suprarrenais/metabolismo , Pré-Escolar , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Lactente , Fator de Crescimento Insulin-Like II/análise , Masculino , Fatores de Crescimento Neural/farmacologia , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/análise , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/genética , Receptor trkA , Receptores de Fator de Crescimento Neural/análise , Receptores de Fator de Crescimento Neural/genética , Transfecção
14.
J Immunol ; 162(10): 6268-77, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10229874

RESUMO

Neurologic abnormalities are common in HIV-1-infected patients and often represent the dominant clinical manifestation of pediatric AIDS. The neurological dysfunction has been directly related to CNS invasion by HIV-1 that is principally, if not exclusively, supported by blood-derived monocytes/macrophages and lymphocytes. By using primary long term cultures of human fetal sensory neurons as well as sympathetic precursors-like neuronal cells, we determined that blood-derived mononuclear cells from HIV-1-infected individuals spontaneously release soluble mediators that can potently inhibit the growth and survival of developing neurons as well as the viability of postmitotic neuronal cells by inducing apoptotic cell death. Analysis of the cytokines produced by lymphomonocytic cells, HIV-1 infected or activated, indicated that oncostatin M (oncM) is a major mediator of these effects. Since low TGF-beta1 concentrations were capable of enhancing oncM-mediated neuronal alterations, our data indicate that by acting in concert with other cytokines, oncM may induce neuronal demise in both the developing and the mature brain. Thus, this cytokine may contribute to the setting of the neuronal cell damage observed in HIV-1-infected individuals.


Assuntos
Citocinas/metabolismo , Infecções por HIV/imunologia , HIV-1/imunologia , Mediadores da Inflamação/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Neurotoxinas/metabolismo , Peptídeos/metabolismo , Apoptose , Bioensaio , Fragmentação do DNA , Interações Medicamentosas , Infecções por HIV/patologia , Humanos , Marcação In Situ das Extremidades Cortadas , Leucócitos Mononucleares/imunologia , Neurônios Aferentes/patologia , Oncostatina M , Fatores de Tempo , Fator de Crescimento Transformador beta/farmacologia
15.
Neuroscience ; 86(3): 881-93, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9692724

RESUMO

Throughout life, olfactory sensory neurons are renewed from a population of dividing stem cells. Little is known about the molecular mechanisms that regulate the activation, self-renewal and differentiation of olfactory neuronal precursors; however, evidence indicates that soluble mediators may play a central role in olfactory neurogenesis. To identify molecules that regulate olfactory self-renewal and differentiation, we have recently established, cloned and propagated in vitro primary long-term cell cultures from the human fetal olfactory neuroepithelium. Here we show that primary human olfactory neuroblasts synthesize and release biologically active basic fibroblast growth factor which, in turn, supports neuroblast growth by autocrine/paracrine mechanisms. The growth-promoting activity of basic fibroblast growth factor is dose dependent and is accompanied by morphological changes of the cells and by an increase in the expression of neuronal-related genes. These observations indicate that endogenous basic fibroblast growth factor participates in controlling olfactory self-renewal and suggest that this cytokine represents a key regulatory element of olfactory neurogenesis.


Assuntos
Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Substâncias de Crescimento/farmacologia , Mucosa Nasal/inervação , Neurônios/citologia , Neurônios/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Feto , Fator 2 de Crescimento de Fibroblastos/farmacologia , Substâncias de Crescimento/fisiologia , Humanos , Mucosa Nasal/citologia , Mucosa Nasal/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/farmacologia
16.
Oncogene ; 16(25): 3337-43, 1998 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-9681834

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular , Proteínas Associadas aos Microtúbulos , Proteínas Supressoras de Tumor , Antineoplásicos/farmacologia , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor p16 de Quinase Dependente de Ciclina/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/efeitos dos fármacos , Ciclinas/metabolismo , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Genes Supressores de Tumor , Humanos , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Neuroblastoma/genética , Neuroblastoma/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tretinoína/farmacologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
18.
J Biol Chem ; 272(42): 26693-701, 1997 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-9334253

RESUMO

Naturally occurring retinoids, like all-trans retinoic acid and 9-cis retinoic acid, are known to affect proliferation and differentiation of sensitive neuroblastoma cell lines. Cellular responsiveness to retinoic acid depends on its interaction with two distinct classes of receptors, the retinoic acid receptors (RARs) and the retinoic X receptors (RXRs). Both receptor classes have three different subtypes (RARalpha, RARbeta, and RARgamma and RXRalpha, RARbeta, and RARgamma) that act as ligand-dependent transcription factors. To examine the involvement of the different receptor classes and subtypes in the biological responses of neuroblastoma cells to retinoids, we analyzed the effects of a panel of receptor-selective retinoids on cell growth, differentiation, and gene expression on in vitro cultured KCNR cells. Any association of per se inactive RXR-selective with RAR-selective ligands efficiently regulates growth inhibition, differentiation (neurite extension), and expression of RARbeta, TrkB, and N-myc. SR11383 alone, a very potent retinoid, entirely reproduces the pattern of biological responses induced by naturally occurring retinoids. In contrast to other tumor cell lines, the growth of neuroblastoma cell lines is not altered using AP1-antagonistic retinoids. These studies raise the possibility that three distinct RXR/RAR heterodimers mediate the effects of retinoids on neuroblastoma cells through an AP-1 antagonism-independent mechanism.


Assuntos
Neuroblastoma/metabolismo , Receptores do Ácido Retinoico/metabolismo , Fatores de Transcrição/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dimerização , Regulação da Expressão Gênica/efeitos dos fármacos , Neuroblastoma/patologia , Receptores X de Retinoides , Retinoides/farmacologia , Células Tumorais Cultivadas
19.
J Neurovirol ; 3(4): 290-8, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9291237

RESUMO

Neurologic abnormalities are common in HIV-1 infected patients and often represent the dominant clinical manifestation of pediatric AIDS. Although the neurological dysfunction has been directly related to CNS invasion by HIV-1, the pathogenesis of neurologic disorders remains unclear. Microglia and macrophages are major HIV-1 targets in the brain, whereas HIV-1 infected neurons or glial cells have been rarely reported. This suggests that indirect mechanisms may account for the severe neuronal damage observed in these patients. Nevertheless, immature, mitotically active neuronal and glial cells, which are present during fetal development, are susceptible to HIV-1 infection and replication in vitro, suggesting that HIV-1 infection during organ development may present unique features. To better characterize virus-host cells interactions in the developing CNS, we have examined the susceptibility of embryologically and biochemically distinct neuronal cell lines to HIV-1 infection. Here we show that mitotically active, immature neurons of distinct lineages, have different susceptibilities to HIV-1 infection and replication and different abilities to support viral gene expression. Mutational analysis of HIV-1 LTR reveals that a region of the viral promoter between nucleotide -255 to -166 is responsible for most quantitative and qualitative differences in viral transactivation among different neuroblasts. This suggests that specific regions of the viral promoter and cellular factors, either lineage- or differentiation-dependent, which bind to those regions, may contribute to control the levels of virus replication and possibly restrict the viral tropism in the developing brain. This may contribute to the establishment of a virus reservoir in the immature CNS and participate by either direct or indirect mechanisms to the severity of the AIDS-related pediatric neurological dysfunction.


Assuntos
Sistema Nervoso Central/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Linhagem Celular , Linhagem da Célula , Sistema Nervoso Central/crescimento & desenvolvimento , Regulação Viral da Expressão Gênica , Infecções por HIV/genética , Repetição Terminal Longa de HIV , HIV-1/genética , Humanos , Replicação Viral
20.
J Biol Chem ; 272(18): 12195-201, 1997 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-9115293

RESUMO

A cDNA, 7G1, was isolated from retinoic acid (RA) differentiated neuroblastoma cells whose expression was high in human fetal brain and spinal cord mRNA but undetectable in adult brain or non-neuronal tissues. Sequence analysis indicates that 7G1 is homologous to the Caenorhabditis elegans gene unc-33. A 5.5-kilobase pair full-length cDNA from a human fetal brain cDNA library contains an 1710-base pair open reading frame. Because the predicted 570 amino acid sequence of 7G1 shares 98% identity with the murine Ulip gene product, an unc-33-like-phosphoprotein, we refer to 7G1 as the human Ulip (hUlip). hUlip is also similar to the bacterial enzyme D-hydantoinase and the recently described vertebrate gene products CRMP62, TOAD-64, CRMP1, CRMP2, and mUNC. RA stimulates an increase in hUlip mRNA that is transcriptionally regulated. RA stimulates an increase in polypeptides of 58, 60, 65, and 70 kDa with the 58- and 65-kDa species being dephosphorylated forms of the 60- and 70-kDa species. This study presents a model in which to study the regulation and expression of the hUlip gene, a member of an emerging family of molecules that potentially mediates signals involved in axonal outgrowth.


Assuntos
Encéfalo/metabolismo , Proteínas de Caenorhabditis elegans , Proteínas Musculares , Neuroblastoma/metabolismo , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Medula Espinal/metabolismo , Tretinoína/farmacologia , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Feto , Regulação Neoplásica da Expressão Gênica , Biblioteca Gênica , Proteínas de Helminto/química , Humanos , Camundongos , Dados de Sequência Molecular , Fatores de Crescimento Neural/química , Neuroblastoma/genética , Neurônios/metabolismo , Fragmentos de Peptídeos/química , Fosfoproteínas/química , RNA Mensageiro/biossíntese , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Células Tumorais Cultivadas , Vertebrados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...