Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 141(4): 595-605, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20478253

RESUMEN

Actively dividing cells perform robust and accurate DNA replication during fluctuating nutrient availability, yet factors that prevent disruption of replication remain largely unknown. Here we report that DksA, a nutrient-responsive transcription factor, ensures replication completion in Escherichia coli by removing transcription roadblocks. In the absence of DksA, replication is rapidly arrested upon amino acid starvation. This arrest requires active transcription and is alleviated by RNA polymerase mutants that compensate for DksA activity. This replication arrest occurs independently of exogenous DNA damage, yet it induces the DNA-damage response and recruits the main recombination protein RecA. This function of DksA is independent of its transcription initiation activity but requires its less-studied transcription elongation activity. Finally, GreA/B elongation factors also prevent replication arrest during nutrient stress. We conclude that transcription elongation factors alleviate fundamental conflicts between replication and transcription, thereby protecting replication fork progression and DNA integrity.


Asunto(s)
Replicación del ADN , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Transcripción Genética , Aminoácidos/metabolismo , Daño del ADN , Reparación del ADN , Guanosina Pentafosfato/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(32): 19415-19424, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32719125

RESUMEN

Synthetic lethality strategies for cancer therapy exploit cancer-specific genetic defects to identify targets that are uniquely essential to the survival of tumor cells. Here we show RAD27/FEN1, which encodes flap endonuclease 1 (FEN1), a structure-specific nuclease with roles in DNA replication and repair, and has the greatest number of synthetic lethal interactions with Saccharomyces cerevisiae genome instability genes, is a druggable target for an inhibitor-based approach to kill cancers with defects in homologous recombination (HR). The vulnerability of cancers with HR defects to FEN1 loss was validated by studies showing that small-molecule FEN1 inhibitors and FEN1 small interfering RNAs (siRNAs) selectively killed BRCA1- and BRCA2-defective human cell lines. Furthermore, the differential sensitivity to FEN1 inhibition was recapitulated in mice, where a small-molecule FEN1 inhibitor reduced the growth of tumors established from drug-sensitive but not drug-resistant cancer cell lines. FEN1 inhibition induced a DNA damage response in both sensitive and resistant cell lines; however, sensitive cell lines were unable to recover and replicate DNA even when the inhibitor was removed. Although FEN1 inhibition activated caspase to higher levels in sensitive cells, this apoptotic response occurred in p53-defective cells and cell killing was not blocked by a pan-caspase inhibitor. These results suggest that FEN1 inhibitors have the potential for therapeutically targeting HR-defective cancers such as those resulting from BRCA1 and BRCA2 mutations, and other genetic defects.


Asunto(s)
Antineoplásicos/farmacología , Endonucleasas de ADN Solapado/antagonistas & inhibidores , Recombinación Homóloga/efectos de los fármacos , Neoplasias/genética , Animales , Proteína BRCA1/deficiencia , Proteína BRCA1/genética , Proteína BRCA2/deficiencia , Proteína BRCA2/genética , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Endonucleasas de ADN Solapado/genética , Inestabilidad Genómica/genética , Humanos , Ratones , Neoplasias/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Mutaciones Letales Sintéticas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 116(35): 17377-17382, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31409704

RESUMEN

Gross Chromosomal Rearrangements (GCRs) play an important role in human diseases, including cancer. Although most of the nonessential Genome Instability Suppressing (GIS) genes in Saccharomyces cerevisiae are known, the essential genes in which mutations can cause increased GCR rates are not well understood. Here 2 S. cerevisiae GCR assays were used to screen a targeted collection of temperature-sensitive mutants to identify mutations that caused increased GCR rates. This identified 94 essential GIS (eGIS) genes in which mutations cause increased GCR rates and 38 candidate eGIS genes that encode eGIS1 protein-interacting or family member proteins. Analysis of TCGA data using the human genes predicted to encode the proteins and protein complexes implicated by the S. cerevisiae eGIS genes revealed a significant enrichment of mutations affecting predicted human eGIS genes in 10 of the 16 cancers analyzed.


Asunto(s)
Genes Supresores , Genoma Fúngico , Inestabilidad Genómica , Neoplasias/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas Supresoras de Tumor/genética , Daño del ADN , Humanos , Mutación , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Supresoras de Tumor/metabolismo
4.
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
5.
Hum Mol Genet ; 26(2): 258-269, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28013290

RESUMEN

The integrity and dynamic properties of the microtubule cytoskeleton are indispensable for the development of the mammalian brain. Consequently, mutations in the genes that encode the structural component (the α/ß-tubulin heterodimer) can give rise to severe, sporadic neurodevelopmental disorders. These are commonly referred to as the tubulinopathies. Here we report the addition of recessive quadrupedalism, also known as Uner Tan syndrome (UTS), to the growing list of diseases caused by tubulin variants. Analysis of a consanguineous UTS family identified a biallelic TUBB2B mutation, resulting in a p.R390Q amino acid substitution. In addition to the identifying quadrupedal locomotion, all three patients showed severe cerebellar hypoplasia. None, however, displayed the basal ganglia malformations typically associated with TUBB2B mutations. Functional analysis of the R390Q substitution revealed that it did not affect the ability of ß-tubulin to fold or become assembled into the α/ß-heterodimer, nor did it influence the incorporation of mutant-containing heterodimers into microtubule polymers. The 390Q mutation in S. cerevisiae TUB2 did not affect growth under basal conditions, but did result in increased sensitivity to microtubule-depolymerizing drugs, indicative of a mild impact of this mutation on microtubule function. The TUBB2B mutation described here represents an unusual recessive mode of inheritance for missense-mediated tubulinopathies and reinforces the sensitivity of the developing cerebellum to microtubule defects.


Asunto(s)
Cerebelo/anomalías , Malformaciones del Desarrollo Cortical/genética , Microtúbulos/genética , Malformaciones del Sistema Nervioso/genética , Tubulina (Proteína)/genética , Adulto , Sustitución de Aminoácidos/genética , Ganglios Basales/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Cerebelo/fisiopatología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Femenino , Homocigoto , Humanos , Masculino , Malformaciones del Desarrollo Cortical/fisiopatología , Microtúbulos/patología , Mutación , Malformaciones del Sistema Nervioso/fisiopatología , Fenotipo , Saccharomyces cerevisiae/genética
6.
PLoS Genet ; 10(5): e1004327, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24811092

RESUMEN

In Saccharomyces cerevisiae, the essential mismatch repair (MMR) endonuclease Mlh1-Pms1 forms foci promoted by Msh2-Msh6 or Msh2-Msh3 in response to mispaired bases. Here we analyzed the Mlh1-Mlh2 complex, whose role in MMR has been unclear. Mlh1-Mlh2 formed foci that often colocalized with and had a longer lifetime than Mlh1-Pms1 foci. Mlh1-Mlh2 foci were similar to Mlh1-Pms1 foci: they required mispair recognition by Msh2-Msh6, increased in response to increased mispairs or downstream defects in MMR, and formed after induction of DNA damage by phleomycin but not double-stranded breaks by I-SceI. Mlh1-Mlh2 could be recruited to mispair-containing DNA in vitro by either Msh2-Msh6 or Msh2-Msh3. Deletion of MLH2 caused a synergistic increase in mutation rate in combination with deletion of MSH6 or reduced expression of Pms1. Phylogenetic analysis demonstrated that the S. cerevisiae Mlh2 protein and the mammalian PMS1 protein are homologs. These results support a hypothesis that Mlh1-Mlh2 is a non-essential accessory factor that acts to enhance the activity of Mlh1-Pms1.


Asunto(s)
Disparidad de Par Base , ADN de Hongos/genética , Proteínas Nucleares/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/genética , Proteínas Portadoras/fisiología , Daño del ADN , Mutación del Sistema de Lectura , Proteínas MutL
7.
J Biol Chem ; 290(35): 21580-90, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26170454

RESUMEN

Previous studies reported the reconstitution of an Mlh1-Pms1-independent 5' nick-directed mismatch repair (MMR) reaction using Saccharomyces cerevisiae proteins. Here we describe the reconstitution of a mispair-dependent Mlh1-Pms1 endonuclease activation reaction requiring Msh2-Msh6 (or Msh2-Msh3), proliferating cell nuclear antigen (PCNA), and replication factor C (RFC) and a reconstituted Mlh1-Pms1-dependent 3' nick-directed MMR reaction requiring Msh2-Msh6 (or Msh2-Msh3), exonuclease 1 (Exo1), replication protein A (RPA), RFC, PCNA, and DNA polymerase δ. Both reactions required Mg(2+) and Mn(2+) for optimal activity. The MMR reaction also required two reaction stages in which the first stage required incubation of Mlh1-Pms1 with substrate DNA, with or without Msh2-Msh6 (or Msh2-Msh3), PCNA, and RFC but did not require nicking of the substrate, followed by a second stage in which other proteins were added. Analysis of different mutant proteins demonstrated that both reactions required a functional Mlh1-Pms1 endonuclease active site, as well as mispair recognition and Mlh1-Pms1 recruitment by Msh2-Msh6 but not sliding clamp formation. Mutant Mlh1-Pms1 and PCNA proteins that were defective for Exo1-independent but not Exo1-dependent MMR in vivo were partially defective in the Mlh1-Pms1 endonuclease and MMR reactions, suggesting that both reactions reflect the activation of Mlh1-Pms1 seen in Exo1-independent MMR in vivo. The availability of this reconstituted MMR reaction should now make it possible to better study both Exo1-independent and Exo1-dependent MMR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Reparación de la Incompatibilidad de ADN , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Biocatálisis/efectos de los fármacos , Cationes Bivalentes/farmacología , Reparación de la Incompatibilidad de ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Genes Dominantes , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteínas Mutantes/metabolismo , Mutación , Saccharomyces cerevisiae/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 110(46): 18472-7, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24187148

RESUMEN

A problem in understanding eukaryotic DNA mismatch repair (MMR) mechanisms is linking insights into MMR mechanisms from genetics and cell-biology studies with those from biochemical studies of MMR proteins and reconstituted MMR reactions. This type of analysis has proven difficult because reconstitution approaches have been most successful for human MMR whereas analysis of MMR in vivo has been most advanced in the yeast Saccharomyces cerevisiae. Here, we describe the reconstitution of MMR reactions using purified S. cerevisiae proteins and mispair-containing DNA substrates. A mixture of MutS homolog 2 (Msh2)-MutS homolog 6, Exonuclease 1, replication protein A, replication factor C-Δ1N, proliferating cell nuclear antigen and DNA polymerase δ was found to repair substrates containing TG, CC, +1 (+T), +2 (+GC), and +4 (+ACGA) mispairs and either a 5' or 3' strand interruption with different efficiencies. The Msh2-MutS homolog 3 mispair recognition protein could substitute for the Msh2-Msh6 mispair recognition protein and showed a different specificity of repair of the different mispairs whereas addition of MutL homolog 1-postmeiotic segregation 1 had no affect on MMR. Repair was catalytic, with as many as 11 substrates repaired per molecule of Exo1. Repair of the substrates containing either a 5' or 3' strand interruption occurred by mispair binding-dependent 5' excision and subsequent resynthesis with excision tracts of up to ~2.9 kb occurring during the repair of the substrate with a 3' strand interruption. The availability of this reconstituted MMR reaction now makes possible detailed biochemical studies of the wealth of mutations identified that affect S. cerevisiae MMR.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , ADN de Hongos/metabolismo , Inestabilidad Genómica/genética , Complejos Multiproteicos/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Catálisis , ADN Polimerasa III/metabolismo , Cartilla de ADN/genética , ADN de Hongos/genética , Exodesoxirribonucleasas/metabolismo , Microscopía Electrónica de Transmisión , Complejos Multiproteicos/genética , Proteína 2 Homóloga a MutS/genética , Reacción en Cadena de la Polimerasa , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de Replicación A/metabolismo
9.
J Biol Chem ; 289(13): 9352-64, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24550389

RESUMEN

DNA mismatch repair is initiated by either the Msh2-Msh6 or the Msh2-Msh3 mispair recognition heterodimer. Here we optimized the expression and purification of Saccharomyces cerevisiae Msh2-Msh3 and performed a comparative study of Msh2-Msh3 and Msh2-Msh6 for mispair binding, sliding clamp formation, and Mlh1-Pms1 recruitment. Msh2-Msh3 formed sliding clamps and recruited Mlh1-Pms1 on +1, +2, +3, and +4 insertion/deletions and CC, AA, and possibly GG mispairs, whereas Msh2-Msh6 formed mispair-dependent sliding clamps and recruited Mlh1-Pms1 on 7 of the 8 possible base:base mispairs, the +1 insertion/deletion mispair, and to a low level on the +2 but not the +3 or +4 insertion/deletion mispairs and not on the CC mispair. The mispair specificity of sliding clamp formation and Mlh1-Pms1 recruitment but not mispair binding alone correlated best with genetic data on the mispair specificity of Msh2-Msh3- and Msh2-Msh6-dependent mismatch repair in vivo. Analysis of an Msh2-Msh6/Msh3 chimeric protein and mutant Msh2-Msh3 complexes showed that the nucleotide binding domain and communicating regions but not the mispair binding domain of Msh2-Msh3 are responsible for the extremely rapid dissociation of Msh2-Msh3 sliding clamps from DNA relative to that seen for Msh2-Msh6, and that amino acid residues predicted to stabilize Msh2-Msh3 interactions with bent, strand-separated mispair-containing DNA are more critical for the recognition of small +1 insertion/deletions than larger +4 insertion/deletions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Unión al ADN/genética , Mutación INDEL , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteína 3 Homóloga de MutS , Mutación , Proteínas de Saccharomyces cerevisiae/genética
10.
PLoS Genet ; 6(1): e1000810, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20090829

RESUMEN

In many bacteria, there is a genome-wide bias towards co-orientation of replication and transcription, with essential and/or highly-expressed genes further enriched co-directionally. We previously found that reversing this bias in the bacterium Bacillus subtilis slows replication elongation, and we proposed that this effect contributes to the evolutionary pressure selecting the transcription-replication co-orientation bias. This selection might have been based purely on selection for speedy replication; alternatively, the slowed replication might actually represent an average of individual replication-disruption events, each of which is counter-selected independently because genome integrity is selected. To differentiate these possibilities and define the precise forces driving this aspect of genome organization, we generated new strains with inversions either over approximately 1/4 of the chromosome or at ribosomal RNA (rRNA) operons. Applying mathematical analysis to genomic microarray snapshots, we found that replication rates vary dramatically within the inverted genome. Replication is moderately impeded throughout the inverted region, which results in a small but significant competitive disadvantage in minimal medium. Importantly, replication is strongly obstructed at inverted rRNA loci in rich medium. This obstruction results in disruption of DNA replication, activation of DNA damage responses, loss of genome integrity, and cell death. Our results strongly suggest that preservation of genome integrity drives the evolution of co-orientation of replication and transcription, a conserved feature of genome organization.


Asunto(s)
Bacillus subtilis/genética , Replicación del ADN , Genoma Bacteriano , Transcripción Genética , Bacillus subtilis/crecimiento & desarrollo , Mutación
11.
PLoS Genet ; 4(8): e1000139, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18670626

RESUMEN

Whole-genome sequencing is a powerful technique for obtaining the reference sequence information of multiple organisms. Its use can be dramatically expanded to rapidly identify genomic variations, which can be linked with phenotypes to obtain biological insights. We explored these potential applications using the emerging next-generation sequencing platform Solexa Genome Analyzer, and the well-characterized model bacterium Bacillus subtilis. Combining sequencing with experimental verification, we first improved the accuracy of the published sequence of the B. subtilis reference strain 168, then obtained sequences of multiple related laboratory strains and different isolates of each strain. This provides a framework for comparing the divergence between different laboratory strains and between their individual isolates. We also demonstrated the power of Solexa sequencing by using its results to predict a defect in the citrate signal transduction pathway of a common laboratory strain, which we verified experimentally. Finally, we examined the molecular nature of spontaneously generated mutations that suppress the growth defect caused by deletion of the stringent response mediator relA. Using whole-genome sequencing, we rapidly mapped these suppressor mutations to two small homologs of relA. Interestingly, stable suppressor strains had mutations in both genes, with each mutation alone partially relieving the relA growth defect. This supports an intriguing three-locus interaction module that is not easily identifiable through traditional suppressor mapping. We conclude that whole-genome sequencing can drastically accelerate the identification of suppressor mutations and complex genetic interactions, and it can be applied as a standard tool to investigate the genetic traits of model organisms.


Asunto(s)
Bacillus subtilis/genética , Genoma Bacteriano , Análisis de Secuencia de ADN/métodos , Bacillus subtilis/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Variación Genética , Datos de Secuencia Molecular , Fenotipo , Análisis de Secuencia de ADN/normas , Supresión Genética
12.
Curr Opin Microbiol ; 11(2): 100-5, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18359660

RESUMEN

The small nucleotides pppGpp and ppGpp (or (p)ppGpp) are rapidly synthesized in response to nutritional stress. In Escherichia coli, the enzymes RelA and SpoT are triggered by different starvation signals to produce (p)ppGpp. In many Gram-positive bacteria this is carried out by RelA and two small homologs. (p)ppGpp, along with the transcription factor DksA, has profound effects on transcription initiation in E. coli. (p)ppGpp/DksA exert differential effects on promoters by playing upon their intrinsic kinetic parameters, and by facilitating the utilization of alternative sigma factors. (p)ppGpp also regulates replication and translation. These studies highlight (p)ppGpp as a key factor in bacterial physiology that responds rapidly to diverse stresses, by shutting down growth and priming cellular defensive and adaptive processes.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Escherichia coli/fisiología , Regulación Bacteriana de la Expresión Génica , Guanosina Tetrafosfato/metabolismo , Biosíntesis de Proteínas , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Transcripción Genética
13.
Methods Mol Biol ; 1672: 43-61, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29043616

RESUMEN

Genome rearrangements underlie different human diseases including many cancers. Determining the rates at which genome rearrangements arise and isolating unique, independent genome rearrangements is critical to understanding the genes and pathways that prevent or promote genome rearrangements. Here, we describe quantitative S. cerevisiae genetic assays for measuring the rates of accumulating genome rearrangements including deletions, translocations, and broken chromosomes healed by de novo telomere addition that result in the deletion of two counter-selectable genes, CAN1 and URA3, placed in the nonessential regions of the S. cerevisiae genome. The assays also allow for the isolation of individual genome rearrangements for structural studies, and a method for analyzing genome rearrangements by next-generation DNA sequencing is provided.


Asunto(s)
Genoma Fúngico , Genómica , Recombinación Genética , Saccharomyces cerevisiae/genética , Inestabilidad Genómica , Biblioteca Genómica , Genómica/métodos , Eliminación de Secuencia , Translocación Genética , Secuenciación Completa del Genoma
14.
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
15.
Nat Commun ; 7: 11256, 2016 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-27071721

RESUMEN

Gross chromosomal rearrangements (GCRs) play an important role in human diseases, including cancer. The identity of all Genome Instability Suppressing (GIS) genes is not currently known. Here multiple Saccharomyces cerevisiae GCR assays and query mutations were crossed into arrays of mutants to identify progeny with increased GCR rates. One hundred eighty two GIS genes were identified that suppressed GCR formation. Another 438 cooperatively acting GIS genes were identified that were not GIS genes, but suppressed the increased genome instability caused by individual query mutations. Analysis of TCGA data using the human genes predicted to act in GIS pathways revealed that a minimum of 93% of ovarian and 66% of colorectal cancer cases had defects affecting one or more predicted GIS gene. These defects included loss-of-function mutations, copy-number changes associated with reduced expression, and silencing. In contrast, acute myeloid leukaemia cases did not appear to have defects affecting the predicted GIS genes.


Asunto(s)
Reordenamiento Génico/genética , Redes Reguladoras de Genes , Genoma Fúngico , Neoplasias/genética , Saccharomyces cerevisiae/genética , Cromosomas Fúngicos/genética , Elementos de Facilitación Genéticos/genética , Epistasis Genética , Genes Fúngicos , Inestabilidad Genómica , Humanos , Mutación/genética
16.
Science ; 334(6063): 1713-6, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22194578

RESUMEN

In eukaryotes, it is unknown whether mismatch repair (MMR) is temporally coupled to DNA replication and how strand-specific MMR is directed. We fused Saccharomyces cerevisiae MSH6 with cyclins to restrict the availability of the Msh2-Msh6 mismatch recognition complex to either S phase or G2/M phase of the cell cycle. The Msh6-S cyclin fusion was proficient for suppressing mutations at three loci that replicate at mid-S phase, whereas the Msh6-G2/M cyclin fusion was defective. However, the Msh6-G2/M cyclin fusion was functional for MMR at a very late-replicating region of the genome. In contrast, the heteroduplex rejection function of MMR during recombination was partially functional during both S phase and G2/M phase. These results indicate a temporal coupling of MMR, but not heteroduplex rejection, to DNA replication.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Replicación del ADN , ADN de Hongos/metabolismo , Ácidos Nucleicos Heterodúplex/metabolismo , Saccharomyces cerevisiae/metabolismo , División Celular , Ciclina B/genética , Ciclina B/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Mutación del Sistema de Lectura , Fase G2 , Genes Fúngicos , Mutación , Proteínas Recombinantes de Fusión/metabolismo , Recombinación Genética , Fase S , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Cell ; 123(5): 945-57, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16325586

RESUMEN

Formation of normal olfactory memory requires the expression of the wild-type amnesiac gene in the dorsal paired medial (DPM) neurons. Imaging the activity in the processes of DPM neurons revealed that the neurons respond when the fly is stimulated with electric shock or with any odor that was tested. Pairing odor and electric-shock stimulation increases odor-evoked calcium signals and synaptic release from DPM neurons. These memory traces form in only one of the two branches of the DPM neuron process. Moreover, trace formation requires the expression of the wild-type amnesiac gene in the DPM neurons. The cellular memory traces first appear at 30 min after conditioning and persist for at least 1 hr, a time window during which DPM neuron synaptic transmission is required for normal memory. DPM neurons are therefore "odor generalists" and form a delayed, branch-specific, and amnesiac-dependent memory trace that may guide behavior after acquisition.


Asunto(s)
Condicionamiento Clásico/fisiología , Drosophila melanogaster , Memoria/fisiología , Neuronas/metabolismo , Olfato/fisiología , Animales , Calcio/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Estimulación Eléctrica , Neuronas/citología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Odorantes , Transmisión Sináptica/fisiología , Factores de Tiempo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA