Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
Chromosoma ; 124(2): 277-87, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25413176

RESUMO

The centromere directs the segregation of chromosomes during mitosis and meiosis. It is a distinct genetic locus whose identity is established through epigenetic mechanisms that depend on the deposition of centromere-specific centromere protein A (CENP-A) nucleosomes. This important chromatin domain has so far escaped comprehensive molecular analysis due to its typical association with highly repetitive satellite DNA. In previous work, we discovered that the centromere of horse chromosome 11 is completely devoid of satellite DNA; this peculiar feature makes it a unique model to dissect the molecular architecture of mammalian centromeres. Here, we exploited this native satellite-free centromere to determine the precise localization of its functional domains in five individuals: We hybridized DNA purified from chromatin immunoprecipitated with an anti CENP-A antibody to a high resolution array (ChIP-on-chip) of the region containing the primary constriction of horse chromosome 11. Strikingly, each individual exhibited a different arrangement of CENP-A binding domains. We then analysed the organization of each domain using a single nucleotide polymorphism (SNP)-based approach and single molecule analysis on chromatin fibres. Examination of the ten instances of chromosome 11 in the five individuals revealed seven distinct 'positional alleles', each one extending for about 80-160 kb, were found across a region of about 500 kb. Our results demonstrate that CENP-A binding domains are autonomous relative to the underlying DNA sequence and are characterized by positional instability causing the sliding of centromere position. We propose that this dynamic behaviour may be common in mammalian centromeres and may determine the establishment of epigenetic alleles.


Assuntos
Centrômero/genética , Cromossomos de Mamíferos/genética , Cavalos/genética , Alelos , Animais , Autoantígenos/genética , Linhagem Celular , Proteína Centromérica A , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Clonagem Molecular , DNA Satélite , Epigênese Genética , Feminino , Masculino , Meiose , Procedimentos Analíticos em Microchip , Mitose , Nucleossomos/genética , Polimorfismo de Nucleotídeo Único
3.
Genome Res ; 21(1): 137-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21081712

RESUMO

Despite its importance in cell biology and evolution, the centromere has remained the final frontier in genome assembly and annotation due to its complex repeat structure. However, isolation and characterization of the centromeric repeats from newly sequenced species are necessary for a complete understanding of genome evolution and function. In recent years, various genomes have been sequenced, but the characterization of the corresponding centromeric DNA has lagged behind. Here, we present a computational method (RepeatNet) to systematically identify higher-order repeat structures from unassembled whole-genome shotgun sequence and test whether these sequence elements correspond to functional centromeric sequences. We analyzed genome datasets from six species of mammals representing the diversity of the mammalian lineage, namely, horse, dog, elephant, armadillo, opossum, and platypus. We define candidate monomer satellite repeats and demonstrate centromeric localization for five of the six genomes. Our analysis revealed the greatest diversity of centromeric sequences in horse and dog in contrast to elephant and armadillo, which showed high-centromeric sequence homogeneity. We could not isolate centromeric sequences within the platypus genome, suggesting that centromeres in platypus are not enriched in satellite DNA. Our method can be applied to the characterization of thousands of other vertebrate genomes anticipated for sequencing in the near future, providing an important tool for annotation of centromeres.


Assuntos
Centrômero/genética , Biologia Computacional/métodos , DNA Satélite/genética , Genoma/genética , Mamíferos/classificação , Mamíferos/genética , Animais , Tatus/genética , Sequência de Bases , Sequência Consenso , Cães/genética , Elefantes/genética , Cavalos/genética , Dados de Sequência Molecular , Gambás/genética , Ornitorrinco/genética , Análise de Sequência de DNA , Especificidade da Espécie
4.
J Biol Chem ; 285(25): 19532-43, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20233711

RESUMO

Increased expression of specific ATP-binding cassette (ABC) transporters is known to mediate the efflux of chemotherapeutic agents from cancer cells. Therefore, establishing how ABC transporter genes are controlled at their transcription level may help provide insight into the role of these multifaceted transporters in the malignant phenotype. We have investigated ABC transporter gene expression in a large neuroblastoma data set of 251 tumor samples. Clustering analysis demonstrated a strong association between differential ABC gene expression patterns in tumor samples and amplification of the MYCN oncogene, suggesting a correlation with MYCN function. Using expression profiling and chromatin immunoprecipitation studies, we show that MYCN oncoprotein coordinately regulates transcription of specific ABC transporter genes, by acting as either an activator or a repressor. Finally, we extend these notions to c-MYC showing that it can also regulate the same set of ABC transporter genes in other tumor cells through similar dynamics. Overall our findings provide insight into MYC-driven molecular mechanisms that contribute to coordinate transcriptional regulation of a large set of ABC transporter genes, thus affecting global drug efflux.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Perfilação da Expressão Gênica , Humanos , Concentração Inibidora 50 , Modelos Genéticos , Fenótipo , Retinoblastoma/metabolismo , Transcrição Gênica
6.
Hum Mol Genet ; 18(17): 3178-93, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19482874

RESUMO

DNA methylation is an epigenetically imposed mark of transcriptional repression that is essential for maintenance of chromatin structure and genomic stability. Genome-wide methylation patterns are mediated by the combined action of three DNA methyltransferases: DNMT1, DNMT3A and DNMT3B. Compelling links exist between DNMT3B and chromosome stability as emphasized by the mitotic defects that are a hallmark of ICF syndrome, a disease arising from germline mutations in DNMT3B. Centromeric and pericentromeric regions are essential for chromosome condensation and the fidelity of segregation. Centromere regions contain distinct epigenetic marks, including dense DNA hypermethylation, yet the mechanisms by which DNA methylation is targeted to these regions remains largely unknown. In the present study, we used a yeast two-hybrid screen and identified a novel interaction between DNMT3B and constitutive centromere protein CENP-C. CENP-C is itself essential for mitosis. We confirm this interaction in mammalian cells and map the domains responsible. Using siRNA knock downs, bisulfite genomic sequencing and ChIP, we demonstrate for the first time that CENP-C recruits DNA methylation and DNMT3B to both centromeric and pericentromeric satellite repeats and that CENP-C and DNMT3B regulate the histone code in these regions, including marks characteristic of centromeric chromatin. Finally, we demonstrate that loss of CENP-C or DNMT3B leads to elevated chromosome misalignment and segregation defects during mitosis and increased transcription of centromeric repeats. Taken together, our data reveal a novel mechanism by which DNA methylation is targeted to discrete regions of the genome and contributes to chromosomal stability.


Assuntos
Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Código das Histonas , Linhagem Celular , Centrômero/genética , Proteínas Cromossômicas não Histona/genética , DNA (Citosina-5-)-Metiltransferases/genética , Células HCT116 , Células HeLa , Humanos , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , DNA Metiltransferase 3B
7.
J Exp Med ; 195(7): 907-18, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11927634

RESUMO

The neurodegenerative changes in Alzheimer's disease (AD) are elicited by the accumulation of beta-amyloid peptides (Abeta), which damage neurons either directly by interacting with components of the cell surface to trigger cell death signaling or indirectly by activating astrocytes and microglia to produce inflammatory mediators. It has been recently proposed that the p75 neurotrophin receptor (p75(NTR)) is responsible for neuronal damage by interacting with Abeta. By using neuroblastoma cell clones lacking the expression of all neurotrophin receptors or engineered to express full-length or various truncated forms of p75(NTR), we could show that p75(NTR) is involved in the direct signaling of cell death by Abeta via the function of its death domain. This signaling leads to the activation of caspases-8 and -3, the production of reactive oxygen intermediates and the induction of an oxidative stress. We also found that the direct and indirect (inflammatory) mechanisms of neuronal damage by Abeta could act synergistically. In fact, TNF-alpha and IL-1beta, cytokines produced by Abeta-activated microglia, could potentiate the neurotoxic action of Abeta mediated by p75(NTR) signaling. Together, our results indicate that neurons expressing p75(NTR), mostly if expressing also proinflammatory cytokine receptors, might be preferential targets of the cytotoxic action of Abeta in AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Citocinas/toxicidade , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Receptores de Fator de Crescimento Neural/fisiologia , Células Clonais , Sinergismo Farmacológico , Regulação da Expressão Gênica , Humanos , Cinética , Neuroblastoma , Neurônios/patologia , Fragmentos de Peptídeos/toxicidade , Receptor de Fator de Crescimento Neural , Receptor trkA/genética , Receptor trkA/fisiologia , Receptores de Fator de Crescimento Neural/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transfecção , Células Tumorais Cultivadas
8.
Proc Natl Acad Sci U S A ; 104(47): 18682-7, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18003922

RESUMO

Histone deacetylase (HDAC) inhibitors reactivate tumor suppressor gene transcription; induce cancer cell differentiation, growth arrest, and programmed cell death; and are among the most promising new classes of anticancer drugs. Myc oncoproteins can block cell differentiation and promote cell proliferation and malignant transformation, in some cases by modulating target gene transcription. Here, we show that tissue transglutaminase (TG2) was commonly reactivated by HDAC inhibitors in neuroblastoma and breast cancer cells but not normal cells and contributed to HDAC inhibitor-induced growth arrest. TG2 was the gene most significantly repressed by N-Myc in neuroblastoma cells in a cDNA microarray analysis and was commonly repressed by N-Myc in neuroblastoma cells and c-Myc in breast cancer cells. Repression of TG2 expression by N-Myc in neuroblastoma cells was necessary for the inhibitory effect of N-Myc on neuroblastoma cell differentiation. Dual step cross-linking chromatin immunoprecipitation and protein coimmunoprecipitation assays showed that N-Myc acted as a transrepressor by recruiting the HDAC1 protein to an Sp1-binding site in the TG2 core promoter in a manner distinct from it's action as a transactivator at E-Box binding sites. HDAC inhibitor treatment blocked the N-Myc-mediated HDAC1 recruitment and TG2 repression in vitro. In neuroblastoma-bearing N-Myc transgenic mice, HDAC inhibitor treatment induced TG2 expression and demonstrated marked antitumor activity in vivo. Taken together, our data indicate the critical roles of HDAC1 and TG2 in Myc-induced oncogenesis and have significant implications for the use of HDAC inhibitor therapy in Myc-driven oncogenesis.


Assuntos
Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transcrição Gênica/genética , Transglutaminases/genética , Transglutaminases/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Camundongos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteína 2 Glutamina gama-Glutamiltransferase , Regulação para Cima/efeitos dos fármacos
9.
FEBS Lett ; 581(5): 821-5, 2007 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17289033

RESUMO

In this study, we report that the human p14(ARF) associates in vivo with the N-Myc and inhibits N-Myc mediated transcriptional activation. We have determined that the region (aa 140-300) encompassing the N-Myc BoxIII is required for efficient interaction in vivo. Furthermore, we demonstrate that in the SK-N-BE neuroblastoma cell line p14(ARF) over-expression delocalized N-Myc from the nucleoplasm into nucleoli and that N-Myc regions required for interaction with p14(ARF) are also important for nucleoli co-localization. Finally, we determine that the N-terminal region of the p14(ARF) protein is involved in binding to c-Myc and N-Myc proteins.


Assuntos
Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transcrição Gênica , Transfecção , Proteína Supressora de Tumor p14ARF/química , Proteína Supressora de Tumor p14ARF/genética
10.
Biochem J ; 391(Pt 1): 59-67, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15966860

RESUMO

Aging is the single most important risk factor for AD (Alzheimer's disease). However, the molecular events that connect normal aging to AD are mostly unknown. The abnormal accumulation of Abeta (amyloid beta-peptide) in the form of senile plaques is one of the main characteristics of AD. In the present study, we show that two members of the neurotrophin receptor superfamily, TrkA (tyrosine kinase receptor A) and p75NTR (p75 neurotrophin receptor), differentially regulate the processing of APP (amyloid precursor protein): TrkA reduces, whereas p75NTR activates, beta-cleavage of APP. The p75NTR-dependent effect requires NGF (nerve growth factor) binding and activation of the second messenger ceramide. We also show that normal aging activates Abeta generation in the brain by 'switching' from the TrkA to the p75NTR receptor system. Such an effect is abolished in p75NTR 'knockout' animals, and can be blocked by both caloric restriction and inhibitors of nSMase (neutral sphingomyelinase). In contrast with caloric restriction, which prevents the age-associated up-regulation of p75NTR expression, nSMase inhibitors block the activation of ceramide. When taken together, these results indicate that the p75NTR-ceramide signalling pathway activates the rate of Abeta generation in an age-dependent fashion, and provide a new target for both the understanding and the prevention of late-onset AD.


Assuntos
Envelhecimento/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Animais , Restrição Calórica , Linhagem Celular Tumoral , Ceramidas/metabolismo , Córtex Cerebral/metabolismo , Deleção de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polienos , Alcamidas Poli-Insaturadas , Receptor de Fator de Crescimento Neural/genética , Receptor trkA/genética
11.
Cancer Res ; 71(2): 404-12, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21123453

RESUMO

Neuroblastoma is the most common extracranial solid tumor of childhood. One important factor that predicts a favorable prognosis is the robust expression of the TRKA and p75NTR neurotrophin receptor genes. Interestingly, TRKA and p75NTR expression is often attenuated in aggressive MYCN-amplified tumors, suggesting a causal link between elevated MYCN activity and the transcriptional repression of TRKA and p75NTR, but the precise mechanisms involved are unclear. Here, we show that MYCN acts directly to repress TRKA and p75NTR gene transcription. Specifically, we found that MYCN levels were critical for repression and that MYCN targeted proximal/core promoter regions by forming a repression complex with transcription factors SP1 and MIZ1. When bound to the TRKA and p75NTR promoters, MYCN recruited the histone deacetylase HDAC1 to induce a repressed chromatin state. Forced re-expression of endogenous TRKA and p75NTR with exposure to the HDAC inhibitor TSA sensitized neuroblastoma cells to NGF-mediated apoptosis. By directly connecting MYCN to the repression of TRKA and p75NTR, our findings establish a key pathway of clinical pathogenicity and aggressiveness in neuroblastoma.


Assuntos
Histona Desacetilase 1/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Receptor trkA/genética , Receptores de Fator de Crescimento Neural/genética , Fator de Transcrição Sp1/genética , Células HEK293 , Células HeLa , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Proteína Proto-Oncogênica N-Myc , Proteínas do Tecido Nervoso/biossíntese , Neuroblastoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Receptor trkA/biossíntese , Receptores de Fator de Crescimento Neural/biossíntese , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica , Transfecção
12.
PLoS One ; 4(6): e5832, 2009 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-19503796

RESUMO

CENP-C is a fundamental component of functional centromeres. The elucidation of its structure-function relationship with centromeric DNA and other kinetochore proteins is critical to the understanding of centromere assembly. CENP-C carries two regions, the central and the C-terminal domains, both of which are important for the ability of CENP-C to associate with the centromeric DNA. However, while the central region is largely divergent in CENP-C homologues, the C-terminal moiety contains two regions that are highly conserved from yeast to humans, named Mif2p homology domains (blocks II and III). The activity of these two domains in human CENP-C is not well defined. In this study we performed a functional dissection of C-terminal CENP-C region analyzing the role of single Mif2p homology domains through in vivo and in vitro assays. By immunofluorescence and Chromatin immunoprecipitation assay (ChIP) we were able to elucidate the ability of the Mif2p homology domain II to target centromere and contact alpha satellite DNA. We also investigate the interactions with other conserved inner kinetochore proteins by means of coimmunoprecipitation and bimolecular fluorescence complementation on cell nuclei. We found that the C-terminal region of CENP-C (Mif2p homology domain III) displays multiple activities ranging from the ability to form higher order structures like homo-dimers and homo-oligomers, to mediate interaction with CENP-A and histone H3. Overall, our findings support a model in which the Mif2p homology domains of CENP-C, by virtue of their ability to establish multiple contacts with DNA and centromere proteins, play a critical role in the structuring of kinethocore chromatin.


Assuntos
Centrômero/ultraestrutura , Proteínas Cromossômicas não Histona/química , Autoantígenos/química , Linhagem Celular , Proteína Centromérica A , DNA/química , Proteínas de Ligação a DNA/química , Dimerização , Epitopos/química , Corantes Fluorescentes/farmacologia , Glutaral/química , Histonas/química , Humanos , Cinetocoros/química , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química
13.
Genome Res ; 19(5): 778-84, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19411601

RESUMO

Molecular cytogenetics provides a visual, pictorial record of the tree of life, and in this respect the fusion origin of human chromosome 2 is a well-known paradigmatic example. Here we report on a variant chromosome 6 in which the centromere jumped to 6p22.1. ChIP-chip experiments with antibodies against the centromeric proteins CENP-A and CENP-C exactly defined the neocentromere as lying at chr6:26,407-26,491 kb. We investigated in detail the evolutionary history of chromosome 6 in primates and found that the primate ancestor had a homologous chromosome with the same marker order, but with the centromere located at 6p22.1. Sometime between 17 and 23 million years ago (Mya), in the common ancestor of humans and apes, the centromere of chromosome 6 moved from 6p22.1 to its current location. The neocentromere we discovered, consequently, has jumped back to the ancestral position, where a latent centromere-forming potentiality persisted for at least 17 Myr. Because all living organisms form a tree of life, as first conceived by Darwin, evolutionary perspectives can provide compelling underlying explicative grounds for contemporary genomic phenomena.


Assuntos
Centrômero/genética , Cromossomos Humanos Par 6/genética , Evolução Molecular , Animais , Autoantígenos/genética , Linhagem Celular Tumoral , Proteína Centromérica A , Proteínas Cromossômicas não Histona/genética , Variação Genética , Genoma Humano , Genômica , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Linhagem
14.
Chromosoma ; 117(4): 339-44, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18274768

RESUMO

It has been hypothesized that human clinical neocentromeres and evolutionary novel centromeres (ENC) represent two faces of the same phenomenon. However, there are only two reports of loci harboring both a novel centromere and a clinical neocentromere. We suggest that only the tip of the iceberg has been scratched because most neocentromerization events have a very low chance of being observed. In support of this view, we report here on a neocentromere at 9q33.1 that emerged in a ring chromosome of about 12 Mb. The ring was produced by a balanced rearrangement that was fortuitously discovered because of its malsegregation in the propositus. Chromatin-immunoprecipitation-on-chip experiments using anti-centromere protein (CENP)-A and anti-CENP-C antibodies strongly indicated that a novel centromeric domain was present in the ring, in a chromosomal domain where an ENC emerged in the ancestor to Old World monkeys.


Assuntos
Centrômero/patologia , Transtornos Cromossômicos/genética , Cromossomos Humanos Par 9/genética , Evolução Molecular , Cromossomos em Anel , Anticorpos/imunologia , Autoantígenos/imunologia , Centrômero/genética , Proteína Centromérica A , Criança , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/imunologia , Transtornos Cromossômicos/patologia , Humanos , Hibridização in Situ Fluorescente , Masculino , Análise em Microsséries
15.
Exp Cell Res ; 313(14): 2980-92, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17619016

RESUMO

The biological complexity of NGF action is achieved by binding two distinct neurotrophin receptors, TrkA and p75(NTR). While several reports have provided lines of evidence on the interaction between TrkA and p75(NTR) at the plasma membrane, much fewer data are available on the consequence of such an interaction in terms of intracellular signaling. In this study, we have focused on how p75(NTR) may affect TrkA downstream signaling with respect to neuronal differentiation. Here, we have shown that cooperation between p75(NTR) and TrkA results in an increased NGF-mediated TrkA autophosphorylation, leads to a sustained activation of ERK1/2 and accelerates neurite outgrowth. Interestingly, neurite outgrowth is concomitant with a selective enhancement of the AP-1 activity and the transcriptional activation of genes such as GAP-43 and p21(CIP/WAF), known to be involved in the differentiation process. Collectively, our results unveil a functional link between the specific expression profile of neurotrophin receptors in neuronal cells and the NGF-mediated regulation of the differentiation process possibly through a persistent ERKs activation and the selective control of the AP-1 activity.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21 , Proteína GAP-43 , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/fisiologia , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ativação Enzimática , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Fator de Crescimento Neural/metabolismo , Neurônios/citologia , Fosforilação , Receptor de Fator de Crescimento Neural/genética , Receptor trkA/genética , Transdução de Sinais/fisiologia , Fator de Transcrição AP-1/genética , Transcrição Gênica , Células Tumorais Cultivadas
16.
Proc Natl Acad Sci U S A ; 102(34): 12117-22, 2005 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-16093321

RESUMO

N-Myc is a transcription factor that forms heterodimers with the protein Max and binds gene promoters by recognizing a DNA sequence, CACGTG, called E-box. The identification of N-myc target genes is an important step for understanding N-Myc biological functions in both physiological and pathological contexts. In this study, we describe the identification of N-Myc-responsive genes through chromatin immunoprecipitation and methylation-sensitive restriction analysis. Results show that N-Myc is a direct regulator of several identified genes, and that methylation of the CpG dinucleotide within the E-box prevents the access of N-Myc to gene promoters in vivo. Furthermore, methylation profile of the E-box within the promoters of EGFR and CASP8, two genes directly controlled by Myc, is cell type-specific, suggesting that differential E-box methylation may contribute to generating unique patterns of Myc-dependent transcription. This study illuminates a central role of DNA methylation in controlling N-Myc occupancy at gene promoters and modulating its transcriptional activity in cancer cells.


Assuntos
Metilação de DNA , Elementos E-Box/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Northern Blotting , Caspase 8 , Caspases/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Elementos E-Box/genética , Ensaio de Desvio de Mobilidade Eletroforética , Genes erbB-1/genética , Humanos , Immunoblotting , Luciferases , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética
17.
J Biol Chem ; 277(51): 49896-902, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12368293

RESUMO

The transcription factor cAMP-response element-binding protein (CREB) mediates survival in many cells, including neurons. Recently, death of cerebellar granule neurons due to nitric oxide (NO) deprivation was shown to be accompanied by down-regulation of CREB activity (). We now provide evidence that overproduction of endogenous NO or supplementation with exogenous NO renders SK-N-BE human neuroblastoma cells more resistant to apoptosis induced by serum deprivation. Parental cells underwent apoptosis after 24 h of serum deprivation, an outcome largely absent in clones overexpressing human neuronal nitric oxide synthase (nNOS). This protective effect was reversed by the inhibition of NOS itself or soluble guanylyl cyclase, pointing at cGMP as an intermediate effector of NO-mediated rescue. A slow-releasing NO donor protected parental cells to a significant extent, thus confirming the survival effect of NO. The impaired viability of serum-deprived parental cells was accompanied by a strong decrease of CREB phosphorylation and transcriptional activity, effects significantly attenuated in nNOS-overexpressing clones. To confirm the role of CREB in survival, the ectopic expression of CREB and/or protein kinase A largely counteracted serum deprivation-induced cell death of SK-N-BE cells, whereas transfection with a CREB negative mutant was ineffective. These experiments indicate that CREB activity is an important step for NO-mediated survival in neuronal cells.


Assuntos
Apoptose , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Neuroblastoma/patologia , Óxido Nítrico/farmacologia , Western Blotting , Sobrevivência Celular , Colforsina/farmacologia , Meios de Cultura Livres de Soro/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Genes Reporter , Humanos , Marcação In Situ das Extremidades Cortadas , Luciferases/metabolismo , Microscopia de Fluorescência , Mutação , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Fosforilação , Plasmídeos/metabolismo , Isoformas de Proteínas , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
18.
J Cell Sci ; 115(Pt 11): 2317-27, 2002 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12006616

RESUMO

CENP-C is a fundamental component of the centromere, highly conserved among species and necessary for the proper assembly of the kinetochore structure and for the metaphase-anaphase transition. Although CENP-C can bind DNA in vitro, the identification of the DNA sequences associated with it in vivo and the significance of such an interaction have been, until now, elusive. To address this problem we took advantage of a chromatin-immunoprecipitation procedure and applied this technique to human HeLa cells. Through this approach we could establish that: (1) CENP-C binds the alpha-satellite DNA selectively; (2) the CENP-C region between amino acids 410 and 537, previously supposed to contain a DNA-binding domain, is indeed required to perform such a function in vivo; and (3) the profile of the alpha-satellite DNA associated with CENP-C is essentially identical to that recognized by CENP-B. However, further biochemical and ultrastructural characterization of CENP-B/DNA and CENP-C/DNA complexes, relative to their DNA components and specific spatial distribution in interphase nuclei, surprisingly reveals that CENP-C and CENP-B associate with the same types of alpha-satellite arrays but in distinct non-overlapping centromere domains. Our results, besides extending previous observations on the role of CENP-C in the formation of active centromeres, show, for the first time, that CENP-C can associate with the centromeric DNA sequences in vivo and, together with CENP-B, defines a highly structured organization of the alpha-satellite DNA within the human centromere.


Assuntos
Autoantígenos , Divisão Celular/genética , Centrômero/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA Satélite/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Eucarióticas/metabolismo , Sequência de Bases/genética , Bioensaio , Proteína B de Centrômero , Proteínas Cromossômicas não Histona/genética , DNA Satélite/genética , Proteínas de Ligação a DNA/genética , Células Eucarióticas/citologia , Células HeLa , Humanos , Estrutura Terciária de Proteína/genética
19.
J Struct Biol ; 140(1-3): 39-48, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12490152

RESUMO

CENP-C is a fundamental component of the inner kinetochore plate and contributes to the formation of functional centromeres in eukaryotic organisms. Recruitment of CENP-C to kinetochore requires other centromere proteins, particularly CENP-A, CENP-H, and CENP-I. However, how CENP-C is correctly localized at the kinetochore is not clearly determined, mainly due to the functional variety of its domains, which hints at a complex recruitment mechanism. Here, by both immunofluorescent labeling and chromatin/immunoprecipitation we could show that human CENP-C contains two distinct domains, one in the central region, between amino acids 426 and 537, and the second one in the carboxyl terminal region, between amino acids 638 and 943, which are both capable of localizing at centromeres and binding alpha-satellite DNA. The presence of two domains that iterate the same function despite being significantly different in their amino acid sequence and structure suggests that CENP-C may target the centromere by establishing multiple contacts with both the DNA and protein constituents of the kinetochore.


Assuntos
Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/fisiologia , Western Blotting , Centrômero/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Reagentes de Ligações Cruzadas/farmacologia , DNA Satélite/metabolismo , Eletroforese em Gel de Poliacrilamida , Formaldeído/farmacologia , Humanos , Cinetocoros/metabolismo , Microscopia de Fluorescência , Mutação , Testes de Precipitina , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA