Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
G3 (Bethesda) ; 14(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38526099

RESUMEN

Cellular pathways that detect DNA damage are useful for identifying genes that suppress DNA damage, which can cause genome instability and cancer predisposition syndromes when mutated. We identified 199 high-confidence and 530 low-confidence DNA damage-suppressing (DDS) genes in Saccharomyces cerevisiae through a whole-genome screen for mutations inducing Hug1 expression, a focused screen for mutations inducing Ddc2 foci, and data from previous screens for mutations causing Rad52 foci accumulation and Rnr3 induction. We also identified 286 high-confidence and 394 low-confidence diverse genome instability-suppressing (DGIS) genes through a whole-genome screen for mutations resulting in increased gross chromosomal rearrangements and data from previous screens for mutations causing increased genome instability as assessed in a diversity of genome instability assays. Genes that suppress both pathways (DDS+ DGIS+) prevent or repair DNA replication damage and likely include genes preventing collisions between the replication and transcription machineries. DDS+ DGIS- genes, including many transcription-related genes, likely suppress damage that is normally repaired properly or prevent inappropriate signaling, whereas DDS- DGIS+ genes, like PIF1, do not suppress damage but likely promote its proper, nonmutagenic repair. Thus, induction of DNA damage markers is not a reliable indicator of increased genome instability, and the DDS and DGIS categories define mechanistically distinct groups of genes.


Asunto(s)
Daño del ADN , Inestabilidad Genómica , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Mutación , Genes Supresores , Reparación del ADN/genética , Genoma Fúngico
2.
Nat Commun ; 12(1): 5568, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34552065

RESUMEN

Eukaryotic DNA Mismatch Repair (MMR) involves redundant exonuclease 1 (Exo1)-dependent and Exo1-independent pathways, of which the Exo1-independent pathway(s) is not well understood. The exo1Δ440-702 mutation, which deletes the MutS Homolog 2 (Msh2) and MutL Homolog 1 (Mlh1) interacting peptides (SHIP and MIP boxes, respectively), eliminates the Exo1 MMR functions but is not lethal in combination with rad27Δ mutations. Analyzing the effect of different combinations of the exo1Δ440-702 mutation, a rad27Δ mutation and the pms1-A99V mutation, which inactivates an Exo1-independent MMR pathway, demonstrated that each of these mutations inactivates a different MMR pathway. Furthermore, it was possible to reconstitute a Rad27- and Msh2-Msh6-dependent MMR reaction in vitro using a mispaired DNA substrate and other MMR proteins. Our results demonstrate Rad27 defines an Exo1-independent eukaryotic MMR pathway that is redundant with at least two other MMR pathways.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Exodesoxirribonucleasas/metabolismo , Endonucleasas de ADN Solapado/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , ADN Ligasas/metabolismo , ADN de Hongos/metabolismo , Exodesoxirribonucleasas/genética , Endonucleasas de ADN Solapado/genética , Proteínas MutL/genética , Proteínas MutL/metabolismo , Mutación , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
3.
Elife ; 92020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32762846

RESUMEN

Foldback inversions, also called inverted duplications, have been observed in human genetic diseases and cancers. Here, we used a Saccharomyces cerevisiae genetic system that generates gross chromosomal rearrangements (GCRs) mediated by foldback inversions combined with whole-genome sequencing to study their formation. Foldback inversions were mediated by formation of single-stranded DNA hairpins. Two types of hairpins were identified: small-loop hairpins that were suppressed by MRE11, SAE2, SLX1, and YKU80 and large-loop hairpins that were suppressed by YEN1, TEL1, SWR1, and MRC1. Analysis of CRISPR/Cas9-induced double strand breaks (DSBs) revealed that long-stem hairpin-forming sequences could form foldback inversions when proximal or distal to the DSB, whereas short-stem hairpin-forming sequences formed foldback inversions when proximal to the DSB. Finally, we found that foldback inversion GCRs were stabilized by secondary rearrangements, mostly mediated by different homologous recombination mechanisms including single-strand annealing; however, POL32-dependent break-induced replication did not appear to be involved forming secondary rearrangements.


Asunto(s)
Inversión Cromosómica , Cromosomas Fúngicos/genética , Inestabilidad Genómica , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Roturas del ADN de Doble Cadena , Reordenamiento Génico , Genoma Fúngico , Recombinación Homóloga
4.
Nat Struct Mol Biol ; 26(12): 1184, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31686054

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nat Commun ; 9(1): 3680, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206225

RESUMEN

Genome instability is associated with tumorigenesis. Here, we identify a role for the histone Htz1, which is deposited by the Swr1 chromatin-remodeling complex (SWR-C), in preventing genome instability in the absence of the replication fork/replication checkpoint proteins Mrc1, Csm3, or Tof1. When combined with deletion of SWR1 or HTZ1, deletion of MRC1, CSM3, or TOF1 or a replication-defective mrc1 mutation causes synergistic increases in gross chromosomal rearrangement (GCR) rates, accumulation of a broad spectrum of GCRs, and hypersensitivity to replication stress. The double mutants have severe replication defects and accumulate aberrant replication intermediates. None of the individual mutations cause large increases in GCR rates; however, defects in MRC1, CSM3 or TOF1 cause activation of the DNA damage checkpoint and replication defects. We propose a model in which Htz1 deposition and retention in chromatin prevents transiently stalled replication forks that occur in mrc1, tof1, or csm3 mutants from being converted to DNA double-strand breaks that trigger genome instability.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina , Replicación del ADN , Inestabilidad Genómica , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Ciclo Celular , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Cromosomas Fúngicos/genética , Daño del ADN , Replicación del ADN/efectos de los fármacos , Reordenamiento Génico/genética , Recombinación Homóloga/genética , Hidroxiurea/farmacología , Modelos Biológicos , Mutación/genética , Saccharomyces cerevisiae/metabolismo , Estrés Fisiológico/efectos de los fármacos
6.
Nat Struct Mol Biol ; 25(8): 650-659, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30061603

RESUMEN

Eukaryotic DNA mismatch repair (MMR) involves both exonuclease 1 (Exo1)-dependent and Exo1-independent pathways. We found that the unstructured C-terminal domain of Saccharomyces cerevisiae Exo1 contains two MutS homolog 2 (Msh2)-interacting peptide (SHIP) boxes downstream from the MutL homolog 1 (Mlh1)-interacting peptide (MIP) box. These three sites were redundant in Exo1-dependent MMR in vivo and could be replaced by a fusion protein between an N-terminal fragment of Exo1 and Msh6. The SHIP-Msh2 interactions were eliminated by the msh2M470I mutation, and wild-type but not mutant SHIP peptides eliminated Exo1-dependent MMR in vitro. We identified two S. cerevisiae SHIP-box-containing proteins and three candidate human SHIP-box-containing proteins. One of these, Fun30, had a small role in Exo1-dependent MMR in vivo. The Remodeling of the Structure of Chromatin (Rsc) complex also functioned in both Exo1-dependent and Exo1-independent MMR in vivo. Our results identified two modes of Exo1 recruitment and a peptide module that mediates interactions between Msh2 and other proteins, and they support a model in which Exo1 functions in MMR by being tethered to the Msh2-Msh6 complex.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Dominios y Motivos de Interacción de Proteínas , Secuencia de Aminoácidos , Secuencia Conservada , Humanos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología de Secuencia de Aminoácido
7.
PLoS Genet ; 14(3): e1007250, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29505562

RESUMEN

Mms21, a subunit of the Smc5/6 complex, possesses an E3 ligase activity for the Small Ubiquitin-like MOdifier (SUMO). Here we show that the mms21-CH mutation, which inactivates Mms21 ligase activity, causes increased accumulation of gross chromosomal rearrangements (GCRs) selected in the dGCR assay. These dGCRs are formed by non-allelic homologous recombination between divergent DNA sequences mediated by Rad52-, Rrm3- and Pol32-dependent break-induced replication. Combining mms21-CH with sgs1Δ caused a synergistic increase in GCRs rates, indicating the distinct roles of Mms21 and Sgs1 in suppressing GCRs. The mms21-CH mutation also caused increased rates of accumulating uGCRs mediated by breakpoints in unique sequences as revealed by whole genome sequencing. Consistent with the accumulation of endogenous DNA lesions, mms21-CH mutants accumulate increased levels of spontaneous Rad52 and Ddc2 foci and had a hyper-activated DNA damage checkpoint. Together, these findings support that Mms21 prevents the accumulation of spontaneous DNA lesions that cause diverse GCRs.


Asunto(s)
Daño del ADN/genética , Proteína SUMO-1/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Cromosomas Fúngicos , Reparación del ADN , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Epistasis Genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Genoma Fúngico , Mutación , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Proteína SUMO-1/genética , Proteínas de Saccharomyces cerevisiae/genética
8.
PLoS Genet ; 14(1): e1007170, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29320491

RESUMEN

Defects in the genes encoding the Paf1 complex can cause increased genome instability. Loss of Paf1, Cdc73, and Ctr9, but not Rtf1 or Leo1, caused increased accumulation of gross chromosomal rearrangements (GCRs). Combining the cdc73Δ mutation with individual deletions of 43 other genes, including TEL1 and YKU80, which are involved in telomere maintenance, resulted in synergistic increases in GCR rates. Whole genome sequence analysis of GCRs indicated that there were reduced relative rates of GCRs mediated by de novo telomere additions and increased rates of translocations and inverted duplications in cdc73Δ single and double mutants. Analysis of telomere lengths and telomeric gene silencing in strains containing different combinations of cdc73Δ, tel1Δ and yku80Δ mutations suggested that combinations of these mutations caused increased defects in telomere maintenance. A deletion analysis of Cdc73 revealed that a central 105 amino acid region was necessary and sufficient for suppressing the defects observed in cdc73Δ strains; this region was required for the binding of Cdc73 to the Paf1 complex through Ctr9 and for nuclear localization of Cdc73. Taken together, these data suggest that the increased GCR rate of cdc73Δ single and double mutants is due to partial telomere dysfunction and that Ctr9 and Paf1 play a central role in the Paf1 complex potentially by scaffolding the Paf1 complex subunits or by mediating recruitment of the Paf1 complex to the different processes it functions in.


Asunto(s)
Inestabilidad Genómica/genética , Proteínas Nucleares/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Homeostasis del Telómero/genética , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/genética , Organismos Modificados Genéticamente , Fenotipo , Unión Proteica , Proteínas Serina-Treonina Quinasas/fisiología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Telómero/metabolismo , Factores de Elongación Transcripcional/genética
9.
Pol J Radiol ; 82: 152-160, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28392852

RESUMEN

BACKGROUND: A systematic review and meta-analysis of all available publications was performed to evaluate the diagnostic accuracy of percutaneous transthoracic needle biopsy (PTNB) using a C-Arm Cone-Beam CT (CBCT) system in patients with lung nodules. MATERIAL/METHODS: Thedatabases of PUBMED, OVID, EBSCO, EMBASE, and China National Knowledge Infrastructure (CNKI) were systematically searched for relevant original articles on the diagnostic accuracy of CBCT-guided PTNB for the diagnosis of nodules in the lungs. Diagnostic indices including sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR) and diagnostic score (DS) were calculated. Moreover,summary receiver operating characteristic curves (SROC) were constructed with Stata (version 13.0), Rev Man (version 5.3), and Meta-disc (version 1.4) software. Other clinical indices such as incidence of complications were also recorded. RESULTS: Eight studies met the inclusion and exclusion criteria for the meta-analysis. The pooled sensitivity, specificity, PLR, NLR, DOR, DS, and SROC with 95% confidence intervals were 0.96 (0.93-0.98), 1.00 (0.91-1.00), 711.15 (9.48-53325.89), 0.04 (0.02-0.07), 16585.29 (284.88-9.7e+05), 9.72 (5.65-13.78), and 0.99 (0.97-0.99), respectively. The incidence of pneumothorax and hemorrhage was 10-29.27% and 1.22-47.25%, respectively. CONCLUSIONS: CBCT-guided PTNB has an acceptable rate of complications and is associated with a reasonable radiation exposure. Moreover, it is a highly accurate and safe technique for the diagnosis of lung nodules and can be recommended to be used in routine clinical practice.

10.
Nucleic Acids Res ; 43(8): 3986-97, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25845601

RESUMEN

Growth arrest and DNA-damage-inducible protein 45 (Gadd45) family members have been implicated in DNA demethylation in vertebrates. However, it remained unclear how they contribute to the demethylation process. Here, we demonstrate that Gadd45a promotes active DNA demethylation through thymine DNA glycosylase (TDG) which has recently been shown to excise 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) generated in Ten-eleven-translocation (Tet)-initiated oxidative demethylation. The connection of Gadd45a with oxidative demethylation is evidenced by the enhanced activation of a methylated reporter gene in HEK293T cells expressing Gadd45a in combination with catalytically active TDG and Tet. Gadd45a interacts with TDG physically and increases the removal of 5fC and 5caC from genomic and transfected plasmid DNA by TDG. Knockout of both Gadd45a and Gadd45b from mouse ES cells leads to hypermethylation of specific genomic loci most of which are also targets of TDG and show 5fC enrichment in TDG-deficient cells. These observations indicate that the demethylation effect of Gadd45a is mediated by TDG activity. This finding thus unites Gadd45a with the recently defined Tet-initiated demethylation pathway.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Nucleares/fisiología , Timina ADN Glicosilasa/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Citosina/análogos & derivados , Citosina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Células Madre Embrionarias/metabolismo , Células HEK293 , Humanos , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/metabolismo , Activación Transcripcional
11.
Science ; 333(6047): 1303-7, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21817016

RESUMEN

The prevalent DNA modification in higher organisms is the methylation of cytosine to 5-methylcytosine (5mC), which is partially converted to 5-hydroxymethylcytosine (5hmC) by the Tet (ten eleven translocation) family of dioxygenases. Despite their importance in epigenetic regulation, it is unclear how these cytosine modifications are reversed. Here, we demonstrate that 5mC and 5hmC in DNA are oxidized to 5-carboxylcytosine (5caC) by Tet dioxygenases in vitro and in cultured cells. 5caC is specifically recognized and excised by thymine-DNA glycosylase (TDG). Depletion of TDG in mouse embyronic stem cells leads to accumulation of 5caC to a readily detectable level. These data suggest that oxidation of 5mC by Tet proteins followed by TDG-mediated base excision of 5caC constitutes a pathway for active DNA demethylation.


Asunto(s)
Citosina/análogos & derivados , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Timina ADN Glicosilasa/metabolismo , 5-Metilcitosina/metabolismo , Animales , Línea Celular , Citosina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/genética , Dioxigenasas , Células Madre Embrionarias , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Oxidación-Reducción , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño , Timina ADN Glicosilasa/genética , Transfección
12.
Cell Res ; 21(8): 1172-81, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21606950

RESUMEN

Cytosine methylation of genomic DNA controls gene expression and maintains genome stability. How a specific DNA sequence is targeted for methylation by a methyltransferase is largely unknown. Here, we show that histone H3 tails lacking lysine 4 (K4) methylation function as an allosteric activator for methyltransferase Dnmt3a by binding to its plant homeodomain (PHD). In vitro, histone H3 peptides stimulated the methylation activity of Dnmt3a up to 8-fold, in a manner reversely correlated with the level of K4 methylation. The biological significance of allosteric regulation was manifested by molecular modeling and identification of key residues in both the PHD and the catalytic domain of Dnmt3a whose mutations impaired the stimulation of methylation activity by H3 peptides but not the binding of H3 peptides. Significantly, these mutant Dnmt3a proteins were almost inactive in DNA methylation when expressed in mouse embryonic stem cells while their recruitment to genomic targets was unaltered. We therefore propose a two-step mechanism for de novo DNA methylation - first recruitment of the methyltransferase probably assisted by a chromatin- or DNA-binding factor, and then allosteric activation depending on the interaction between Dnmt3a and the histone tails - the latter might serve as a checkpoint for the methylation activity.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Histonas/metabolismo , Regulación Alostérica , Animales , Línea Celular , Inmunoprecipitación de Cromatina , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Activación Enzimática , Histonas/química , Humanos , Ratones , Mutación , Unión Proteica , Estructura Terciaria de Proteína
13.
Mol Cell Biol ; 27(24): 8748-59, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17938196

RESUMEN

DNA methylation plays an important role in gene silencing in mammals. Two de novo methyltransferases, Dnmt3a and Dnmt3b, are required for the establishment of genomic methylation patterns in development. However, little is known about their coordinate function in the silencing of genes critical for embryonic development and how their activity is regulated. Here we show that Dnmt3a and Dnmt3b are the major components of a native complex purified from embryonic stem cells. The two enzymes directly interact and mutually stimulate each other both in vitro and in vivo. The stimulatory effect is independent of the catalytic activity of the enzyme. In differentiating embryonic carcinoma or embryonic stem cells and mouse postimplantation embryos, they function synergistically to methylate the promoters of the Oct4 and Nanog genes. Inadequate methylation caused by ablating Dnmt3a and Dnmt3b is associated with dysregulated expression of Oct4 and Nanog during the differentiation of pluripotent cells and mouse embryonic development. These results suggest that Dnmt3a and Dnmt3b form a complex through direct contact in living cells and cooperate in the methylation of the promoters of Oct4 and Nanog during cell differentiation. The physical and functional interaction between Dnmt3a and Dnmt3b represents a novel regulatory mechanism to ensure the proper establishment of genomic methylation patterns for gene silencing in development.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cromatografía de Afinidad , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/aislamiento & purificación , Metilación de ADN/efectos de los fármacos , ADN Metiltransferasa 3A , Desarrollo Embrionario/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/metabolismo , Ratones , Proteína Homeótica Nanog , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Tretinoina/farmacología , ADN Metiltransferasa 3B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...