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.
Mol Cell Biol ; 41(9): e0030321, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34228493

RESUMO

Germline mutations in the mismatch repair (MMR) genes MSH2, MSH6, MLH1, and PMS2 are linked to cancer of the colon and other organs, characterized by microsatellite instability and a large increase in mutation frequency. Unexpectedly, mutations in EXO1, encoding the only exonuclease genetically implicated in MMR, are not linked to familial cancer and cause a substantially weaker mutator phenotype. This difference could be explained if eukaryotic cells possessed additional exonucleases redundant with EXO1. Analysis of the MLH1 interactome identified FANCD2-associated nuclease 1 (FAN1), a novel enzyme with biochemical properties resembling EXO1. We now show that FAN1 efficiently substitutes for EXO1 in MMR assays and that this functional complementation is modulated by its interaction with MLH1. FAN1 also contributes to MMR in vivo; cells lacking both EXO1 and FAN1 have an MMR defect and display resistance to N-methyl-N-nitrosourea (MNU) and 6-thioguanine (TG). Moreover, FAN1 loss amplifies the mutational profile of EXO1-deficient cells, suggesting that the two nucleases act redundantly in the same antimutagenic pathway. However, the increased drug resistance and mutator phenotype of FAN1/EXO1-deficient cells are less prominent than those seen in cells lacking MSH6 or MLH1. Eukaryotic cells thus apparently possess additional mechanisms that compensate for the loss of EXO1.


Assuntos
Proteínas Aviárias/metabolismo , Reparo de Erro de Pareamento de DNA , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/metabolismo , Enzimas Multifuncionais/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Galinhas , Endodesoxirribonucleases/química , Exodesoxirribonucleases/química , Exodesoxirribonucleases/deficiência , Exodesoxirribonucleases/genética , Guanosina/análogos & derivados , Células HEK293 , Humanos , Metilnitronitrosoguanidina , Enzimas Multifuncionais/química , Mutação/genética , Tionucleosídeos
2.
J Biol Chem ; 293(37): 14384-14392, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30082315

RESUMO

Comprised of telomeric TTAGGG repeats and shelterin, telomeres ensure that the natural ends of chromosomes remain impervious to the DNA damage response. Telomeres carry a long constitutive 3' overhang that can bind replication protein A (RPA) and activate the ATR Ser/Thr kinase (ATR), which induces cell cycle arrest. A single-stranded (ss) TTAGGG repeat-binding protein in mouse shelterin, POT1a, has been proposed to repress ATR signaling by preventing RPA binding. Repression of ATR at telomeres requires tethering of POT1a to the other shelterin subunits situated on the double-stranded (ds) telomeric DNA. The simplest model of ATR repression, the "tethered exclusion model," suggests that the only critical features of POT1a are its connection to shelterin and its binding to ss telomeric DNA. In agreement with the model, we show here that a shelterin-tethered variant of RPA70 (lacking the ATR recruitment domain) can repress ATR signaling at telomeres that lack POT1a. However, arguing against the tethered exclusion model, the nearly identical POT1b subunit of shelterin has been shown to be much less proficient than POT1a in repression of ATR. We now show that POT1b has the intrinsic ability to fully repress ATR but is prevented from doing so when bound to Ctc1, Stn1, Ten1 (CST), the complex needed for telomere end processing. These results establish that shelterin represses ATR with a tethered ssDNA-binding domain that excludes RPA from the 3' overhang and also reveal an unexpected effect of CST on the ability of POT1b to repress ATR.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína de Replicação A/metabolismo , Transdução de Sinais , Telômero , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Células Cultivadas , DNA/metabolismo , Células HEK293 , Humanos , Camundongos , Modelos Teóricos , Ligação Proteica
3.
Cell Rep ; 18(1): 41-53, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28052260

RESUMO

Shelterin is a six-subunit protein complex that plays crucial roles in telomere length regulation, protection, and maintenance. Although several shelterin subunits have been studied in vitro, the biochemical properties of the fully assembled shelterin complex are not well defined. Here, we characterize shelterin using ensemble biochemical methods, electron microscopy, and single-molecule imaging to determine how shelterin recognizes and assembles onto telomeric repeats. We show that shelterin complexes can exist in solution and primarily locate telomeric DNA through a three-dimensional diffusive search. Shelterin can diffuse along non-telomeric DNA but is impeded by nucleosomes, arguing against extensive one-dimensional diffusion as a viable assembly mechanism. Our work supports a model in which individual shelterin complexes rapidly bind to telomeric repeats as independent functional units, which do not alter the DNA-binding mode of neighboring complexes but, rather, occupy telomeric DNA in a "beads on a string" configuration.


Assuntos
Mamíferos/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Animais , DNA/metabolismo , Difusão , Células HEK293 , Humanos , Cinética , Complexos Multiproteicos/metabolismo , Ligação Proteica , Sequências Repetitivas de Ácido Nucleico , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo
4.
PLoS One ; 10(3): e0118940, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25742417

RESUMO

African trypanosomes cause a parasitic disease known as sleeping sickness. Mitochondrial transcript maturation in these organisms requires a RNA editing reaction that is characterized by the insertion and deletion of U-nucleotides into otherwise non-functional mRNAs. Editing represents an ideal target for a parasite-specific therapeutic intervention since the reaction cycle is absent in the infected host. In addition, editing relies on a macromolecular protein complex, the editosome, that only exists in the parasite. Therefore, all attempts to search for editing interfering compounds have been focused on molecules that bind to proteins of the editing machinery. However, in analogy to other RNA-driven biochemical pathways it should be possible to stall the reaction by targeting its substrate RNAs. Here we demonstrate inhibition of editing by specific aminoglycosides. The molecules bind into the major groove of the gRNA/pre-mRNA editing substrates thereby causing a stabilization of the RNA molecules through charge compensation and an increase in stacking. The data shed light on mechanistic details of the editing process and identify critical parameters for the development of new trypanocidal compounds.


Assuntos
Edição de RNA , RNA de Protozoário/metabolismo , Trypanosoma/metabolismo , RNA de Protozoário/genética , Termodinâmica , Trypanosoma/genética
5.
DNA Repair (Amst) ; 24: 87-97, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25288561

RESUMO

RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.


Assuntos
Replicação do DNA , Proteínas de Ligação a DNA/genética , Sequência de Aminoácidos , Animais , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/efeitos da radiação , Galinhas , Cisplatino/farmacologia , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Técnicas de Inativação de Genes , Teste de Complementação Genética , Humanos , Hidroxiureia/farmacologia , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Radiação Ionizante , Vertebrados/genética
6.
PLoS One ; 7(2): e24555, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22383942

RESUMO

The small nematode Caenorhabditis elegans displays a spectrum of DNA damage responses similar to humans. In order to identify new DNA damage response genes, we isolated in a forward genetic screen 14 new mutations conferring hypersensitivity to ionizing radiation. We present here our characterization of lem-3, one of the genes identified in this screen. LEM-3 contains a LEM domain and a GIY nuclease domain. We confirm that LEM-3 has DNase activity in vitro. lem-3(lf) mutants are hypersensitive to various types of DNA damage, including ionizing radiation, UV-C light and crosslinking agents. Embryos from irradiated lem-3 hermaphrodites displayed severe defects during cell division, including chromosome mis-segregation and anaphase bridges. The mitotic defects observed in irradiated lem-3 mutant embryos are similar to those found in baf-1 (barrier-to-autointegration factor) mutants. The baf-1 gene codes for an essential and highly conserved protein known to interact with the other two C. elegans LEM domain proteins, LEM-2 and EMR-1. We show that baf-1, lem-2, and emr-1 mutants are also hypersensitive to DNA damage and that loss of lem-3 sensitizes baf-1 mutants even in the absence of DNA damage. Our data suggest that BAF-1, together with the LEM domain proteins, plays an important role following DNA damage - possibly by promoting the reorganization of damaged chromatin.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Dano ao DNA , Endodesoxirribonucleases/fisiologia , Alelos , Animais , Apoptose , Proteínas de Bactérias/metabolismo , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/química , Ciclo Celular , Proteínas de Ciclo Celular , Cromatina/química , Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Endodesoxirribonucleases/química , Metanossulfonato de Etila/farmacologia , Teste de Complementação Genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/química , Modelos Genéticos , Mutação , Proteínas Nucleares/química , Fenótipo , Estrutura Terciária de Proteína , Radiação Ionizante , Frações Subcelulares , Transgenes , Raios Ultravioleta
7.
Proc Natl Acad Sci U S A ; 107(50): 21553-7, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115814

RESUMO

Fanconi anemia (FA) is a rare genetic disease characterized by congenital defects, bone marrow failure, chromosomal instability, and cancer susceptibility. One hallmark of cells from FA patients is hypersensitivity to interstrand cross-linking agents, such as the chemotherapeutics cisplatin and mitomycin C (MMC). We have recently characterized a FANCD2/FANCI-associated nuclease, KIAA1018/FAN1, the depletion of which sensitizes human cells to these agents. However, as the down-regulation of FAN1 in human cells was mediated by siRNA and thus only transient, we were unable to study the long-term effects of FAN1 loss on chromosomal stability. We now describe the generation of chicken DT40 B cells, in which the FAN1 locus was disrupted by gene targeting. FAN1-null cells are highly sensitive to cisplatin and MMC, but not to ionizing or UV radiation, methyl methanesulfonate, or camptothecin. The cells do not display elevated sister chromatid exchange frequencies, either sporadic or MMC-induced. Interestingly, MMC treatment causes chromosomal instability that is quantitatively, but not qualitatively, comparable to that seen in FA cells. This finding, coupled with evidence showing that DT40 cells deficient in both FAN1 and FANCC, or FAN1 and FANCJ, exhibited increased sensitivity to cisplatin compared with cells lacking only FAN1, suggests that, despite its association with FANCD2/FANCI, FAN1 in DT40 cells participates in the processing of damage induced by interstrand cross-linking-generating agents also independently of the classical FA pathway.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , DNA/efeitos dos fármacos , Exodesoxirribonucleases/metabolismo , Instabilidade Genômica/efeitos dos fármacos , Animais , Linhagem Celular , Galinhas , Dano ao DNA , Reparo do DNA , Endodesoxirribonucleases , Exodesoxirribonucleases/genética , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Enzimas Multifuncionais
8.
Cell ; 142(1): 77-88, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20603016

RESUMO

Cytotoxicity of cisplatin and mitomycin C (MMC) is ascribed largely to their ability to generate interstrand crosslinks (ICLs) in DNA, which block the progression of replication forks. The processing of ICLs requires the Fanconi anemia (FA) pathway, excision repair, and translesion DNA synthesis (TLS). It also requires homologous recombination (HR), which repairs double-strand breaks (DSBs) generated by cleavage of the blocked replication forks. Here we describe KIAA1018, an evolutionarily conserved protein that has an N-terminal ubiquitin-binding zinc finger (UBZ) and a C-terminal nuclease domain. KIAA1018 is a 5'-->3' exonuclease and a structure-specific endonuclease that preferentially incises 5' flaps. Like cells from FA patients, human cells depleted of KIAA1018 are sensitized to ICL-inducing agents and display chromosomal instability. The link of KIAA1018 to the FA pathway is further strengthened by its recruitment to DNA damage through interaction of its UBZ domain with monoubiquitylated FANCD2. We therefore propose to name KIAA1018 FANCD2-associated nuclease, FAN1.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Reparo do DNA , Exodesoxirribonucleases/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Mitomicina/farmacologia , Sequência de Aminoácidos , Animais , Caenorhabditis elegans , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Dano ao DNA/efeitos dos fármacos , Endodesoxirribonucleases , Endonucleases/metabolismo , Exodesoxirribonucleases/química , Humanos , Dados de Sequência Molecular , Enzimas Multifuncionais , Fosfodiesterase I/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...