Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 40(24): e105862, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34786738

RESUMO

The onset of random X chromosome inactivation in mouse requires the switch from a symmetric to an asymmetric state, where the identities of the future inactive and active X chromosomes are assigned. This process is known as X chromosome choice. Here, we show that RIF1 and KAP1 are two fundamental factors for the definition of this transcriptional asymmetry. We found that at the onset of differentiation of mouse embryonic stem cells (mESCs), biallelic up-regulation of the long non-coding RNA Tsix weakens the symmetric association of RIF1 with the Xist promoter. The Xist allele maintaining the association with RIF1 goes on to up-regulate Xist RNA expression in a RIF1-dependent manner. Conversely, the promoter that loses RIF1 gains binding of KAP1, and KAP1 is required for the increase in Tsix levels preceding the choice. We propose that the mutual exclusion of Tsix and RIF1, and of RIF1 and KAP1, at the Xist promoters establish a self-sustaining loop that transforms an initially stochastic event into a stably inherited asymmetric X-chromosome state.


Assuntos
Células-Tronco Embrionárias Murinas/citologia , RNA Longo não Codificante/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Feminino , Camundongos , Regiões Promotoras Genéticas , Processos Estocásticos , Regulação para Cima , Inativação do Cromossomo X
2.
Sci Rep ; 7(1): 2119, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28522851

RESUMO

Rif1 is a conserved protein that plays essential roles in orchestrating DNA replication timing, controlling nuclear architecture, telomere length and DNA repair. However, the relationship between these different roles, as well as the molecular basis of Rif1 function is still unclear. The association of Rif1 with insoluble nuclear lamina has thus far hampered exhaustive characterization of the associated protein complexes. We devised a protocol that overcomes this problem, and were thus able to discover a number of novel Rif1 interactors, involved in chromatin metabolism and phosphorylation. Among them, we focus here on PP1. Data from different systems have suggested that Rif1-PP1 interaction is conserved and has important biological roles. Using mutagenesis, NMR, isothermal calorimetry and surface plasmon resonance we demonstrate that Rif1 is a high-affinity PP1 adaptor, able to out-compete the well-established PP1-inhibitor I2 in vitro. Our conclusions have important implications for understanding Rif1 diverse roles and the relationship between the biological processes controlled by Rif1.


Assuntos
Proteínas de Ligação a Telômeros/química , Animais , Sítios de Ligação , Linhagem Celular , Camundongos , Mutação Puntual , Ligação Proteica , Proteína Fosfatase 1/química , Proteína Fosfatase 1/metabolismo , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo
3.
Adv Exp Med Biol ; 1042: 259-272, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29357062

RESUMO

Eukaryotic genomes are replicated starting from multiple origins of replication. Their usage is tightly regulated, and not all the potential origins are activated during a single cell cycle. In addition, the ones that are activated are activated in a sequential order. Why don't origins of replication normally all fire together? Is this important? And if so, why? Would any order of firing do, or does the specific sequence matter? How is this process regulated? These questions concern all eukaryotes but have proven extremely hard to address because replication timing is a process intricately connected with multiple aspects of nuclear function.


Assuntos
Replicação do DNA/fisiologia , Genoma/genética , Mamíferos/genética , Proteínas de Ligação a Telômeros/fisiologia , Animais , Proteínas de Ciclo Celular/fisiologia , Divisão Celular/genética , Período de Replicação do DNA/genética , Instabilidade Genômica/fisiologia , Humanos
4.
J Biol Chem ; 289(20): 13903-11, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24634216

RESUMO

Mammalian Rif1 is a key regulator of DNA replication timing, double-stranded DNA break repair, and replication fork restart. Dissecting the molecular functions of Rif1 is essential to understand how it regulates such diverse processes. However, Rif1 is a large protein that lacks well defined functional domains and is predicted to be largely intrinsically disordered; these features have hampered recombinant expression of Rif1 and subsequent functional characterization. Here we applied ESPRIT (expression of soluble proteins by random incremental truncation), an in vitro evolution-like approach, to identify high yielding soluble fragments encompassing conserved regions I and II (CRI and CRII) at the C-terminal region of murine Rif1. NMR analysis showed CRI to be intrinsically disordered, whereas CRII is partially folded. CRII binds cruciform DNA with high selectivity and micromolar affinity and thus represents a functional DNA binding domain. Mutational analysis revealed an α-helical region of CRII to be important for cruciform DNA binding and identified critical residues. Thus, we present the first structural study of the mammalian Rif1, identifying a domain that directly links its function to DNA binding. The high specificity of Rif1 for cruciform structures is significant given the role of this key protein in regulating origin firing and DNA repair.


Assuntos
Fenômenos Biofísicos , DNA Cruciforme/metabolismo , Proteínas de Ligação a Telômeros/química , Proteínas de Ligação a Telômeros/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Cruciforme/genética , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Solubilidade , Especificidade por Substrato
5.
Science ; 339(6120): 700-4, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23306437

RESUMO

The choice between double-strand break (DSB) repair by either homology-directed repair (HDR) or nonhomologous end joining (NHEJ) is tightly regulated. Defects in this regulation can induce genome instability and cancer. 53BP1 is critical for the control of DSB repair, promoting NHEJ, and inhibiting the 5' end resection needed for HDR. Using dysfunctional telomeres and genome-wide DSBs, we identify Rif1 as the main factor used by 53BP1 to impair 5' end resection. Rif1 inhibits resection involving CtIP, BLM, and Exo1; limits accumulation of BRCA1/BARD1 complexes at sites of DNA damage; and defines one of the mechanisms by which 53BP1 causes chromosomal abnormalities in Brca1-deficient cells. These data establish Rif1 as an important contributor to the control of DSB repair by 53BP1.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Animais , Proteína BRCA1/metabolismo , Células Cultivadas , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Camundongos , Proteína de Replicação A/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
6.
EMBO J ; 31(18): 3678-90, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22850673

RESUMO

The eukaryotic genome is replicated according to a specific spatio-temporal programme. However, little is known about both its molecular control and biological significance. Here, we identify mouse Rif1 as a key player in the regulation of DNA replication timing. We show that Rif1 deficiency in primary cells results in an unprecedented global alteration of the temporal order of replication. This effect takes place already in the first S-phase after Rif1 deletion and is neither accompanied by alterations in the transcriptional landscape nor by major changes in the biochemical identity of constitutive heterochromatin. In addition, Rif1 deficiency leads to both defective G1/S transition and chromatin re-organization after DNA replication. Together, these data offer a novel insight into the global regulation and biological significance of the replication-timing programme in mammalian cells.


Assuntos
Replicação do DNA , Regulação da Expressão Gênica , Proteínas de Ligação a Telômeros/genética , Alelos , Animais , Ciclo Celular , Feminino , Fase G1 , Genoma , Genótipo , Heterocromatina/química , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal/métodos , Matriz Nuclear/metabolismo , Fase S , Transcrição Gênica
7.
J Cell Biol ; 187(3): 385-98, 2009 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-19948482

RESUMO

Rif1, originally recognized for its role at telomeres in budding yeast, has been implicated in a wide variety of cellular processes in mammals, including pluripotency of stem cells, response to double-strand breaks, and breast cancer development. As the molecular function of Rif1 is not known, we examined the consequences of Rif1 deficiency in mouse cells. Rif1 deficiency leads to failure in embryonic development, and conditional deletion of Rif1 from mouse embryo fibroblasts affects S-phase progression, rendering cells hypersensitive to replication poisons. Rif1 deficiency does not alter the activation of the DNA replication checkpoint but rather affects the execution of repair. RNA interference to human Rif1 decreases the efficiency of homology-directed repair (HDR), and Rif1 deficiency results in aberrant aggregates of the HDR factor Rad51. Consistent with a role in S-phase progression, Rif1 accumulates at stalled replication forks, preferentially around pericentromeric heterochromatin. Collectively, these findings reveal a function for Rif1 in the repair of stalled forks by facilitating HDR.


Assuntos
Reparo do DNA , Replicação do DNA , Proteínas de Ligação a Telômeros/fisiologia , Animais , Dano ao DNA , Desenvolvimento Embrionário/genética , Heterocromatina , Humanos , Camundongos , Interferência de RNA , Rad51 Recombinase/metabolismo , Fase S/genética , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo
8.
Genes Dev ; 18(17): 2108-19, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342490

RESUMO

We report on the function of the human ortholog of Saccharomyces cerevisiae Rif1 (Rap1-interacting factor 1). Yeast Rif1 associates with telomeres and regulates their length. In contrast, human Rif1 did not accumulate at functional telomeres, but localized to dysfunctional telomeres and to telomeric DNA clusters in ALT cells, a pattern of telomere association typical of DNA-damage-response factors. After induction of double-strand breaks (DSBs), Rif1 formed foci that colocalized with other DNA-damage-response factors. This response was strictly dependent on ATM (ataxia telangiectasia mutated) and 53BP1, but not affected by diminished function of ATR (ATM- and Rad3-related kinase), BRCA1, Chk2, Nbs1, and Mre11. Rif1 inhibition resulted in radiosensitivity and a defect in the intra-S-phase checkpoint. The S-phase checkpoint phenotype was independent of Nbs1 status, arguing that Rif1 and Nbs1 act in different pathways to inhibit DNA replication after DNA damage. These data reveal that human Rif1 contributes to the ATM-mediated protection against DNA damage and point to a remarkable difference in the primary function of this protein in yeast and mammals.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Serina-Treonina Quinases/metabolismo , Fase S/fisiologia , Proteínas de Ligação a Telômeros/metabolismo , Telômero/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Sequência de Bases , Proteínas de Transporte , Proteínas de Ciclo Celular , Células Cultivadas , Dano ao DNA/fisiologia , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/fisiologia , Imunofluorescência , Humanos , Immunoblotting , Fosfoproteínas , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Repressoras/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Proteínas de Ligação a Telômeros/fisiologia , Transfecção , Proteínas Supressoras de Tumor , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Raios Ultravioleta
9.
Dev Cell ; 4(5): 727-39, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12737807

RESUMO

Disjunction of maternal and paternal centromeres during meiosis I requires crossing over between homologous chromatids, which creates chiasmata that hold homologs together. It also depends on a mechanism ensuring that maternal and paternal sister kinetochore pairs attach to oppositely oriented microtubules. Proteolytic cleavage of cohesin's Rec8 subunit by separase destroys cohesion between sister chromatid arms at anaphase I and thereby resolves chiasmata. The Spo12 and Slk19 proteins have been implicated in regulating meiosis I kinetochore orientation and/or in preventing cleavage of Rec8 at centromeres. We show here that the role of these proteins is instead to promote nucleolar segregation, including release of the Cdc14 phosphatase required for Cdk1 inactivation and disassembly of the anaphase I spindle. Separase is also required but surprisingly not its protease activity. It has two mechanistically different roles during meiosis I. Loss of the protease-independent function alone results in a second meiotic division occurring on anaphase I spindles in spo12delta and slk19delta mutants.


Assuntos
Anáfase , Proteínas de Ciclo Celular/metabolismo , Nucléolo Celular/metabolismo , Endopeptidases , Proteínas Fúngicas/metabolismo , Meiose , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ciclina B , Ciclinas/metabolismo , Regulação para Baixo , Proteínas Fúngicas/genética , Hibridização in Situ Fluorescente , Proteínas Associadas aos Microtúbulos/genética , Proteínas Nucleares , Proteínas Quinases/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Separase , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...