Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
PLoS Genet ; 20(5): e1011148, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38776358

RESUMO

The helicase MCM and the ribonucleotide reductase RNR are the complexes that provide the substrates (ssDNA templates and dNTPs, respectively) for DNA replication. Here, we demonstrate that MCM interacts physically with RNR and some of its regulators, including the kinase Dun1. These physical interactions encompass small subpopulations of MCM and RNR, are independent of the major subcellular locations of these two complexes, augment in response to DNA damage and, in the case of the Rnr2 and Rnr4 subunits of RNR, depend on Dun1. Partial disruption of the MCM/RNR interactions impairs the release of Rad52 -but not RPA-from the DNA repair centers despite the lesions are repaired, a phenotype that is associated with hypermutagenesis but not with alterations in the levels of dNTPs. These results suggest that a specifically regulated pool of MCM and RNR complexes plays non-canonical roles in genetic stability preventing persistent Rad52 centers and hypermutagenesis.


Assuntos
Proteínas de Ciclo Celular , Dano ao DNA , Reparo do DNA , Replicação do DNA , Instabilidade Genômica , Proteína Rad52 de Recombinação e Reparo de DNA , Ribonucleotídeo Redutases , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Replicação do DNA/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Dano ao DNA/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Reparo do DNA/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteínas de Manutenção de Minicromossomo/genética , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , Ribonucleosídeo Difosfato Redutase/genética , Ribonucleosídeo Difosfato Redutase/metabolismo
2.
EMBO Rep ; 22(1): e50410, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33289333

RESUMO

DNA damage tolerance relies on homologous recombination (HR) and translesion synthesis (TLS) mechanisms to fill in the ssDNA gaps generated during passing of the replication fork over DNA lesions in the template. Whereas TLS requires specialized polymerases able to incorporate a dNTP opposite the lesion and is error-prone, HR uses the sister chromatid and is mostly error-free. We report that the HR protein Rad52-but not Rad51 and Rad57-acts in concert with the TLS machinery (Rad6/Rad18-mediated PCNA ubiquitylation and polymerases Rev1/Pol ζ) to repair MMS and UV light-induced ssDNA gaps through a non-recombinogenic mechanism, as inferred from the different phenotypes displayed in the absence of Rad52 and Rad54 (essential for MMS- and UV-induced HR); accordingly, Rad52 is required for efficient DNA damage-induced mutagenesis. In addition, Rad52, Rad51, and Rad57, but not Rad54, facilitate Rad6/Rad18 binding to chromatin and subsequent DNA damage-induced PCNA ubiquitylation. Therefore, Rad52 facilitates the tolerance process not only by HR but also by TLS through Rad51/Rad57-dependent and -independent processes, providing a novel role for the recombination proteins in maintaining genome integrity.


Assuntos
Dano ao DNA , Reparo do DNA , Replicação do DNA , Proteína Rad52 de Recombinação e Reparo de DNA , DNA de Cadeia Simples/genética , DNA Polimerase Dirigida por DNA/genética
3.
PLoS Genet ; 16(12): e1008603, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370275

RESUMO

Telomeres have the ability to adopt a lariat conformation and hence, engage in long and short distance intra-chromosome interactions. Budding yeast telomeres were proposed to fold back into subtelomeric regions, but a robust assay to quantitatively characterize this structure has been lacking. Therefore, it is not well understood how the interactions between telomeres and non-telomeric regions are established and regulated. We employ a telomere chromosome conformation capture (Telo-3C) approach to directly analyze telomere folding and its maintenance in S. cerevisiae. We identify the histone modifiers Sir2, Sin3 and Set2 as critical regulators for telomere folding, which suggests that a distinct telomeric chromatin environment is a major requirement for the folding of yeast telomeres. We demonstrate that telomeres are not folded when cells enter replicative senescence, which occurs independently of short telomere length. Indeed, Sir2, Sin3 and Set2 protein levels are decreased during senescence and their absence may thereby prevent telomere folding. Additionally, we show that the homologous recombination machinery, including the Rad51 and Rad52 proteins, as well as the checkpoint component Rad53 are essential for establishing the telomere fold-back structure. This study outlines a method to interrogate telomere-subtelomere interactions at a single unmodified yeast telomere. Using this method, we provide insights into how the spatial arrangement of the chromosome end structure is established and demonstrate that telomere folding is compromised throughout replicative senescence.


Assuntos
Replicação do DNA , Histona Desacetilases/metabolismo , Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/metabolismo , Telômero/genética , Histona Desacetilases/genética , Metiltransferases/genética , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteínas Repressoras/genética , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Sirtuína 2/genética , Telômero/química , Homeostase do Telômero
4.
PLoS Genet ; 14(6): e1007407, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29879139

RESUMO

Upon telomerase inactivation, telomeres gradually shorten with each cell division until cells enter replicative senescence. In Saccharomyces cerevisiae, the kinases Mec1/ATR and Tel1/ATM protect the genome during pre-senescence by preventing telomere-telomere fusions (T-TFs) and the subsequent genetic instability associated with fusion-bridge-breakage cycles. Here we report that T-TFs in mec1Δ tel1Δ cells can be suppressed by reducing the pool of available histones. This protection associates neither with changes in bulk telomere length nor with major changes in the structure of subtelomeric chromatin. We show that the absence of Mec1 and Tel1 strongly augments double-strand break (DSB) repair by non-homologous end joining (NHEJ), which might contribute to the high frequency of T-TFs in mec1Δ tel1Δ cells. However, histone depletion does not prevent telomere fusions by inhibiting NHEJ, which is actually increased in histone-depleted cells. Rather, histone depletion protects telomeres from fusions by homologous recombination (HR), even though HR is proficient in maintaining the proliferative state of pre-senescent mec1Δ tel1Δ cells. Therefore, HR during pre-senescence not only helps stalled replication forks but also prevents T-TFs by a mechanism that, in contrast to the previous one, is promoted by a reduction in the histone pool and can occur in the absence of Rad51. Our results further suggest that the Mec1-dependent depletion of histones that occurs during pre-senescence in cells without telomerase (tlc1Δ) prevents T-TFs by favoring the processing of unprotected telomeres by Rad51-independent HR.


Assuntos
Senescência Celular/genética , Histonas/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiologia , Telômero/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Reparo de DNA por Recombinação/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Telomerase/genética , Telomerase/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(48): 14840-5, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26578803

RESUMO

RNA polymerase II (RNAPII) transcription elongation is a highly regulated process that greatly influences mRNA levels as well as pre-mRNA splicing. Despite many studies in vitro, how chromatin modulates RNAPII elongation in vivo is still unclear. Here, we show that a decrease in the level of available canonical histones leads to more accessible chromatin with decreased levels of canonical histones and variants H2A.X and H2A.Z and increased levels of H3.3. With this altered chromatin structure, the RNAPII elongation rate increases, and the kinetics of pre-mRNA splicing is delayed with respect to RNAPII elongation. Consistent with the kinetic model of cotranscriptional splicing, the rapid RNAPII elongation induced by histone depletion promotes the skipping of variable exons in the CD44 gene. Indeed, a slowly elongating mutant of RNAPII was able to rescue this defect, indicating that the defective splicing induced by histone depletion is a direct consequence of the increased elongation rate. In addition, genome-wide analysis evidenced that histone reduction promotes widespread alterations in pre-mRNA processing, including intron retention and changes in alternative splicing. Our data demonstrate that pre-mRNA splicing may be regulated by chromatin structure through the modulation of the RNAPII elongation rate.


Assuntos
Histonas/metabolismo , RNA Polimerase II/metabolismo , Precursores de RNA/biossíntese , Splicing de RNA/fisiologia , Elongação da Transcrição Genética/fisiologia , Linhagem Celular Tumoral , Histonas/genética , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , RNA Polimerase II/genética , Precursores de RNA/genética
6.
EMBO J ; 32(9): 1307-21, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23563117

RESUMO

Homologous recombination (HR) is essential for genome integrity. Recombination proteins participate in tolerating DNA lesions that interfere with DNA replication, but can also generate toxic recombination intermediates and genetic instability when they are not properly regulated. Here, we have studied the role of the recombination proteins Rad51 and Rad52 at replication forks and replicative DNA lesions. We show that Rad52 loads Rad51 onto unperturbed replication forks, where they facilitate replication of alkylated DNA by non-repair functions. The recruitment of Rad52 and Rad51 to chromatin during DNA replication is a prerequisite for the repair of the non-DSB DNA lesions, presumably single-stranded DNA gaps, which are generated during the replication of alkylated DNA. We also show that the repair of these lesions requires CDK1 and is not coupled to the fork but rather restricted to G2/M by the replicative checkpoint. We propose a new scenario for HR where Rad52 and Rad51 are recruited to the fork to promote DNA damage tolerance by distinct and cell cycle-regulated replicative and repair functions.


Assuntos
Dano ao DNA , Replicação do DNA , DNA de Cadeia Simples/metabolismo , Rad51 Recombinase/metabolismo , Rad51 Recombinase/fisiologia , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Reparo do DNA/genética , Reparo do DNA/fisiologia , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Endodesoxirribonucleases/fisiologia , Metanossulfonato de Metila/farmacologia , Nuclease do Micrococo/metabolismo , Modelos Biológicos , Ligação Proteica/fisiologia , Rad51 Recombinase/genética , Recombinação Genética/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiologia , Proteínas de Saccharomyces cerevisiae/genética
7.
RNA Biol ; 14(3): 281-286, 2017 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-27211514

RESUMO

Histone proteins are main structural components of the chromatin and major determinants of gene regulation. Expression of canonical histone genes is strictly controlled during the cell cycle in order to couple DNA replication with histone deposition. Indeed, reductions in the levels of canonical histones or defects in chromatin assembly cause genetic instability. Early data from yeast demonstrated that severe histone depletion also causes strong gene expression changes. We have recently reported that a moderated depletion of canonical histones in human cells leads to an open chromatin configuration, which in turn increases RNA polymerase II elongation rates and causes pre-mRNA splicing defects. Interestingly, some of the observed defects accompany the scheduled histone depletion that is associated with several senescence and aging processes. Thus, our comparison of induced and naturally-occurring histone depletion processes suggests that a programmed reduction of the level of canonical histones might be a strategy to control gene expression during specific physiological processes.


Assuntos
Regulação da Expressão Gênica , Histonas/metabolismo , Animais , Ciclo Celular/genética , Senescência Celular/genética , Humanos , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , Transcrição Gênica
8.
Bioessays ; 36(5): 451-62, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24615940

RESUMO

Homologous recombination (HR) is required to protect and restart stressed replication forks. Paradoxically, the Mrc1 branch of the S phase checkpoints, which is activated by replicative stress, prevents HR repair at breaks and arrested forks. Indeed, the mechanisms underlying HR can threaten genome integrity if not properly regulated. Thus, understanding how cells avoid genetic instability associated with replicative stress, a hallmark of cancer, is still a challenge. Here I discuss recent results that support a model by which HR responds to replication stress through replicative and repair activities that operate at different stages of the cell cycle (S and G2, respectively) and in distinct subnuclear structures. Remarkably, the replication checkpoint appears to control this scenario by inhibiting the assembly of HR repair centers at stressed forks during S phase, thereby avoiding genetic instability.


Assuntos
Ciclo Celular/genética , Senescência Celular/genética , Reparo do DNA , Replicação do DNA , Instabilidade Genômica , Recombinação Homóloga/genética , Receptores Imunológicos/metabolismo , Compartimento Celular , Núcleo Celular/genética , Fase G2/genética , Humanos , Glicoproteínas de Membrana , Fase S/genética , Fatores de Tempo
9.
Nucleic Acids Res ; 42(20): 12469-82, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25300489

RESUMO

The structural organization of chromosomes is essential for their correct function and dynamics during the cell cycle. The assembly of DNA into chromatin provides the substrate for topoisomerases and condensins, which introduce the different levels of superhelical torsion required for DNA metabolism. In particular, Top2 and condensin are directly involved in both the resolution of precatenanes that form during replication and the formation of the intramolecular loop that detects tension at the centromeric chromatin during chromosome biorientation. Here we show that histone depletion activates the spindle assembly checkpoint (SAC) and impairs sister chromatid decatenation, leading to chromosome mis-segregation and lethality in the absence of the SAC. We demonstrate that histone depletion impairs chromosome biorientation and activates the Aurora-dependent pathway, which detects tension problems at the kinetochore. Interestingly, SAC activation is suppressed by the absence of Top2 and Smc2, an essential component of condensin. Indeed, smc2-8 suppresses catenanes accumulation, mitotic arrest and growth defects induced by histone depletion at semi-permissive temperature. Remarkably, SAC activation by histone depletion is associated with condensin-mediated alterations of the centromeric chromatin. Therefore, our results reveal the importance of a precise interplay between histone supply and condensin/Top2 for pericentric chromatin structure, precatenanes resolution and centromere biorientation.


Assuntos
Adenosina Trifosfatases/fisiologia , Cromatina/química , Cromossomos Fúngicos/química , Proteínas de Ligação a DNA/fisiologia , Histonas/fisiologia , Pontos de Checagem da Fase M do Ciclo Celular , Complexos Multiproteicos/fisiologia , Aurora Quinases/fisiologia , Centrômero/fisiologia , Segregação de Cromossomos , DNA Topoisomerases Tipo II/fisiologia , Metáfase , Nucleossomos/fisiologia , Pontos de Checagem da Fase S do Ciclo Celular , Proteínas de Saccharomyces cerevisiae/fisiologia
10.
PLoS Genet ; 7(11): e1002376, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22102830

RESUMO

Chromatin assembly mutants accumulate recombinogenic DNA damage and are sensitive to genotoxic agents. Here we have analyzed why impairment of the H3K56 acetylation-dependent CAF1 and Rtt106 chromatin assembly pathways, which have redundant roles in H3/H4 deposition during DNA replication, leads to genetic instability. We show that the absence of H3K56 acetylation or the simultaneous knock out of CAF1 and Rtt106 increases homologous recombination by affecting the integrity of advancing replication forks, while they have a minor effect on stalled replication fork stability in response to the replication inhibitor hydroxyurea. This defect in replication fork integrity is not due to defective checkpoints. In contrast, H3K56 acetylation protects against replicative DNA damaging agents by DNA repair/tolerance mechanisms that do not require CAF1/Rtt106 and are likely subsequent to the process of replication-coupled nucleosome deposition. We propose that the tight connection between DNA synthesis and histone deposition during DNA replication mediated by H3K56ac/CAF1/Rtt106 provides a mechanism for the stabilization of advancing replication forks and the maintenance of genome integrity, while H3K56 acetylation has an additional, CAF1/Rtt106-independent function in the response to replicative DNA damage.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Histonas/metabolismo , Lisina/metabolismo , Chaperonas Moleculares/genética , Nucleossomos/metabolismo , Ribonucleases/genética , Ribonucleases/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Acetilação , DNA/biossíntese , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histonas/genética , Recombinação Homóloga/genética , Chaperonas Moleculares/metabolismo , Mutação , Nucleossomos/genética , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais
11.
bioRxiv ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38915629

RESUMO

Stalled replication forks can be processed by several distinct mechanisms collectively called post-replication repair which includes homologous recombination, fork regression, and translesion DNA synthesis. However, the regulation of the usage between these pathways is not fully understood. The Rad51 protein plays a pivotal role in maintaining genomic stability through its roles in HR and in protecting stalled replication forks from degradation. We report the isolation of separation-of-function mutations in Saccharomyces cerevisiae Rad51 that retain their recombination function but display a defect in fork protection leading to a shift in post-replication repair pathway usage from HR to alternate pathways including mutagenic translesion synthesis. Rad51-E135D and Rad51-K305N show normal in vivo and in vitro recombination despite changes in their DNA binding profiles, in particular to dsDNA, with a resulting effect on their ATPase activities. The mutants lead to a defect in Rad51 recruitment to stalled forks in vivo as well as a defect in the protection of dsDNA from degradation by Dna2-Sgs1 and Exo1 in vitro . A high-resolution cryo-electron microscopy structure of the Rad51-ssDNA filament at 2.4 Å resolution provides a structural basis for a mechanistic understanding of the mutant phenotypes. Together, the evidence suggests a model in which Rad51 binding to duplex DNA is critical to control pathway usage at stalled replication forks.

13.
FEBS J ; 290(12): 3076-3088, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35398961

RESUMO

Cdc7 and its regulator Dbf4 (Dbf4-dependent kinase; DDK) form an essential complex due to its function in replication initiation, which is carried out by phosphorylating different residues at the helicase MCM during the G1/S transition. In response to replication stress, late origins are inhibited to prevent cell cycle progression until the problems are resolved. In yeast, this inhibition is partially achieved by attenuating DDK activity. In addition, evidence from yeast to human shows that Cdc7 is required for a successful DNA damage response by coordinating multiple processes dealing with replication stress (replication checkpoint, DNA damage tolerance and break-induced replication) through mechanisms that go beyond its role in origin activation. These studies reveal the importance of getting a better understanding of the spatiotemporal regulation of DDK. Here, we will discuss how DDK operates in these processes and its putative role in controlling the activity of replication and repair factors at specific nuclease-resistant nucleoprotein scaffolds.


Assuntos
Proteínas Serina-Treonina Quinases , Proteínas de Saccharomyces cerevisiae , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Replicação do DNA , Fosforilação
14.
Cell Rep ; 42(3): 112174, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36862554

RESUMO

The advance and stability of replication forks rely on a tight co-regulation of DNA synthesis and nucleosome assembly. We show that mutants affected in parental histone recycling are impaired in the recombinational repair of the single-stranded DNA gaps generated in response to DNA adducts that hamper replication, which are then filled in by translesion synthesis. These recombination defects are in part due to an excess of parental nucleosomes at the invaded strand that destabilizes the sister chromatid junction formed after strand invasion through a Srs2-dependent mechanism. In addition, we show that a dCas9∗/R-loop is more recombinogenic when the dCas9∗/DNA-RNA hybrid interferes with the lagging than with the leading strand, and this recombination is particularly sensitive to problems in the deposition of parental histones at the strand that contains the hindrance. Therefore, parental histone distribution and location of the replication obstacle at the lagging or leading strand regulate homologous recombination.


Assuntos
Replicação do DNA , Histonas , Histonas/metabolismo , Recombinação Homóloga , Nucleossomos , Reparo do DNA , DNA
15.
Sci Rep ; 13(1): 11397, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37452085

RESUMO

Genome duplication occurs through the coordinated action of DNA replication and nucleosome assembly at replication forks. Defective nucleosome assembly causes DNA lesions by fork breakage that need to be repaired. In addition, it causes a loss of chromatin integrity. These chromatin alterations can be restored, even though the mechanisms are unknown. Here, we show that the process of chromatin restoration can deal with highly severe chromatin defects induced by the absence of the chaperones CAF1 and Rtt106 or a strong reduction in the pool of available histones, and that this process can be followed by analyzing the topoisomer distribution of the 2µ plasmid. Using this assay, we demonstrate that chromatin restoration is slow and independent of checkpoint activation, whereas it requires the action of transcription and the FACT complex. Therefore, cells are able to "repair" not only DNA lesions but also chromatin alterations associated with defective nucleosome assembly.


Assuntos
Replicação do DNA , Nucleossomos , Nucleossomos/genética , Montagem e Desmontagem da Cromatina , Cromatina/genética , DNA
16.
Artigo em Inglês | MEDLINE | ID: mdl-36981853

RESUMO

International students face many impediments under the COVID-19 pandemic. The objectives of this study are to assess the association between the perceptions of international students and the lockdown policy for COVID-19. In 2021, three different levels of lockdown policy were enforced, including level I from January to April, level III from May to July, and level II from August to December. We conducted three surveys for international graduate students using a validated questionnaire during the different lockdown levels. We collected 185, 119, and 83 valid questionnaires in level I, II, and III, respectively. There were linear trends in the correlations of lockdown policy with the knowledge (p = 0.052), attitudes (p = 0.002), and practices (p < 0.001) of COVID-19. In brief, the stricter the lockdown policy, the better the students adhered to sufficient knowledge, positive attitudes, and healthy practices. Furthermore, there were significant linear correlations of lockdown policy with the transportation, school study, leisure, family life, and diet behavior. In conclusion, lockdown policy had important impacts on the knowledge, attitudes, practices, and daily lives of international students. The findings indicated that the lockdown system and its corresponding measures appear to affect perceptions in a positive way.


Assuntos
COVID-19 , Humanos , COVID-19/epidemiologia , Pandemias , Taiwan/epidemiologia , Controle de Doenças Transmissíveis , Estudantes , Inquéritos e Questionários , Conhecimentos, Atitudes e Prática em Saúde
17.
EMBO Rep ; 10(7): 790-6, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19465889

RESUMO

During DNA replication, the advance of replication forks is tightly connected with chromatin assembly, a process that can be impaired by the partial depletion of histone H4 leading to recombinogenic DNA damage. Here, we show that the partial depletion of H4 is rapidly followed by the collapse of unperturbed and stalled replication forks, even though the S-phase checkpoints remain functional. This collapse is characterized by a reduction in the amount of replication intermediates, but an increase in single Ys relative to bubbles, defects in the integrity of the replisome and an accumulation of DNA double-strand breaks. This collapse is also associated with an accumulation of Rad52-dependent X-shaped molecules. Consistently, a Rad52-dependent--although Rad51-independent--mechanism is able to rescue these broken replication forks. Our findings reveal that correct nucleosome deposition is required for replication fork stability, and provide molecular evidence for homologous recombination as an efficient mechanism of replication fork restart.


Assuntos
Montagem e Desmontagem da Cromatina , Replicação do DNA , Saccharomyces cerevisiae/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Histonas/metabolismo , Complexos Multienzimáticos/metabolismo , Recombinação Genética/genética , Fase S , Saccharomyces cerevisiae/citologia
18.
Genes (Basel) ; 12(10)2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34680945

RESUMO

The DNA damage tolerance (DDT) response is aimed to timely and safely complete DNA replication by facilitating the advance of replication forks through blocking lesions. This process is associated with an accumulation of single-strand DNA (ssDNA), both at the fork and behind the fork. Lesion bypass and ssDNA filling can be performed by translation synthesis (TLS) and template switching mechanisms. TLS uses low-fidelity polymerases to incorporate a dNTP opposite the blocking lesion, whereas template switching uses a Rad51/ssDNA nucleofilament and the sister chromatid to bypass the lesion. Rad51 is loaded at this nucleofilament by two mediator proteins, BRCA2 and Rad52, and these three factors are critical for homologous recombination (HR). Here, we review recent advances showing that Rad51, BRCA2, and Rad52 perform some of these functions through mechanisms that do not require the strand exchange activity of Rad51: the formation and protection of reversed fork structures aimed to bypass blocking lesions, and the promotion of TLS. These findings point to the central HR proteins as potential molecular switches in the choice of the mechanism of DDT.


Assuntos
Proteína BRCA1/metabolismo , Reparo do DNA , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Animais , Proteína BRCA1/genética , Dano ao DNA , Humanos , Rad51 Recombinase/genética , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Recombinação Genética
19.
Methods Mol Biol ; 2153: 447-458, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32840798

RESUMO

Homologous recombination (HR) has been extensively studied in response to DNA double-strand breaks (DSBs). In contrast, much less is known about how HR deals with DNA lesions other than DSBs (e.g., at single-stranded DNA) and replication forks, despite the fact that these DNA structures are associated with most spontaneous recombination events. A major handicap for studying the role of HR at non-DSB DNA lesions and replication forks is the difficulty of discriminating whether a recombination protein is associated with the non-DSB lesion per se or rather with a DSB generated during their processing. Here, we describe a method to follow the in vivo binding of recombination proteins to non-DSB DNA lesions and replication forks. This approach is based on the cleavage and subsequent electrophoretic analysis of the target DNA by the recombination protein fused to the micrococcal nuclease.


Assuntos
Quebras de DNA , Desoxirribonucleases/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Bactérias/metabolismo , Cromatina/genética , Cromatina/metabolismo , Replicação do DNA , Eletroforese em Gel Bidimensional , Recombinação Homóloga , Micrococcus/enzimologia , Saccharomyces cerevisiae/genética
20.
Cell Rep ; 36(4): 109440, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320356

RESUMO

The minichromosome maintenance (MCM) helicase physically interacts with the recombination proteins Rad51 and Rad52 from yeast to human cells. We show, in Saccharomyces cerevisiae, that these interactions occur within a nuclease-insoluble scaffold enriched in replication/repair factors. Rad51 accumulates in a MCM- and DNA-binding-independent manner and interacts with MCM helicases located outside of the replication origins and forks. MCM, Rad51, and Rad52 accumulate in this scaffold in G1 and are released during the S phase. In the presence of replication-blocking lesions, Cdc7 prevents their release from the scaffold, thus maintaining the interactions. We identify a rad51 mutant that is impaired in its ability to bind to MCM but not to the scaffold. This mutant is proficient in recombination but partially defective in single-stranded DNA (ssDNA) gap filling and replication fork progression through damaged DNA. Therefore, cells accumulate MCM/Rad51/Rad52 complexes at specific nuclear scaffolds in G1 to assist stressed forks through non-recombinogenic functions.


Assuntos
Replicação do DNA , DNA de Cadeia Simples/metabolismo , Recombinação Homóloga/genética , Complexos Multiproteicos/metabolismo , Rad51 Recombinase/metabolismo , Saccharomyces cerevisiae/metabolismo , Ciclo Celular/genética , Núcleo Celular/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Metanossulfonato de Metila , Modelos Biológicos , Ligação Proteica , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA