Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 287(5): 3156-64, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22167198

RESUMO

The highly conserved DYNLL1 (LC8) protein was originally discovered as a light chain of the dynein motor complex, but is increasingly emerging as a sequence-specific regulator of protein dimerization with hundreds of targets and wide-ranging cellular functions. Despite its important roles, DYNLL1's own regulation remains poorly understood. Here we identify ASCIZ (ATMIN/ZNF822), an essential Zn(2+) finger protein with dual roles in the DNA base damage response and as a developmental transcription factor, as a conserved regulator of Dynll1 gene expression. DYNLL1 levels are reduced by ∼10-fold in the absence of ASCIZ in human, mouse and chicken cells. ASCIZ binds directly to the Dynll1 promoter and regulates its activity in a Zn(2+) finger-dependent manner. DYNLL1 protein in turn interacts with ten binding sites in the ASCIZ transcription activation domain, and high DYNLL1 levels inhibit the transcriptional activity of ASCIZ. In addition, DYNLL1 was also required for DNA damage-induced ASCIZ focus formation. The dual ability of ASCIZ to activate Dynll1 gene expression and to sense free DYNLL1 protein levels enables a simple dynamic feedback loop to adjust DYNLL1 levels to cellular needs. The ASCIZ-DYNLL1 feedback loop represents a novel mechanism for auto-regulation of gene expression, where the gene product directly inhibits the transcriptional activator while bound at its own promoter.


Assuntos
Proteínas de Transporte/metabolismo , Dineínas do Citoplasma/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/fisiologia , Zinco/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/genética , Linhagem Celular , Galinhas , Dineínas do Citoplasma/genética , Humanos , Camundongos , Proteínas Nucleares/genética , Fatores de Transcrição , Transcrição Gênica/fisiologia , Dedos de Zinco
2.
PLoS Genet ; 6(10): e1001170, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20975950

RESUMO

Zn²(+)-finger proteins comprise one of the largest protein superfamilies with diverse biological functions. The ATM substrate Chk2-interacting Zn²(+)-finger protein (ASCIZ; also known as ATMIN and ZNF822) was originally linked to functions in the DNA base damage response and has also been proposed to be an essential cofactor of the ATM kinase. Here we show that absence of ASCIZ leads to p53-independent late-embryonic lethality in mice. Asciz-deficient primary fibroblasts exhibit increased sensitivity to DNA base damaging agents MMS and H2O2, but Asciz deletion knock-down does not affect ATM levels and activation in mouse, chicken, or human cells. Unexpectedly, Asciz-deficient embryos also exhibit severe respiratory tract defects with complete pulmonary agenesis and severe tracheal atresia. Nkx2.1-expressing respiratory precursors are still specified in the absence of ASCIZ, but fail to segregate properly within the ventral foregut, and as a consequence lung buds never form and separation of the trachea from the oesophagus stalls early. Comparison of phenotypes suggests that ASCIZ functions between Wnt2-2b/ß-catenin and FGF10/FGF-receptor 2b signaling pathways in the mesodermal/endodermal crosstalk regulating early respiratory development. We also find that ASCIZ can activate expression of reporter genes via its SQ/TQ-cluster domain in vitro, suggesting that it may exert its developmental functions as a transcription factor. Altogether, the data indicate that, in addition to its role in the DNA base damage response, ASCIZ has separate developmental functions as an essential regulator of respiratory organogenesis.


Assuntos
Proteínas de Transporte/fisiologia , Reparo do DNA/fisiologia , Pulmão/embriologia , Proteínas Nucleares/fisiologia , Organogênese/fisiologia , Animais , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Senescência Celular , Dano ao DNA , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Genótipo , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidantes/farmacologia , Fatores de Tempo , Traqueia/embriologia , Fatores de Transcrição , Raios Ultravioleta
3.
Mol Cell Biol ; 26(14): 5360-72, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16809772

RESUMO

Mutations in the LMNA gene, which encodes all A-type lamins, including lamin A and lamin C, cause a variety of tissue-specific degenerative diseases termed laminopathies. Little is known about the pathogenesis of these disorders. Previous studies have indicated that A-type lamins interact with the retinoblastoma protein (pRB). Here we probe the functional consequences of this association and further examine links between nuclear structure and cell cycle control. Since pRB is required for cell cycle arrest by p16(ink4a), we tested the responsiveness of multiple lamin A/C-depleted cell lines to overexpression of this CDK inhibitor and tumor suppressor. We find that the loss of A-type lamin expression results in marked destabilization of pRB. This reduction in pRB renders cells resistant to p16(ink4a)-mediated G(1) arrest. Reintroduction of lamin A, lamin C, or pRB restores p16(ink4a)-responsiveness to Lmna(-/-) cells. An array of lamin A mutants, representing a variety of pathologies as well as lamin A processing mutants, was introduced into Lmna(-/-) cells. Of these, a mutant associated with mandibuloacral dysplasia (MAD R527H), as well as two lamin A processing mutants, but not other disease-associated mutants, failed to restore p16(ink4a) responsiveness. Although our findings do not rule out links between altered pRB function and laminopathies, they fail to support such an assertion. These findings do link lamin A/C to the functional activation of a critical tumor suppressor pathway and further the possibility that somatic mutations in LMNA contribute to tumor progression.


Assuntos
Ciclo Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Lamina Tipo A/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Sequência de Bases , Linhagem Celular , DNA Complementar/genética , Estabilidade de Medicamentos , Fase G1 , Humanos , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Camundongos , Camundongos Knockout , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética
4.
Trends Cell Biol ; 15(7): 378-85, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15936946

RESUMO

Numerous studies have identified key binding partners and functional activities of nuclear tumor-suppressor proteins such as the retinoblastoma protein, p53 and BRCA1. Historically, less attention has been given to the subnuclear locations of these proteins. Here, we describe several recent studies that promote the view that regulated association with subcompartments of the nucleus is inherent to tumor-suppressor function.


Assuntos
Núcleo Celular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteína BRCA1/metabolismo , Núcleo Celular/ultraestrutura , Proliferação de Células , Fibroblastos , Regulação da Expressão Gênica , Humanos , Camundongos , Células NIH 3T3 , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo
5.
EMBO J ; 24(13): 2447-57, 2005 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-15933716

RESUMO

Nuclear Rad51 focus formation is required for homology-directed repair of DNA double-strand breaks (DSBs), but its regulation in response to non-DSB lesions is poorly understood. Here we report a novel human SQ/TQ cluster domain-containing protein termed ASCIZ that forms Rad51-containing foci in response to base-modifying DNA methylating agents but not in response to DSB-inducing agents. ASCIZ foci seem to form prior to Rad51 recruitment, and an ASCIZ core domain can concentrate Rad51 in focus-like structures independently of DNA damage. ASCIZ depletion dramatically increases apoptosis after methylating DNA damage and impairs Rad51 focus formation in response to methylating agents but not after ionizing radiation. ASCIZ focus formation and increased apoptosis in ASCIZ-depleted cells depend on the mismatch repair protein MLH1. Interestingly, ASCIZ foci form efficiently during G1 phase, when sister chromatids are unavailable as recombination templates. We propose that ASCIZ acts as a lesion-specific focus scaffold in a Rad51-dependent pathway that resolves cytotoxic repair intermediates, most likely single-stranded DNA gaps, resulting from MLH1-dependent processing of base lesions.


Assuntos
Apoptose , Dano ao DNA , Metilação de DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Pareamento Incorreto de Bases , Proteínas de Transporte , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/fisiologia , Estrutura Terciária de Proteína , Rad51 Recombinase , Transdução de Sinais
6.
Oncogene ; 23(1): 307-10, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14712236

RESUMO

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are present in variable number in most human cell types and have been linked to various cellular functions, including roles as depots for DNA repair proteins. Here, we show that treatment of human cells with DNA methylating agents leads to redistribution of PML from NBs to a diffuse nuclear localization. Biochemically, this correlates with a specific reduction of PML levels in the nuclear matrix fraction without affecting total PML levels. Similar results were obtained for the other major PML NB component, the Sp100 protein, indicating that DNA methylating agents lead to a general disassembly of PML NBs. Similar to the dispersal of PML NBs in response to some viral infections, PML redistribution after DNA damage was inhibited by the proteasome inhibitor MG132. We propose that the regulated dispersal of PML NBs may facilitate the enhanced release of DNA repair proteins from NB depots in order to respond adequately to extensive DNA damage.


Assuntos
Núcleo Celular/metabolismo , Cisteína Endopeptidases/fisiologia , Dano ao DNA , Metilação de DNA , Complexos Multienzimáticos/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Fatores de Transcrição/metabolismo , Humanos , Proteína da Leucemia Promielocítica , Complexo de Endopeptidases do Proteassoma , Proteínas Supressoras de Tumor
7.
IUBMB Life ; 55(1): 23-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12716058

RESUMO

Forkhead-associated (FHA) domains are present in >200 diverse proteins in all phyla from bacteria to mammals and seem to be particularly prevalent in proteins with cell cycle control functions. Recent work from several laboratories has considerably improved our understanding of the structure and function of these domains that were virtually unknown a few years ago, and the first disease associations of FHA domains have now emerged. FHA domains form 11-stranded beta-sandwiches that contain some 100-180 amino acid residues with a high degree of sequence diversity. FHA domains act as phosphorylation-dependent protein-protein interaction modules that preferentially bind to phospho-threonine residues in their targets. Interestingly, point mutations in the human CHK2 gene that lead to single-residue amino acid substitutions in the FHA domain of this cell cycle checkpoint kinase have been found to cause a subset of cases of the Li-Fraumeni multi-cancer syndrome.


Assuntos
Fosfotreonina/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Ciclo Celular/fisiologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência
8.
FEBS Lett ; 527(1-3): 71-5, 2002 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-12220636

RESUMO

Parathyroid hormone-related protein (PTHrP) has a diverse range of proposed biological activities participating in both extracellular and intracellular signaling. In order to identify candidate protein effectors, yeast two-hybrid screens were conducted using mature human PTHrP (residues 1-141) and the COOH-terminus (residues 107-141). Both PTHrP baits interacted with a beta-arrestin 1B fragment, an important component of G-protein-coupled receptor desensitization and MAPK signaling. Co-immunoprecipitation, in vitro binding assays and colocalization experiments confirmed this interaction in human cells and this required residues 122-141 of PTHrP. These findings suggest that beta-arrestin 1 acts as an effector for a novel function of PTHrP in cytoplasm.


Assuntos
Arrestinas/metabolismo , Hormônios Peptídicos/metabolismo , Arrestinas/genética , Arrestinas/isolamento & purificação , Sequência de Bases , Núcleo Celular , Células Cultivadas , Citoplasma/metabolismo , Humanos , Dados de Sequência Molecular , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Técnicas do Sistema de Duplo-Híbrido , beta-Arrestina 1 , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...