Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Nature ; 590(7847): 655-659, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33473214

RESUMO

Break-induced replication (BIR) repairs one-ended double-strand breaks in DNA similar to those formed by replication collapse or telomere erosion, and it has been implicated in the initiation of genome instability in cancer and other human diseases1,2. Previous studies have defined the enzymes that are required for BIR1-5; however, understanding of initial and extended BIR synthesis, and of how the migrating D-loop proceeds through known replication roadblocks, has been precluded by technical limitations. Here we use a newly developed assay to show that BIR synthesis initiates soon after strand invasion and proceeds more slowly than S-phase replication. Without primase, leading strand synthesis is initiated efficiently, but is unable to proceed beyond 30 kilobases, suggesting that primase is needed for stabilization of the nascent leading strand. DNA synthesis can initiate in the absence of Pif1 or Pol32, but does not proceed efficiently. Interstitial telomeric DNA disrupts and terminates BIR progression, and BIR initiation is suppressed by transcription proportionally to the transcription level. Collisions between BIR and transcription lead to mutagenesis and chromosome rearrangements at levels that exceed instabilities induced by transcription during normal replication. Together, these results provide fundamental insights into the mechanism of BIR and how BIR contributes to genome instability.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Replicação do DNA , Saccharomyces cerevisiae , Cromossomos Fúngicos/genética , DNA Helicases/deficiência , DNA Primase/metabolismo , DNA Fúngico/biossíntese , DNA Polimerase Dirigida por DNA/deficiência , Instabilidade Genômica , Cinética , Mutagênese , Mutação , Fase S , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae , Telômero/genética , Fatores de Tempo , Transcrição Gênica
2.
Mol Cell Biol ; 40(17)2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32541066

RESUMO

Rad3 is the orthologue of ATR and the sensor kinase of the DNA replication checkpoint in Schizosaccharomyces pombe Under replication stress, it initiates checkpoint signaling at the forks necessary for maintaining genome stability and cell survival. To better understand the checkpoint initiation process, we have carried out a genetic screen in fission yeast by random mutation of the genome, looking for mutants defective in response to the replication stress induced by hydroxyurea. In addition to the previously reported mutant with a C-to-Y change at position 307 encoded by tel2 (tel2-C307Y mutant) (Y.-J. Xu, S. Khan, A. C. Didier, M. Wozniak, et al., Mol Cell Biol 39:e00175-19, 2019, https://doi.org/10.1128/MCB.00175-19), this screen has identified six mutations in rqh1 encoding a RecQ DNA helicase. Surprisingly, these rqh1 mutations, except for a start codon mutation, are all in the helicase domain, indicating that the helicase activity of Rqh1 plays an important role in the replication checkpoint. In support of this notion, integration of two helicase-inactive mutations or deletion of rqh1 generated a similar Rad3 signaling defect, and heterologous expression of human RECQ1, BLM, and RECQ4 restored the Rad3 signaling and partially rescued a rqh1 helicase mutant. Therefore, the replication checkpoint function of Rqh1 is highly conserved, and mutations in the helicase domain of these human enzymes may cause the checkpoint defect and contribute to the cancer predisposition syndromes.


Assuntos
Quinase do Ponto de Checagem 2/metabolismo , DNA Helicases/metabolismo , Replicação do DNA , DNA Fúngico/biossíntese , Proteínas de Schizosaccharomyces pombe/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Quinase do Ponto de Checagem 2/genética , DNA Helicases/genética , DNA Fúngico/genética , DNA Fúngico/metabolismo , Instabilidade Genômica , Hidroxiureia/farmacologia , Proteínas Quinases/metabolismo , RecQ Helicases/genética , RecQ Helicases/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Transdução de Sinais/efeitos dos fármacos
3.
Mol Cell ; 66(3): 398-410.e4, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475874

RESUMO

Replication stress and mitotic abnormalities are key features of cancer cells. Temporarily paused forks are stabilized by the intra-S phase checkpoint and protected by the association of Rad51, which prevents Mre11-dependent resection. However, if a fork becomes dysfunctional and cannot resume, this terminally arrested fork is rescued by a converging fork to avoid unreplicated parental DNA during mitosis. Alternatively, dysfunctional forks are restarted by homologous recombination. Using fission yeast, we report that Rad52 and the DNA binding activity of Rad51, but not its strand-exchange activity, act to protect terminally arrested forks from unrestrained Exo1-nucleolytic activity. In the absence of recombination proteins, large ssDNA gaps, up to 3 kb long, occur behind terminally arrested forks, preventing efficient fork merging and leading to mitotic sister chromatid bridging. Thus, Rad52 and Rad51 prevent temporarily and terminally arrested forks from degrading and, despite the availability of converging forks, converting to anaphase bridges causing aneuploidy and cell death.


Assuntos
Replicação do DNA , DNA Fúngico/biossíntese , DNA de Cadeia Simples/biossíntese , Mitose/fisiologia , Origem de Replicação , Schizosaccharomyces/metabolismo , Troca de Cromátide Irmã , Aneuploidia , Cromossomos Fúngicos/genética , Cromossomos Fúngicos/metabolismo , Quebras de DNA de Cadeia Simples , DNA Fúngico/genética , DNA de Cadeia Simples/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Viabilidade Microbiana , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/crescimento & desenvolvimento , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Fatores de Tempo
4.
Proc Natl Acad Sci U S A ; 114(3): 552-557, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28049846

RESUMO

Replication gaps that persist into mitosis likely represent important threats to genome stability, but experimental identification of these gaps has proved challenging. We have developed a technique that allows us to explore the dynamics by which genome replication is completed before mitosis. Using this approach, we demonstrate that excessive allocation of replication resources to origins within repetitive regions, induced by SIR2 deletion, leads to persistent replication gaps and genome instability. Conversely, the weakening of replication origins in repetitive regions suppresses these gaps. Given known age- and cancer-associated changes in chromatin accessibility at repetitive sequences, we suggest that replication gaps resulting from misallocation of replication resources underlie age- and disease-associated genome instability.


Assuntos
Replicação do DNA , Instabilidade Genômica , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/metabolismo , Cromossomos Fúngicos/genética , DNA Fúngico/biossíntese , DNA Fúngico/genética , DNA Ribossômico/biossíntese , DNA Ribossômico/genética , Deleção de Genes , Genoma Fúngico , Humanos , Modelos Biológicos , Sequências Repetitivas de Ácido Nucleico , Origem de Replicação , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/deficiência , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Sirtuína 2/deficiência , Sirtuína 2/genética
5.
Cell Cycle ; 16(1): 45-58, 2017 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-27687866

RESUMO

Acetaldehyde, a primary metabolite of alcohol, forms DNA adducts and disrupts the DNA replication process, causing genomic instability, a hallmark of cancer. Indeed, chronic alcohol consumption accounts for approximately 3.6% of all cancers worldwide. However, how the adducts are prevented and repaired after acetaldehyde exposure is not well understood. In this report, we used the fission yeast Schizosaccharomyces pombe as a model organism to comprehensively understand the genetic controls of DNA damage avoidance in response to acetaldehyde. We demonstrate that Atd1 functions as a major acetaldehyde detoxification enzyme that prevents accumulation of Rad52-DNA repair foci, while Atd2 and Atd3 have minor roles in acetaldehyde detoxification. We found that acetaldehyde causes DNA damage at the replication fork and activates the cell cycle checkpoint to coordinate cell cycle arrest with DNA repair. Our investigation suggests that acetaldehyde-mediated DNA adducts include interstrand-crosslinks and DNA-protein crosslinks. We also demonstrate that acetaldehyde activates multiple DNA repair pathways. Nucleotide excision repair and homologous recombination, which are both epistatically linked to the Fanconi anemia pathway, have major roles in acetaldehyde tolerance, while base excision repair and translesion synthesis also contribute to the prevention of acetaldehyde-dependent genomic instability. We also show the involvement of Wss1-related metalloproteases, Wss1 and Wss2, in acetaldehyde tolerance. These results indicate that acetaldehyde causes cellular stresses that require cells to coordinate multiple cellular processes in order to prevent genomic instability. Considering that acetaldehyde is a human carcinogen, our genetic studies serve as a guiding investigation into the mechanisms of acetaldehyde-dependent genomic instability and carcinogenesis.


Assuntos
Acetaldeído/toxicidade , Dano ao DNA/genética , Schizosaccharomyces/genética , Adaptação Fisiológica/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Adutos de DNA/metabolismo , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA Fúngico/biossíntese , Epistasia Genética/efeitos dos fármacos , Anemia de Fanconi/genética , Genes Fúngicos , Recombinação Homóloga/efeitos dos fármacos , Inativação Metabólica/efeitos dos fármacos , Modelos Biológicos , Schizosaccharomyces/citologia , Schizosaccharomyces/efeitos dos fármacos , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
6.
Mol Cell ; 65(1): 117-130, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27989438

RESUMO

The integrity of eukaryotic genomes requires rapid and regulated chromatin replication. How this is accomplished is still poorly understood. Using purified yeast replication proteins and fully chromatinized templates, we have reconstituted this process in vitro. We show that chromatin enforces DNA replication origin specificity by preventing non-specific MCM helicase loading. Helicase activation occurs efficiently in the context of chromatin, but subsequent replisome progression requires the histone chaperone FACT (facilitates chromatin transcription). The FACT-associated Nhp6 protein, the nucleosome remodelers INO80 or ISW1A, and the lysine acetyltransferases Gcn5 and Esa1 each contribute separately to maximum DNA synthesis rates. Chromatin promotes the regular priming of lagging-strand DNA synthesis by facilitating DNA polymerase α function at replication forks. Finally, nucleosomes disrupted during replication are efficiently re-assembled into regular arrays on nascent DNA. Our work defines the minimum requirements for chromatin replication in vitro and shows how multiple chromatin factors might modulate replication fork rates in vivo.


Assuntos
Cromatina/genética , Replicação do DNA , DNA Fúngico/genética , Nucleossomos/genética , Origem de Replicação , Saccharomyces cerevisiae/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Cromatina/metabolismo , DNA Polimerase I/genética , DNA Polimerase I/metabolismo , DNA Fúngico/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Nucleossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Tempo , Fatores de Elongação da Transcrição/genética , Fatores de Elongação da Transcrição/metabolismo
7.
Mol Cell ; 62(4): 532-45, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27203178

RESUMO

Cell division entails a sequence of processes whose specific demands for biosynthetic precursors and energy place dynamic requirements on metabolism. However, little is known about how metabolic fluxes are coordinated with the cell division cycle. Here, we examine budding yeast to show that more than half of all measured metabolites change significantly through the cell division cycle. Cell cycle-dependent changes in central carbon metabolism are controlled by the cyclin-dependent kinase (Cdk1), a major cell cycle regulator, and the metabolic regulator protein kinase A. At the G1/S transition, Cdk1 phosphorylates and activates the enzyme Nth1, which funnels the storage carbohydrate trehalose into central carbon metabolism. Trehalose utilization fuels anabolic processes required to reliably complete cell division. Thus, the cell cycle entrains carbon metabolism to fuel biosynthesis. Because the oscillation of Cdk activity is a conserved feature of the eukaryotic cell cycle, we anticipate its frequent use in dynamically regulating metabolism for efficient proliferation.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteína Quinase CDC28 de Saccharomyces cerevisiae/metabolismo , Carbono/metabolismo , Ciclo Celular , Proliferação de Células , Metabolismo Energético , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Proteína Quinase CDC2/genética , Proteína Quinase CDC28 de Saccharomyces cerevisiae/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Replicação do DNA , DNA Fúngico/biossíntese , DNA Fúngico/genética , Ativação Enzimática , Pontos de Checagem da Fase G1 do Ciclo Celular , Fosforilação , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais , Fatores de Tempo , Trealase/metabolismo , Trealose/metabolismo
8.
DNA Repair (Amst) ; 42: 94-106, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27045900

RESUMO

Trinucleotide repeat expansions are responsible for at least two dozen neurological disorders. Mechanisms leading to these large expansions of repeated DNA are still poorly understood. It was proposed that transient stalling of the replication fork by the repeat tract might trigger slippage of the newly-synthesized strand over its template, leading to expansions or contractions of the triplet repeat. However, such mechanism was never formally proven. Here we show that replication fork pausing and CAG/CTG trinucleotide repeat instability are not linked, stable and unstable repeats exhibiting the same propensity to stall replication forks when integrated in a yeast natural chromosome. We found that replication fork stalling was dependent on the integrity of the mismatch-repair system, especially the Msh2p-Msh6p complex, suggesting that direct interaction of MMR proteins with secondary structures formed by trinucleotide repeats in vivo, triggers replication fork pauses. We also show by chromatin immunoprecipitation that Msh2p is enriched at trinucleotide repeat tracts, in both stable and unstable orientations, this enrichment being dependent on MSH3 and MSH6. Finally, we show that overexpressing MSH2 favors the formation of heteroduplex regions, leading to an increase in contractions and expansions of CAG/CTG repeat tracts during replication, these heteroduplexes being dependent on both MSH3 and MSH6. These heteroduplex regions were not detected when a mutant msh2-E768A gene in which the ATPase domain was mutated was overexpressed. Our results unravel two new roles for mismatch-repair proteins: stabilization of heteroduplex regions and transient blocking of replication forks passing through such repeats. Both roles may involve direct interactions between MMR proteins and secondary structures formed by trinucleotide repeat tracts, although indirect interactions may not be formally excluded.


Assuntos
Reparo de Erro de Pareamento de DNA , Replicação do DNA , Repetições de Trinucleotídeos/genética , DNA Fúngico/biossíntese , DNA Fúngico/química , DNA Fúngico/genética , DNA Fúngico/metabolismo , Proteína MutS de Ligação de DNA com Erro de Pareamento/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Expansão das Repetições de Trinucleotídeos
9.
J Biol Chem ; 291(16): 8825-35, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26921322

RESUMO

Small ubiquitin-like modifier (SUMO) E3 ligases are known to have a major role in preventing gross chromosomal rearrangements (GCRs); however, relatively little is known about the role of SUMO isopeptidases in genome maintenance and their role in controlling intracellular sumoylation homeostasis. Here we show the SUMO isopeptidase Ulp2 in Saccharomyces cerevisiae does not prevent the accumulation of GCRs, and interestingly, its loss causes subunit-specific changes of sumoylated minichromosome maintenance (MCM) helicase in addition to drastic accumulation of sumoylated nucleolar RENT and inner kinetochore complexes. In contrast, loss of Ulp1 or its mis-localization from the nuclear periphery causes substantial accumulations of GCRs and elevated sumoylation of most proteins except for Ulp2 targets. Interestingly, the E3 ligase Mms21, which has a major role in genome maintenance, preferentially controls the sumoylation of Mcm3 during DNA replication. These findings reveal distinct roles for Ulp1 and Ulp2 in controlling homeostasis of intracellular sumoylation and show that sumoylation of MCM is controlled in a subunit-specific and cell cycle dependent manner.


Assuntos
Nucléolo Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Endopeptidases/metabolismo , Proteína SUMO-1/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sumoilação/fisiologia , Acetiltransferases/genética , Acetiltransferases/metabolismo , Nucléolo Celular/genética , Cisteína Endopeptidases/genética , Replicação do DNA/fisiologia , DNA Fúngico/biossíntese , DNA Fúngico/genética , Endopeptidases/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína SUMO-1/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
10.
Nutr Hosp ; 32(6): 2932-7, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26667755

RESUMO

The objective of this study was to isolate endophytic fungi producing paclitaxel from yew for the purpose of paclitaxel manufacture. Surface sterilized bark of Taxus wallichiana var. mairei was used as source material and potato dextrose agar culture medium was used in isolation of endophytic fungi. Fungal cultures were extracted with a mixture of chloroform / methanol (1:1, v/v) and the paclitaxel in the extracts was determined and authenticated with LC-MS. An endophytic fungus that produced paclitaxel was identified by ITS rDNA and 26S D1/D2 rDNA sequencing. The results showed that a total of 435 endophytic fungal strains were isolated from T. wallichiana var. mairei and purified. Only one of these strains produced paclitaxel and it belongs to Fusarium. The paclitaxel productivity in whole PDB culture and that in spent culture medium from this strain is 0.0153 mg/L and 0.0119 mg/L respectively. The paclitaxel content in dry mycelium is 0.27 mg/kg. This isolated endophytic fungus produced paclitaxel at a considerable level and shows potentiality as a producing strain for paclitaxel manufacture after strain improvement.


El objetivo de este estudio fue aislar hongos endofíticos productores de paclitaxel a partir de tejo con el propósito de fabricar paclitaxel. Se utilizó la superficie de la corteza esterilizada de Taxus wallichiana var. mairei como material de origen y dextrosa de patata en medio de cultivo de agar para el aislamiento de hongos endófitos. Los cultivos de hongos se extrajeron con una mezcla de cloroformo / metanol (1:1, v/v) y el paclitaxel en los extractos se determinó y autentificó con LC-MS. Un hongo endófito que produjo paclitaxel fue identificado por su ADNr 26S y secuenciación D1/D2 ADNr. Los resultados mostraron que un total de 435 cepas de hongos endófitos se aislaron y purificarón a partir de T. wallichiana var. mairei. Solo una de estas cepas produce paclitaxel y pertenece a Fusarium. La productividad del cultivo de paclitaxel procedente de esta cepa es 0,0153 mg/L y 0,0119 mg/L, respectivamente. El contenido de paclitaxel en micelio seco es 0,27 mg/kg. Este aislado de hongos endófitos produjo paclitaxel a un nivel considerable y muestra potencial como cepa para la fabricación de paclitaxel después de llevar a cabo una mejora de las cepas.


Assuntos
Antineoplásicos Fitogênicos/biossíntese , Endófitos/química , Paclitaxel/biossíntese , Taxus/microbiologia , DNA Fúngico/biossíntese , DNA Fúngico/genética , Endófitos/metabolismo , Fusarium/metabolismo , Micélio/química , Micélio/metabolismo , Casca de Planta/microbiologia
11.
Biochem Biophys Res Commun ; 451(1): 36-41, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25065741

RESUMO

BACKGROUND: These experiments were employed to explore the mechanisms underlying baicalin action on Candida albicans. METHODOLOGY AND PRINCIPAL FINDINGS: We detected the baicalin inhibition effects on three isotope-labeled precursors of (3)H-UdR, (3)H-TdR and (3)H-leucine incorporation into C. albicans using the isotope incorporation technology. The activities of Succinate Dehydrogenase (SDH), cytochrome oxidase (CCO) and Ca(2)(+)-Mg(2+) ATPase, cytosolic Ca(2+) concentration, the cell cycle and apoptosis, as well as the ultrastructure of C.albicans were also tested. We found that baicalin inhibited (3)H-UdR, (3)H-TdR and (3)H-leucine incorporation into C.albicans (P<0.005). The activities of the SDH and Ca(2)(+)-Mg(2+) ATPase of C.albicans in baicalin groups were lower than those in control group (P<0.05). Ca(2+) concentrations of C. albicans in baicalin groups were much higher than those in control group (P<0.05). The ratio of C.albicans at the G0/G1 stage increased in baicalin groups in dose dependent manner (P<0.01). There were a significant differences in the apoptosis rate of C.albicans between baicalin and control groups (P<0.01). After 12-48 h incubation with baicalin (1mg/ml), C. albicans shown to be markedly damaged under transmission electron micrographs. INNOVATION AND SIGNIFICANCE: Baicalin can increase the apoptosis rate of C. albicans. These effects of Baicalin may involved in its inhibiting the activities of the SDH and Ca(2)(+)-Mg(2+) ATPase, increasing cytosolic Ca(2+) content and damaging the ultrastructure of C. albicans.


Assuntos
Anti-Infecciosos/farmacologia , Candida albicans/efeitos dos fármacos , Flavonoides/farmacologia , Apoptose/efeitos dos fármacos , ATPase de Ca(2+) e Mg(2+)/antagonistas & inibidores , Cálcio/metabolismo , Candida albicans/genética , Candida albicans/metabolismo , Candida albicans/ultraestrutura , Candidíase/prevenção & controle , Ciclo Celular/efeitos dos fármacos , DNA Fúngico/biossíntese , RNA Fúngico/biossíntese , Succinato Desidrogenase/antagonistas & inibidores
12.
PLoS Genet ; 10(6): e1004358, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945837

RESUMO

Eukaryotic cells must inhibit re-initiation of DNA replication at each of the thousands of origins in their genome because re-initiation can generate genomic alterations with extraordinary frequency. To minimize the probability of re-initiation from so many origins, cells use a battery of regulatory mechanisms that reduce the activity of replication initiation proteins. Given the global nature of these mechanisms, it has been presumed that all origins are inhibited identically. However, origins re-initiate with diverse efficiencies when these mechanisms are disabled, and this diversity cannot be explained by differences in the efficiency or timing of origin initiation during normal S phase replication. This observation raises the possibility of an additional layer of replication control that can differentially regulate re-initiation at distinct origins. We have identified novel genetic elements that are necessary for preferential re-initiation of two origins and sufficient to confer preferential re-initiation on heterologous origins when the control of re-initiation is partially deregulated. The elements do not enhance the S phase timing or efficiency of adjacent origins and thus are specifically acting as re-initiation promoters (RIPs). We have mapped the two RIPs to ∼ 60 bp AT rich sequences that act in a distance- and sequence-dependent manner. During the induction of re-replication, Mcm2-7 reassociates both with origins that preferentially re-initiate and origins that do not, suggesting that the RIP elements can overcome a block to re-initiation imposed after Mcm2-7 associates with origins. Our findings identify a local level of control in the block to re-initiation. This local control creates a complex genomic landscape of re-replication potential that is revealed when global mechanisms preventing re-replication are compromised. Hence, if re-replication does contribute to genomic alterations, as has been speculated for cancer cells, some regions of the genome may be more susceptible to these alterations than others.


Assuntos
Replicação do DNA/genética , DNA Fúngico/biossíntese , Regiões Promotoras Genéticas/genética , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Quinases Ciclina-Dependentes/metabolismo , Variação Genética , Genoma Fúngico , Proteínas de Manutenção de Minicromossomo/genética , Origem de Replicação/genética , Fase S/genética , Proteínas de Saccharomyces cerevisiae/biossíntese
13.
Nature ; 502(7471): 393-6, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24025768

RESUMO

During DNA repair by homologous recombination (HR), DNA synthesis copies information from a template DNA molecule. Multiple DNA polymerases have been implicated in repair-specific DNA synthesis, but it has remained unclear whether a DNA helicase is involved in this reaction. A good candidate DNA helicase is Pif1, an evolutionarily conserved helicase in Saccharomyces cerevisiae important for break-induced replication (BIR) as well as HR-dependent telomere maintenance in the absence of telomerase found in 10-15% of all cancers. Pif1 has a role in DNA synthesis across hard-to-replicate sites and in lagging-strand synthesis with polymerase δ (Polδ). Here we provide evidence that Pif1 stimulates DNA synthesis during BIR and crossover recombination. The initial steps of BIR occur normally in Pif1-deficient cells, but Polδ recruitment and DNA synthesis are decreased, resulting in premature resolution of DNA intermediates into half-crossovers. Purified Pif1 protein strongly stimulates Polδ-mediated DNA synthesis from a D-loop made by the Rad51 recombinase. Notably, Pif1 liberates the newly synthesized strand to prevent the accumulation of topological constraint and to facilitate extensive DNA synthesis via the establishment of a migrating D-loop structure. Our results uncover a novel function of Pif1 and provide insights into the mechanism of HR.


Assuntos
Troca Genética , DNA Helicases/metabolismo , DNA Polimerase III/metabolismo , Replicação do DNA , DNA Fúngico/biossíntese , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , DNA Helicases/deficiência , DNA Helicases/genética , Reparo do DNA , DNA Fúngico/química , DNA Fúngico/metabolismo , Conformação de Ácido Nucleico , Rad51 Recombinase/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
14.
Nature ; 502(7471): 389-92, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24025772

RESUMO

The repair of chromosomal double strand breaks (DSBs) is crucial for the maintenance of genomic integrity. However, the repair of DSBs can also destabilize the genome by causing mutations and chromosomal rearrangements, the driving forces for carcinogenesis and hereditary diseases. Break-induced replication (BIR) is one of the DSB repair pathways that is highly prone to genetic instability. BIR proceeds by invasion of one broken end into a homologous DNA sequence followed by replication that can copy hundreds of kilobases of DNA from a donor molecule all the way through its telomere. The resulting repaired chromosome comes at a great cost to the cell, as BIR promotes mutagenesis, loss of heterozygosity, translocations, and copy number variations, all hallmarks of carcinogenesis. BIR uses most known replication proteins to copy large portions of DNA, similar to S-phase replication. It has therefore been suggested that BIR proceeds by semiconservative replication; however, the model of a bona fide, stable replication fork contradicts the known instabilities associated with BIR such as a 1,000-fold increase in mutation rate compared to normal replication. Here we demonstrate that in budding yeast the mechanism of replication during BIR is significantly different from S-phase replication, as it proceeds via an unusual bubble-like replication fork that results in conservative inheritance of the new genetic material. We provide evidence that this atypical mode of DNA replication, dependent on Pif1 helicase, is responsible for the marked increase in BIR-associated mutations. We propose that the BIR mode of synthesis presents a powerful mechanism that can initiate bursts of genetic instability in eukaryotes, including humans.


Assuntos
Quebra Cromossômica , Quebras de DNA de Cadeia Dupla , Replicação do DNA/genética , DNA Fúngico/biossíntese , Saccharomyces cerevisiae/genética , DNA Helicases/metabolismo , Reparo do DNA/genética , DNA Fúngico/genética , Instabilidade Genômica/genética , Mutagênese/genética , Fase S/genética , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
DNA Repair (Amst) ; 12(11): 878-89, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23988736

RESUMO

Abasic sites in genomic DNA can be a significant source of mutagenesis in biological systems, including human cancers. Such mutagenesis requires translesion DNA synthesis (TLS) bypass of the abasic site by specialized DNA polymerases. The abasic site bypass specificity of TLS proteins had been studied by multiple means in vivo and in vitro, although the generality of the conclusions reached have been uncertain. Here, we introduce a set of yeast reporter strains for investigating the in vivo specificity of abasic site bypass at numerous random positions within chromosomal DNA. When shifted to 37°C, these strains underwent telomere uncapping and resection that exposed reporter genes within a long 3' ssDNA overhang. Human APOBEC3G cytosine deaminase was expressed to create uracils in ssDNA, which were excised by uracil-DNA N-glycosylase. During repair synthesis, error-prone TLS bypassed the resulting abasic sites. Because of APOBEC3G's strict motif specificity and the restriction of abasic site formation to only one DNA strand, this system provides complete information about the location of abasic sites that led to mutations. We recapitulated previous findings on the roles of REV1 and REV3. Further, we found that sequence context can strongly influence the relative frequency of A or C insertion. We also found that deletion of Pol32, a non-essential common subunit of Pols δ and ζ, resulted in residual low-frequency C insertion dependent on Rev1 catalysis. We summarize our results in a detailed model of the interplay between TLS components leading to error-prone bypass of abasic sites. Our results underscore the utility of this system for studying TLS bypass of many types of lesions within genomic DNA.


Assuntos
Cromossomos Fúngicos , DNA Fúngico/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Nucleotídeos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Desaminase APOBEC-3G , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , DNA Fúngico/genética , DNA de Cadeia Simples/metabolismo , Humanos , Mutagênese Insercional , Uracila/metabolismo , Uracila-DNA Glicosidase/metabolismo
16.
BMC Syst Biol ; 6: 78, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22738223

RESUMO

BACKGROUND: Eukaryotic cell proliferation involves DNA replication, a tightly regulated process mediated by a multitude of protein factors. In budding yeast, the initiation of replication is facilitated by the heterohexameric origin recognition complex (ORC). ORC binds to specific origins of replication and then serves as a scaffold for the recruitment of other factors such as Cdt1, Cdc6, the Mcm2-7 complex, Cdc45 and the Dbf4-Cdc7 kinase complex. While many of the mechanisms controlling these associations are well documented, mathematical models are needed to explore the network's dynamic behaviour. We have developed an ordinary differential equation-based model of the protein-protein interaction network describing replication initiation. RESULTS: The model was validated against quantified levels of protein factors over a range of cell cycle timepoints. Using chromatin extracts from synchronized Saccharomyces cerevisiae cell cultures, we were able to monitor the in vivo fluctuations of several of the aforementioned proteins, with additional data obtained from the literature. The model behaviour conforms to perturbation trials previously reported in the literature, and accurately predicts the results of our own knockdown experiments. Furthermore, we successfully incorporated our replication initiation model into an established model of the entire yeast cell cycle, thus providing a comprehensive description of these processes. CONCLUSIONS: This study establishes a robust model of the processes driving DNA replication initiation. The model was validated against observed cell concentrations of the driving factors, and characterizes the interactions between factors implicated in eukaryotic DNA replication. Finally, this model can serve as a guide in efforts to generate a comprehensive model of the mammalian cell cycle in order to explore cancer-related phenotypes.


Assuntos
Replicação do DNA , DNA Fúngico/biossíntese , Modelos Biológicos , Saccharomyces cerevisiae/metabolismo , Biologia de Sistemas/métodos , Calibragem , Ciclo Celular , Saccharomyces cerevisiae/citologia
17.
EMBO J ; 31(4): 883-94, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22234185

RESUMO

Intracellular deoxyribonucleoside triphosphate (dNTP) pools must be tightly regulated to preserve genome integrity. Indeed, alterations in dNTP pools are associated with increased mutagenesis, genomic instability and tumourigenesis. However, the mechanisms by which altered or imbalanced dNTP pools affect DNA synthesis remain poorly understood. Here, we show that changes in intracellular dNTP levels affect replication dynamics in budding yeast in different ways. Upregulation of the activity of ribonucleotide reductase (RNR) increases elongation, indicating that dNTP pools are limiting for normal DNA replication. In contrast, inhibition of RNR activity with hydroxyurea (HU) induces a sharp transition to a slow-replication mode within minutes after S-phase entry. Upregulation of RNR activity delays this transition and modulates both fork speed and origin usage under replication stress. Interestingly, we also observed that chromosomal instability (CIN) mutants have increased dNTP pools and show enhanced DNA synthesis in the presence of HU. Since upregulation of RNR promotes fork progression in the presence of DNA lesions, we propose that CIN mutants adapt to chronic replication stress by upregulating dNTP pools.


Assuntos
Replicação do DNA , Desoxirribonucleosídeos/metabolismo , Origem de Replicação , Saccharomyces cerevisiae/genética , Bromodesoxiuridina , Dano ao DNA , DNA Fúngico/biossíntese , DNA Fúngico/genética , Hidroxiureia/farmacologia , Imunoprecipitação , Ribonucleotídeo Redutases/metabolismo , Fase S , Saccharomyces cerevisiae/enzimologia
18.
Subcell Biochem ; 57: 187-206, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22094423

RESUMO

DNA damage DNA damage is an important factor in aging in all eukaryotes. Although connections between DNA damage DNA damage and aging have been extensively investigated in complex organisms, only a relatively few studies have investigated DNA damage DNA damage as an aging factor in the model organism S. cerevisiae. Several of these studies point to DNA replication stress DNA replication stress as a cause of age-dependent DNA damage DNA damage in the replicative model of aging, which measures how many times budding yeast cells divide before they senesce and die. Even fewer studies have investigated how DNA damage DNA damage contributes to aging in the chronological aging chronological aging model, which measures how long cells in stationary phase cultures retain reproductive capacity. DNA replication stress DNA replication stress also has been implicated as a factor in chronological aging chronological aging . Since cells in stationary phase are generally considered to be "post-mitotic" and to reside in a quiescent G0/G1 state, the notion that defects in DNA replication might contribute to chronological aging chronological aging appears to be somewhat paradoxical. However, the results of recent studies suggest that a significant fraction of cells in stationary phase cultures are not quiescent, especially in experiments that employ defined medium, which is frequently employed to assess chronological lifespan. Most cells that fail to achieve quiescence remain in a viable, but non-dividing state until they eventually die, similar to the senescent state in mammalian cells. In this chapter we discuss the role of DNA damage DNA damage and DNA replication stress DNA replication stress in both replicative and chronological aging chronological aging in S. cerevisiae. We also discuss the relevance of these findings to the emerging view that DNA damage DNA damage and DNA replication stress DNA replication stress are important components of the senescent state that occurs at early stages of cancer.


Assuntos
Envelhecimento/genética , Dano ao DNA , Replicação do DNA , DNA Fúngico/biossíntese , Modelos Genéticos , Saccharomycetales/genética , Estresse Fisiológico/genética , Envelhecimento/metabolismo , Animais , Divisão Celular , Regulação Fúngica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Longevidade , Neoplasias/genética , Neoplasias/metabolismo , Saccharomycetales/crescimento & desenvolvimento , Saccharomycetales/metabolismo , Fatores de Tempo
19.
Genetics ; 187(4): 1031-40, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21242538

RESUMO

This study reports an unusual ploidy-specific response to replication stress presented by a defective minichromosome maintenance (MCM) helicase allele in yeast. The corresponding mouse allele, Mcm4(Chaos3), predisposes mice to mammary gland tumors. While mcm4(Chaos3) causes replication stress in both haploid and diploid yeast, only diploid mutants exhibit G2/M delay, severe genetic instability (GIN), and reduced viability. These different outcomes are associated with distinct repair pathways adopted in haploid and diploid mutants. Haploid mutants use the Rad6-dependent pathways that resume stalled forks, whereas the diploid mutants use the Rad52- and MRX-dependent pathways that repair double strand breaks. The repair pathway choice is irreversible and not regulated by the availability of repair enzymes. This ploidy effect is independent of mating type heterozygosity and not further enhanced by increasing ploidy. In summary, a defective MCM helicase causes GIN only in particular cell types. In response to replication stress, early events associated with ploidy dictate the repair pathway choice. This study uncovers a fundamental difference between haplophase and diplophase in the maintenance of genome integrity.


Assuntos
Proteínas de Ciclo Celular/genética , Reparo do DNA , Replicação do DNA , DNA Fúngico/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Ciclo Celular , Dano ao DNA , DNA Helicases/genética , DNA Helicases/metabolismo , Análise Mutacional de DNA , DNA Fúngico/genética , Diploide , Loci Gênicos , Haploidia , Heterozigoto , Componente 4 do Complexo de Manutenção de Minicromossomo , Mutação , Rad51 Recombinase/metabolismo , Recombinação Genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Enzimas de Conjugação de Ubiquitina/metabolismo
20.
Genetics ; 187(1): 21-35, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980236

RESUMO

A cell's ability to tolerate DNA damage is directly connected to the human development of diseases and cancer. To better understand the processes underlying mutagenesis, we studied the cell's reliance on the potentially error-prone translesion synthesis (TLS), and an error-free, template-switching pathway in Saccharomyces cerevisiae. The primary proteins mediating S. cerevisiae TLS are three DNA polymerases (Pols): Rev1, Pol ζ (Rev3/7), and Pol η (Rad30), all with human homologs. Rev1's noncatalytic role in recruiting other DNA polymerases is known to be important for TLS. However, the biological significance of Rev1's unusual conserved DNA polymerase activity, which inserts dC, is much less well understood. Here, we demonstrate that inactivating Rev1's DNA polymerase function sensitizes cells to both chronic and acute exposure to 4-nitroquinoline-1-oxide (4-NQO) but not to UV or cisplatin. Full Rev1-dependent resistance to 4-NQO, however, also requires the additional Rev1 functions. When error-free tolerance is disrupted through deletion of MMS2, Rev1's catalytic activity is more vital for 4-NQO resistance, possibly explaining why the biological significance of Rev1's catalytic activity has been elusive. In the presence or absence of Mms2-dependent error-free tolerance, the catalytic dead strain of Rev1 exhibits a lower 4-NQO-induced mutation frequency than wild type. Furthermore, Pol ζ, but not Pol η, also contributes to 4-NQO resistance. These results show that Rev1's catalytic activity is important in vivo when the cell has to cope with specific DNA lesions, such as N(2)-dG.


Assuntos
DNA Polimerase Dirigida por DNA/metabolismo , Nucleotidiltransferases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , 4-Nitroquinolina-1-Óxido/farmacologia , Biocatálise , Cisplatino/farmacologia , Adutos de DNA/genética , Adutos de DNA/metabolismo , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , DNA Fúngico/biossíntese , DNA Fúngico/genética , DNA Polimerase Dirigida por DNA/química , Humanos , Mutação/efeitos dos fármacos , Mutação/efeitos da radiação , Nucleotidiltransferases/química , Estrutura Terciária de Proteína , Aldeído Pirúvico/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/efeitos da radiação , Proteínas de Saccharomyces cerevisiae/química , Raios Ultravioleta , DNA Polimerase iota
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA