Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Genet ; 7(4): e1001362, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21490951

ABSTRACT

To better understand telomere biology in budding yeast, we have performed systematic suppressor/enhancer analyses on yeast strains containing a point mutation in the essential telomere capping gene CDC13 (cdc13-1) or containing a null mutation in the DNA damage response and telomere capping gene YKU70 (yku70Δ). We performed Quantitative Fitness Analysis (QFA) on thousands of yeast strains containing mutations affecting telomere-capping proteins in combination with a library of systematic gene deletion mutations. To perform QFA, we typically inoculate 384 separate cultures onto solid agar plates and monitor growth of each culture by photography over time. The data are fitted to a logistic population growth model; and growth parameters, such as maximum growth rate and maximum doubling potential, are deduced. QFA reveals that as many as 5% of systematic gene deletions, affecting numerous functional classes, strongly interact with telomere capping defects. We show that, while Cdc13 and Yku70 perform complementary roles in telomere capping, their genetic interaction profiles differ significantly. At least 19 different classes of functionally or physically related proteins can be identified as interacting with cdc13-1, yku70Δ, or both. Each specific genetic interaction informs the roles of individual gene products in telomere biology. One striking example is with genes of the nonsense-mediated RNA decay (NMD) pathway which, when disabled, suppress the conditional cdc13-1 mutation but enhance the null yku70Δ mutation. We show that the suppressing/enhancing role of the NMD pathway at uncapped telomeres is mediated through the levels of Stn1, an essential telomere capping protein, which interacts with Cdc13 and recruitment of telomerase to telomeres. We show that increased Stn1 levels affect growth of cells with telomere capping defects due to cdc13-1 and yku70Δ. QFA is a sensitive, high-throughput method that will also be useful to understand other aspects of microbial cell biology.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Telomere/pathology , Transcription Factors/metabolism , Gene Expression Regulation, Fungal , Models, Biological , Mutation/genetics , RNA Stability/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere-Binding Proteins/genetics , Temperature
2.
PLoS Genet ; 6(8): e1001072, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20808892

ABSTRACT

Maintenance of telomere capping is absolutely essential to the survival of eukaryotic cells. Telomere capping proteins, such as Cdc13 and POT1, are essential for the viability of budding yeast and mammalian cells, respectively. Here we identify, for the first time, three genetic modifications that allow budding yeast cells to survive without telomere capping by Cdc13. We found that simultaneous inactivation of Sgs1, Exo1, and Rad9, three DNA damage response (DDR) proteins, is sufficient to allow cell division in the absence of Cdc13. Quantitative amplification of ssDNA (QAOS) was used to show that the RecQ helicase Sgs1 plays an important role in the resection of uncapped telomeres, especially in the absence of checkpoint protein Rad9. Strikingly, simultaneous deletion of SGS1 and the nuclease EXO1, further reduces resection at uncapped telomeres and together with deletion of RAD9 permits cell survival without CDC13. Pulsed-field gel electrophoresis studies show that cdc13-1 rad9Delta sgs1Delta exo1Delta strains can maintain linear chromosomes despite the absence of telomere capping by Cdc13. However, with continued passage, the telomeres of such strains eventually become short and are maintained by recombination-based mechanisms. Remarkably, cdc13Delta rad9Delta sgs1Delta exo1Delta strains, lacking any Cdc13 gene product, are viable and can grow indefinitely. Our work has uncovered a critical role for RecQ helicases in limiting the division of cells with uncapped telomeres, and this may provide one explanation for increased tumorigenesis in human diseases associated with mutations of RecQ helicases. Our results reveal the plasticity of the telomere cap and indicate that the essential role of telomere capping is to counteract specific aspects of the DDR.


Subject(s)
Cell Cycle Proteins/metabolism , Exodeoxyribonucleases/metabolism , Microbial Viability , RecQ Helicases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Cell Cycle Proteins/genetics , Exodeoxyribonucleases/genetics , RecQ Helicases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Telomere/genetics , Telomere-Binding Proteins/genetics
3.
PLoS Genet ; 5(7): e1000552, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19593371

ABSTRACT

Polyploidy is frequent in nature and is a hallmark of cancer cells, but little is known about the strategy of DNA repair in polyploid organisms. We have studied DNA repair in the polyploid archaeon Haloferax volcanii, which contains up to 20 genome copies. We have focused on the role of Mre11 and Rad50 proteins, which are found in all domains of life and which form a complex that binds to and coordinates the repair of DNA double-strand breaks (DSBs). Surprisingly, mre11 rad50 mutants are more resistant to DNA damage than the wild-type. However, wild-type cells recover faster from DNA damage, and pulsed-field gel electrophoresis shows that DNA double-strand breaks are repaired more slowly in mre11 rad50 mutants. Using a plasmid repair assay, we show that wild-type and mre11 rad50 cells use different strategies of DSB repair. In the wild-type, Mre11-Rad50 appears to prevent the repair of DSBs by homologous recombination (HR), allowing microhomology-mediated end-joining to act as the primary repair pathway. However, genetic analysis of recombination-defective radA mutants suggests that DNA repair in wild-type cells ultimately requires HR, therefore Mre11-Rad50 merely delays this mode of repair. In polyploid organisms, DSB repair by HR is potentially hazardous, since each DNA end will have multiple partners. We show that in the polyploid archaeon H. volcanii the repair of DSBs by HR is restrained by Mre11-Rad50. The unrestrained use of HR in mre11 rad50 mutants enhances cell survival but leads to slow recovery from DNA damage, presumably due to difficulties in the resolution of DNA repair intermediates. Our results suggest that recombination might be similarly repressed in other polyploid organisms and at repetitive sequences in haploid and diploid species.


Subject(s)
Archaeal Proteins/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Haloferax volcanii/genetics , Archaeal Proteins/metabolism , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Gene Knockout Techniques , Genome, Bacterial , Mutation , Polyploidy , Recombination, Genetic , Sequence Deletion , Ultraviolet Rays
4.
Mol Biol Cell ; 20(6): 1683-94, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19158388

ABSTRACT

Esc2 is a member of the RENi family of SUMO-like domain proteins and is implicated in gene silencing in Saccharomyces cerevisiae. Here, we identify a dual role for Esc2 during S-phase in mediating both intra-S-phase DNA damage checkpoint signaling and preventing the accumulation of Rad51-dependent homologous recombination repair (HRR) intermediates. These roles are qualitatively similar to those of Sgs1, the yeast ortholog of the human Bloom's syndrome protein, BLM. However, whereas mutation of either ESC2 or SGS1 leads to the accumulation of unprocessed HRR intermediates in the presence of MMS, the accumulation of these structures in esc2 (but not sgs1) mutants is entirely dependent on Mph1, a protein that shows structural similarity to the Fanconi anemia group M protein (FANCM). In the absence of both Esc2 and Sgs1, the intra-S-phase DNA damage checkpoint response is compromised after exposure to MMS, and sgs1esc2 cells attempt to undergo mitosis with unprocessed HRR intermediates. We propose a model whereby Esc2 acts in an Mph1-dependent process, separately from Sgs1, to influence the repair/tolerance of MMS-induced lesions during S-phase.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Repair/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Recombination, Genetic/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , DEAD-box RNA Helicases/genetics , DNA Damage/genetics , Genomic Instability/genetics , Mitosis , Mutation/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RecQ Helicases/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
5.
EMBO J ; 27(18): 2400-10, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18756267

ABSTRACT

Exo1 is a nuclease involved in mismatch repair, DSB repair, stalled replication fork processing and in the DNA damage response triggered by dysfunctional telomeres. In budding yeast and mice, Exo1 creates single-stranded DNA (ssDNA) at uncapped telomeres. This ssDNA accumulation activates the checkpoint response resulting in cell cycle arrest. Here, we demonstrate that Exo1 is phosphorylated when telomeres are uncapped in cdc13-1 and yku70Delta yeast cells, and in response to the induction of DNA damage. After telomere uncapping, Exo1 phosphorylation depends on components of the checkpoint machinery such as Rad24, Rad17, Rad9, Rad53 and Mec1, but is largely independent of Chk1, Tel1 and Dun1. Serines S372, S567, S587 and S692 of Exo1 were identified as targets for phosphorylation. Furthermore, mutation of these Exo1 residues altered the DNA damage response to uncapped telomeres and camptothecin treatment, in a manner that suggests Exo1 phosphorylation inhibits its activity. We propose that Rad53-dependent Exo1 phosphorylation is involved in a negative feedback loop to limit ssDNA accumulation and DNA damage checkpoint activation.


Subject(s)
DNA Damage , Exodeoxyribonucleases/physiology , Cell Cycle , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , DNA, Single-Stranded/metabolism , Fungal Proteins/metabolism , Genome, Fungal , Genotype , Models, Biological , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Saccharomyces cerevisiae Proteins/metabolism , Telomere/metabolism , Telomere/ultrastructure , Temperature
6.
Mol Biol Cell ; 18(10): 4062-73, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17671161

ABSTRACT

CSM2, PSY3, SHU1, and SHU2 (collectively referred to as the SHU genes) were identified in Saccharomyces cerevisiae as four genes in the same epistasis group that suppress various sgs1 and top3 mutant phenotypes when mutated. Although the SHU genes have been implicated in homologous recombination repair (HRR), their precise role(s) within this pathway remains poorly understood. Here, we have identified a specific role for the Shu proteins in a Rad51/Rad54-dependent HRR pathway(s) to repair MMS-induced lesions during S-phase. We show that, although mutation of RAD51 or RAD54 prevented the formation of MMS-induced HRR intermediates (X-molecules) arising during replication in sgs1 cells, mutation of SHU genes attenuated the level of these structures. Similar findings were also observed in shu1 cells in which Rmi1 or Top3 function was impaired. We propose a model in which the Shu proteins act in HRR to promote the formation of HRR intermediates that are processed by the Sgs1-Rmi1-Top3 complex.


Subject(s)
RecQ Helicases/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cell Proliferation/drug effects , DNA Repair/drug effects , DNA-Binding Proteins , Epistasis, Genetic , Genes, Fungal , Hydroxyurea/pharmacology , Methyl Methanesulfonate/pharmacology , Models, Biological , Mutant Proteins/metabolism , Mutation/genetics , Recombination, Genetic/drug effects , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects
7.
Appl Environ Microbiol ; 70(2): 943-53, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14766575

ABSTRACT

Since most archaea are extremophilic and difficult to cultivate, our current knowledge of their biology is confined largely to comparative genomics and biochemistry. Haloferax volcanii offers great promise as a model organism for archaeal genetics, but until now there has been a lack of a wide variety of selectable markers for this organism. We describe here isolation of H. volcanii leuB and trpA genes encoding 3-isopropylmalate dehydrogenase and tryptophan synthase, respectively, and development of these genes as a positive selection system. DeltaleuB and DeltatrpA mutants were constructed in a variety of genetic backgrounds and were shown to be auxotrophic for leucine and tryptophan, respectively. We constructed both integrative and replicative plasmids carrying the leuB or trpA gene under control of a constitutive promoter. The use of these selectable markers in deletion of the lhr gene of H. volcanii is described.


Subject(s)
Alcohol Oxidoreductases/genetics , Genetic Markers , Haloferax volcanii/genetics , Selection, Genetic , Tryptophan Synthase/genetics , 3-Isopropylmalate Dehydrogenase , Alcohol Oxidoreductases/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Culture Media , Gene Deletion , Haloferax volcanii/growth & development , Haloferax volcanii/metabolism , Leucine/metabolism , Molecular Sequence Data , Plasmids , Sequence Analysis, DNA , Sodium Chloride/metabolism , Tryptophan/metabolism , Tryptophan Synthase/metabolism
8.
EMBO J ; 22(3): 735-45, 2003 Feb 03.
Article in English | MEDLINE | ID: mdl-12554673

ABSTRACT

PriA protein provides a means to load the DnaB replicative helicase at DNA replication fork and D loop structures, and is therefore a key factor in the rescue of stalled or broken forks and subsequent replication restart. We show that the nucleoid-associated RdgC protein binds non-specifically to single-stranded (ss) DNA and double-stranded DNA. It is also essential for growth of a strain lacking PriA, indicating that it might affect replication fork progression or fork rescue. dnaC suppressors of priA overcome this inviability, especially when RecF, RecO or RecR is inactivated, indicating that RdgC avoids or counters a toxic effect of these proteins. Mutations modifying ssDNA-binding (SSB) protein also negate this toxic effect, suggesting that the toxicity reflects inappropriate loading of RecA on SSB-coated ssDNA, leading to excessive or untimely RecA activity. We suggest that binding of RdgC to DNA limits RecA loading, avoiding problems at replication forks that would otherwise require PriA to promote replication restart. Mutations in RNA polymerase also reduce the toxic effect of RecFOR, providing a further link between DNA replication, transcription and repair.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , DNA/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Cell Division/physiology , Cell Size , DNA, Single-Stranded/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/genetics , Evolution, Molecular , Genes, Bacterial , Mutation , Phylogeny , Protein Binding , Replication Protein A
SELECTION OF CITATIONS
SEARCH DETAIL
...