Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Mol Cell ; 83(16): 2925-2940.e8, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37499663

ABSTRACT

Homologous recombination (HR) is essential for error-free repair of DNA double-strand breaks, perturbed replication forks (RFs), and post-replicative single-stranded DNA (ssDNA) gaps. To initiate HR, the recombination mediator and tumor suppressor protein BRCA2 facilitates nucleation of RAD51 on ssDNA prior to stimulation of RAD51 filament growth by RAD51 paralogs. Although ssDNA binding by BRCA2 has been implicated in RAD51 nucleation, the function of double-stranded DNA (dsDNA) binding by BRCA2 remains unclear. Here, we exploit single-molecule (SM) imaging to visualize BRCA2-mediated RAD51 nucleation in real time using purified proteins. We report that BRCA2 nucleates and stabilizes RAD51 on ssDNA either directly or through an unappreciated diffusion-assisted delivery mechanism involving binding to and sliding along dsDNA, which requires the cooperative action of multiple dsDNA-binding modules in BRCA2. Collectively, our work reveals two distinct mechanisms of BRCA2-dependent RAD51 loading onto ssDNA, which we propose are critical for its diverse functions in maintaining genome stability and cancer suppression.


Subject(s)
BRCA2 Protein , Rad51 Recombinase , Humans , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , DNA-Binding Proteins/metabolism , DNA, Single-Stranded/genetics , DNA/metabolism , DNA Repair , Protein Binding
2.
Nature ; 629(8012): 697-703, 2024 May.
Article in English | MEDLINE | ID: mdl-38658755

ABSTRACT

RAD52 is important for the repair of DNA double-stranded breaks1,2, mitotic DNA synthesis3-5 and alternative telomere length maintenance6,7. Central to these functions, RAD52 promotes the annealing of complementary single-stranded DNA (ssDNA)8,9 and provides an alternative to BRCA2/RAD51-dependent homologous recombination repair10. Inactivation of RAD52 in homologous-recombination-deficient BRCA1- or BRCA2-defective cells is synthetically lethal11,12, and aberrant expression of RAD52 is associated with poor cancer prognosis13,14. As a consequence, RAD52 is an attractive therapeutic target against homologous-recombination-deficient breast, ovarian and prostate cancers15-17. Here we describe the structure of RAD52 and define the mechanism of annealing. As reported previously18-20, RAD52 forms undecameric (11-subunit) ring structures, but these rings do not represent the active form of the enzyme. Instead, cryo-electron microscopy and biochemical analyses revealed that ssDNA annealing is driven by RAD52 open rings in association with replication protein-A (RPA). Atomic models of the RAD52-ssDNA complex show that ssDNA sits in a positively charged channel around the ring. Annealing is driven by the RAD52 N-terminal domains, whereas the C-terminal regions modulate the open-ring conformation and RPA interaction. RPA associates with RAD52 at the site of ring opening with critical interactions occurring between the RPA-interacting domain of RAD52 and the winged helix domain of RPA2. Our studies provide structural snapshots throughout the annealing process and define the molecular mechanism of ssDNA annealing by the RAD52-RPA complex.


Subject(s)
Cryoelectron Microscopy , DNA, Single-Stranded , Multiprotein Complexes , Rad52 DNA Repair and Recombination Protein , Replication Protein A , Humans , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/ultrastructure , Models, Molecular , Protein Binding , Rad52 DNA Repair and Recombination Protein/chemistry , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/ultrastructure , Replication Protein A/chemistry , Replication Protein A/metabolism , Replication Protein A/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Domains , Binding Sites
3.
Mol Cell ; 82(24): 4664-4680.e9, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36455556

ABSTRACT

POLQ is a key effector of DSB repair by microhomology-mediated end-joining (MMEJ) and is overexpressed in many cancers. POLQ inhibitors confer synthetic lethality in HR and Shieldin-deficient cancer cells, which has been proposed to reflect a critical dependence on the DSB repair pathway by MMEJ. Whether POLQ also operates independent of MMEJ remains unexplored. Here, we show that POLQ-deficient cells accumulate post-replicative ssDNA gaps upon BRCA1/2 loss or PARP inhibitor treatment. Biochemically, cooperation between POLQ helicase and polymerase activities promotes RPA displacement and ssDNA-gap fill-in, respectively. POLQ is also capable of microhomology-mediated gap skipping (MMGS), which generates deletions during gap repair that resemble the genomic scars prevalent in POLQ overexpressing cancers. Our findings implicate POLQ in mutagenic post-replicative gap sealing, which could drive genome evolution in cancer and whose loss places a critical dependency on HR for gap protection and repair and cellular viability.


Subject(s)
DNA Breaks, Double-Stranded , Neoplasms , Humans , DNA Replication/genetics , Genomic Instability , DNA, Single-Stranded/genetics , Synthetic Lethal Mutations , DNA End-Joining Repair , Neoplasms/genetics
4.
Nature ; 619(7970): 650-657, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344587

ABSTRACT

Homologous recombination is a fundamental process of life. It is required for the protection and restart of broken replication forks, the repair of chromosome breaks and the exchange of genetic material during meiosis. Individuals with mutations in key recombination genes, such as BRCA2 (also known as FANCD1), or the RAD51 paralogues RAD51B, RAD51C (also known as FANCO), RAD51D, XRCC2 (also known as FANCU) and XRCC3, are predisposed to breast, ovarian and prostate cancers1-10 and the cancer-prone syndrome Fanconi anaemia11-13. The BRCA2 tumour suppressor protein-the product of BRCA2-is well characterized, but the cellular functions of the RAD51 paralogues remain unclear. Genetic knockouts display growth defects, reduced RAD51 focus formation, spontaneous chromosome abnormalities, sensitivity to PARP inhibitors and replication fork defects14,15, but the precise molecular roles of RAD51 paralogues in fork stability, DNA repair and cancer avoidance remain unknown. Here we used cryo-electron microscopy, AlphaFold2 modelling and structural proteomics to determine the structure of the RAD51B-RAD51C-RAD51D-XRCC2 complex (BCDX2), revealing that RAD51C-RAD51D-XRCC2 mimics three RAD51 protomers aligned within a nucleoprotein filament, whereas RAD51B is highly dynamic. Biochemical and single-molecule analyses showed that BCDX2 stimulates the nucleation and extension of RAD51 filaments-which are essential for recombinational DNA repair-in reactions that depend on the coupled ATPase activities of RAD51B and RAD51C. Our studies demonstrate that BCDX2 orchestrates RAD51 assembly on single stranded DNA for replication fork protection and double strand break repair, in reactions that are critical for tumour avoidance.


Subject(s)
Cryoelectron Microscopy , DNA-Binding Proteins , Multiprotein Complexes , Rad51 Recombinase , Tumor Suppressor Proteins , Humans , DNA Repair , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Homologous Recombination , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Rad51 Recombinase/ultrastructure , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/ultrastructure , Poly(ADP-ribose) Polymerase Inhibitors , Neoplasms/genetics , Neoplasms/prevention & control , Proteomics , Computer Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , DNA Breaks, Double-Stranded
5.
Mol Cell ; 81(4): 767-783.e11, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33333017

ABSTRACT

Chromatin is a barrier to efficient DNA repair, as it hinders access and processing of certain DNA lesions. ALC1/CHD1L is a nucleosome-remodeling enzyme that responds to DNA damage, but its precise function in DNA repair remains unknown. Here we report that loss of ALC1 confers sensitivity to PARP inhibitors, methyl-methanesulfonate, and uracil misincorporation, which reflects the need to remodel nucleosomes following base excision by DNA glycosylases but prior to handover to APEX1. Using CRISPR screens, we establish that ALC1 loss is synthetic lethal with homologous recombination deficiency (HRD), which we attribute to chromosome instability caused by unrepaired DNA gaps at replication forks. In the absence of ALC1 or APEX1, incomplete processing of BER intermediates results in post-replicative DNA gaps and a critical dependence on HR for repair. Hence, targeting ALC1 alone or as a PARP inhibitor sensitizer could be employed to augment existing therapeutic strategies for HRD cancers.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , DNA Helicases/genetics , DNA Replication/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , Homologous Recombination/drug effects , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Nucleosomes/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
6.
Cell ; 147(1): 158-72, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21962513

ABSTRACT

The efficient and timely resolution of DNA recombination intermediates is essential for bipolar chromosome segregation. Here, we show that the specialized chromosome segregation patterns of meiosis and mitosis, which require the coordination of recombination with cell-cycle progression, are achieved by regulating the timing of activation of two crossover-promoting endonucleases. In yeast meiosis, Mus81-Mms4 and Yen1 are controlled by phosphorylation events that lead to their sequential activation. Mus81-Mms4 is hyperactivated by Cdc5-mediated phosphorylation in meiosis I, generating the crossovers necessary for chromosome segregation. Yen1 is also tightly regulated and is activated in meiosis II to resolve persistent Holliday junctions. In yeast and human mitotic cells, a similar regulatory network restrains these nuclease activities until mitosis, biasing the outcome of recombination toward noncrossover products while also ensuring the elimination of any persistent joint molecules. Mitotic regulation thereby facilitates chromosome segregation while limiting the potential for loss of heterozygosity and sister-chromatid exchanges.


Subject(s)
DNA, Cruciform , Meiosis , Mitosis , Recombination, Genetic , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Cell Cycle , Crossing Over, Genetic , HeLa Cells , Holliday Junction Resolvases/metabolism , Humans , Saccharomyces cerevisiae/enzymology
7.
Nature ; 586(7828): 292-298, 2020 10.
Article in English | MEDLINE | ID: mdl-32999459

ABSTRACT

The RecQ DNA helicase WRN is a synthetic lethal target for cancer cells with microsatellite instability (MSI), a form of genetic hypermutability that arises from impaired mismatch repair1-4. Depletion of WRN induces widespread DNA double-strand breaks in MSI cells, leading to cell cycle arrest and/or apoptosis. However, the mechanism by which WRN protects MSI-associated cancers from double-strand breaks remains unclear. Here we show that TA-dinucleotide repeats are highly unstable in MSI cells and undergo large-scale expansions, distinct from previously described insertion or deletion mutations of a few nucleotides5. Expanded TA repeats form non-B DNA secondary structures that stall replication forks, activate the ATR checkpoint kinase, and require unwinding by the WRN helicase. In the absence of WRN, the expanded TA-dinucleotide repeats are susceptible to cleavage by the MUS81 nuclease, leading to massive chromosome shattering. These findings identify a distinct biomarker that underlies the synthetic lethal dependence on WRN, and support the development of therapeutic agents that target WRN for MSI-associated cancers.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repeat Expansion/genetics , Dinucleotide Repeats/genetics , Neoplasms/genetics , Werner Syndrome Helicase/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Chromothripsis , DNA Cleavage , DNA Replication , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/metabolism , Genomic Instability , Humans , Recombinases/metabolism
8.
Cell ; 142(1): 65-76, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603015

ABSTRACT

DNA interstrand crosslinks (ICLs) are highly toxic because they block the progression of replisomes. The Fanconi Anemia (FA) proteins, encoded by genes that are mutated in FA, are important for repair of ICLs. The FA core complex catalyzes the monoubiquitination of FANCD2, and this event is essential for several steps of ICL repair. However, how monoubiquitination of FANCD2 promotes ICL repair at the molecular level is unknown. Here, we describe a highly conserved protein, KIAA1018/MTMR15/FAN1, that interacts with, and is recruited to sites of DNA damage by, the monoubiquitinated form of FANCD2. FAN1 exhibits endonuclease activity toward 5' flaps and has 5' exonuclease activity, and these activities are mediated by an ancient VRR_nuc domain. Depletion of FAN1 from human cells causes hypersensitivity to ICLs, defects in ICL repair, and genome instability. These data at least partly explain how ubiquitination of FANCD2 promotes DNA repair.


Subject(s)
DNA Repair , Exodeoxyribonucleases/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins , BRCA2 Protein/metabolism , Cell Line , Cross-Linking Reagents/pharmacology , DNA Damage/drug effects , Endodeoxyribonucleases , Endonucleases/chemistry , Endonucleases/metabolism , Exodeoxyribonucleases/chemistry , Humans , Molecular Sequence Data , Multifunctional Enzymes , Protein Structure, Tertiary , Sequence Alignment , Ubiquitination
9.
Mol Cell ; 65(5): 848-860.e11, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28257701

ABSTRACT

The efficient removal of replication and recombination intermediates is essential for the maintenance of genome stability. Resolution of these potentially toxic structures requires the MUS81-EME1 endonuclease, which is activated at prometaphase by formation of the SMX tri-nuclease containing three DNA repair structure-selective endonucleases: SLX1-SLX4, MUS81-EME1, and XPF-ERCC1. Here we show that SMX tri-nuclease is more active than the three individual nucleases, efficiently cleaving replication forks and recombination intermediates. Within SMX, SLX4 co-ordinates the SLX1 and MUS81-EME1 nucleases for Holliday junction resolution, in a reaction stimulated by XPF-ERCC1. SMX formation activates MUS81-EME1 for replication fork and flap structure cleavage by relaxing substrate specificity. Activation involves MUS81's conserved N-terminal HhH domain, which mediates incision site selection and SLX4 binding. Cell cycle-dependent formation and activation of this tri-nuclease complex provides a unique mechanism by which cells ensure chromosome segregation and preserve genome integrity.


Subject(s)
DNA Repair , DNA Replication , DNA/biosynthesis , Endonucleases/metabolism , Genomic Instability , Cell Cycle , DNA/chemistry , DNA/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Enzyme Activation , Humans , Multienzyme Complexes , Nucleic Acid Conformation , Protein Binding , Protein Interaction Domains and Motifs , Recombinases/genetics , Recombinases/metabolism , Structure-Activity Relationship , Time Factors
10.
Proc Natl Acad Sci U S A ; 119(18): e2123420119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35452329

ABSTRACT

Four-way DNA intermediates, also known as Holliday junctions (HJs), are formed during homologous recombination and DNA repair, and their resolution is necessary for proper chromosome segregation. To facilitate the biochemical analysis of HJ processing, we developed a method involving DNAzyme self-cleavage to generate 1.8-kb DNA molecules containing either single (sHJ) or double Holliday junctions (dHJs). We show that dHJ DNAs (referred to as HoJo DNAs) are dissolved by the human BLM­TopIIIα­RMI1­RMI2 complex to form two noncrossover products. However, structure-selective endonucleases (human GEN1 and SMX complex) resolve DNA containing single or double HJs to yield a mixture of crossover and noncrossover products. Finally, we demonstrate that chromatin inhibits the resolution of the double HJ by GEN or SMX while allowing BTRR-mediated dissolution.


Subject(s)
Chromatin , DNA, Cruciform , Chromatin/genetics , Chromosomes , DNA/genetics , DNA, Cruciform/genetics , Solubility
11.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35042798

ABSTRACT

Mutations in the SETX gene, which encodes Senataxin, are associated with the progressive neurodegenerative diseases ataxia with oculomotor apraxia 2 (AOA2) and amyotrophic lateral sclerosis 4 (ALS4). To identify the causal defect in AOA2, patient-derived cells and SETX knockouts (human and mouse) were analyzed using integrated genomic and transcriptomic approaches. A genome-wide increase in chromosome instability (gains and losses) within genes and at chromosome fragile sites was observed, resulting in changes to gene-expression profiles. Transcription stress near promoters correlated with high GCskew and the accumulation of R-loops at promoter-proximal regions, which localized with chromosomal regions where gains and losses were observed. In the absence of Senataxin, the Cockayne syndrome protein CSB was required for the recruitment of the transcription-coupled repair endonucleases (XPG and XPF) and RAD52 recombination protein to target and resolve transcription bubbles containing R-loops, leading to genomic instability. These results show that transcription stress is an important contributor to SETX mutation-associated chromosome fragility and AOA2.


Subject(s)
Chromosomal Instability/genetics , DNA Helicases/metabolism , Multifunctional Enzymes/metabolism , RNA Helicases/metabolism , Spinocerebellar Ataxias/congenital , Animals , Apraxias/genetics , Ataxia/genetics , Cell Line , Cerebellar Ataxia/genetics , DNA Helicases/genetics , DNA Repair/genetics , Gene Expression Profiling/methods , Genomic Instability/genetics , Genomics/methods , Humans , Mice , Mouse Embryonic Stem Cells , Multifunctional Enzymes/genetics , Mutation/genetics , Neurodegenerative Diseases/genetics , Primary Cell Culture , Promoter Regions, Genetic/genetics , RNA Helicases/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Transcriptome/genetics
12.
Hum Mol Genet ; 31(17): 2899-2917, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35394024

ABSTRACT

Cellular proliferation depends on the accurate and timely replication of the genome. Several genetic diseases are caused by mutations in key DNA replication genes; however, it remains unclear whether these genes influence the normal program of DNA replication timing. Similarly, the factors that regulate DNA replication dynamics are poorly understood. To systematically identify trans-acting modulators of replication timing, we profiled replication in 184 cell lines from three cell types, encompassing 60 different gene knockouts or genetic diseases. Through a rigorous approach that considers the background variability of replication timing, we concluded that most samples displayed normal replication timing. However, mutations in two genes showed consistently abnormal replication timing. The first gene was RIF1, a known modulator of replication timing. The second was MCM10, a highly conserved member of the pre-replication complex. Cells from a single patient carrying MCM10 mutations demonstrated replication timing variability comprising 46% of the genome and at different locations than RIF1 knockouts. Replication timing alterations in the mutated MCM10 cells were predominantly comprised of replication delays and initiation site gains and losses. Taken together, this study demonstrates the remarkable robustness of the human replication timing program and reveals MCM10 as a novel candidate modulator of DNA replication timing.


Subject(s)
DNA Replication Timing , Minichromosome Maintenance Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Replication/genetics , DNA Replication Timing/genetics , Humans , Minichromosome Maintenance Proteins/genetics , Replication Origin
13.
Crit Rev Biochem Mol Biol ; 56(2): 157-177, 2021 04.
Article in English | MEDLINE | ID: mdl-33596761

ABSTRACT

SLX4 provides a molecular scaffold for the assembly of multiple protein complexes required for the maintenance of genome stability. It is involved in the repair of DNA crosslinks, the resolution of recombination intermediates, the response to replication stress and the maintenance of telomere length. To carry out these diverse functions, SLX4 interacts with three structure-selective endonucleases, MUS81-EME1, SLX1 and XPF-ERCC1, as well as the telomere binding proteins TRF2, RTEL1 and SLX4IP. Recently, SLX4 was shown to interact with MutSß, a heterodimeric protein involved in DNA mismatch repair, trinucleotide repeat instability, crosslink repair and recombination. Importantly, MutSß promotes the pathogenic expansion of CAG/CTG trinucleotide repeats, which is causative of myotonic dystrophy and Huntington's disease. The colocalization and specific interaction of MutSß with SLX4, together with their apparently overlapping functions, are suggestive of a common role in reactions that promote DNA maintenance and genome stability. This review will focus on the role of SLX4 in DNA repair, the interplay between MutSß and SLX4, and detail how they cooperate to promote recombinational repair and DNA crosslink repair. Furthermore, we speculate that MutSß and SLX4 may provide an alternative cellular mechanism that modulates trinucleotide instability.


Subject(s)
DNA Repair , Genomic Instability , MutS Homolog 3 Protein/metabolism , Recombinases/metabolism , Animals , DNA Damage , Humans , MutS Homolog 3 Protein/genetics , Neoplasms/genetics , Neoplasms/metabolism , Protein Interaction Maps , Recombinases/genetics
14.
Anal Biochem ; 682: 115347, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37821038

ABSTRACT

DNA molecules that contain single Holliday junctions have served as model substrates to investigate the pathway in which homologous recombination intermediates are processed. However, the preparation of DNA containing Holliday junctions in high yield remains a challenge. In this work, we used a nicking endonuclease to generate gapped DNA, from which α-structured DNA or figure-8 DNA were created via RecA-mediated reactions. The resulting DNA molecules were found to serve as good substrates for Holliday junction resolvases. The simplified method negates the requirement for radioactive labelling of DNA, making the generation of Holliday junction DNA more accessible to non-experts.


Subject(s)
DNA, Cruciform , Escherichia coli Proteins , DNA, Cruciform/metabolism , Escherichia coli Proteins/chemistry , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/genetics , DNA/chemistry
15.
Cell ; 135(2): 261-71, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18957201

ABSTRACT

Homologous recombination (HR) is an important conserved process for DNA repair and ensures maintenance of genome integrity. Inappropriate HR causes gross chromosomal rearrangements and tumorigenesis in mammals. In yeast, the Srs2 helicase eliminates inappropriate recombination events, but the functional equivalent of Srs2 in higher eukaryotes has been elusive. Here, we identify C. elegans RTEL-1 as a functional analog of Srs2 and describe its vertebrate counterpart, RTEL1, which is required for genome stability and tumor avoidance. We find that rtel-1 mutant worms and RTEL1-depleted human cells share characteristic phenotypes with yeast srs2 mutants: lethality upon deletion of the sgs1/BLM homolog, hyperrecombination, and DNA damage sensitivity. In vitro, purified human RTEL1 antagonizes HR by promoting the disassembly of D loop recombination intermediates in a reaction dependent upon ATP hydrolysis. We propose that loss of HR control after deregulation of RTEL1 may be a critical event that drives genome instability and cancer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , DNA Helicases/metabolism , Genomic Instability , Recombination, Genetic , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DNA/metabolism , DNA Helicases/genetics , DNA Repair , Humans , Mutation , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism
16.
Mol Cell ; 54(1): 94-106, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24631285

ABSTRACT

The careful orchestration of cellular events such as DNA replication, repair, and segregation is essential for equal distribution of the duplicated genome into two daughter cells. To ensure that persistent recombination intermediates are resolved prior to cell division, the Yen1 Holliday junction resolvase is activated at anaphase. Here, we show that the master cell-cycle regulators, cyclin-dependent kinase (Cdk) and Cdc14 phosphatase, control the actions of Yen1. During S phase, Cdk-mediated phosphorylation of Yen1 promotes its nuclear exclusion and inhibits catalytic activity by reducing the efficiency of DNA binding. Later in the cell cycle, at anaphase, Cdc14 drives Yen1 dephosphorylation, leading to its nuclear relocalization and enzymatic activation. Using a constitutively activated form of Yen1, we show that uncontrolled Yen1 activity is detrimental to the cell: spatial and temporal restriction of Yen1 protects against genotoxic stress and, by avoiding competition with the noncrossover-promoting repair pathways, prevents loss of heterozygosity.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/enzymology , Cyclin-Dependent Kinases/metabolism , Genomic Instability , Holliday Junction Resolvases/metabolism , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Active Transport, Cell Nucleus , Anaphase , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinases/genetics , DNA Damage , DNA Repair , Enzyme Activation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Holliday Junction Resolvases/genetics , Loss of Heterozygosity , Mutation , Phosphorylation , Protein Tyrosine Phosphatases/genetics , S Phase , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
17.
Genes Dev ; 28(10): 1124-36, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24831703

ABSTRACT

The resolution of recombination intermediates containing Holliday junctions (HJs) is critical for genome maintenance and proper chromosome segregation. Three pathways for HJ processing exist in human cells and involve the following enzymes/complexes: BLM-TopoIIIα-RMI1-RMI2 (BTR complex), SLX1-SLX4-MUS81-EME1 (SLX-MUS complex), and GEN1. Cycling cells preferentially use the BTR complex for the removal of double HJs in S phase, with SLX-MUS and GEN1 acting at temporally distinct phases of the cell cycle. Cells lacking SLX-MUS and GEN1 exhibit chromosome missegregation, micronucleus formation, and elevated levels of 53BP1-positive G1 nuclear bodies, suggesting that defects in chromosome segregation lead to the transmission of extensive DNA damage to daughter cells. In addition, however, we found that the effects of SLX4, MUS81, and GEN1 depletion extend beyond mitosis, since genome instability is observed throughout all phases of the cell cycle. This is exemplified in the form of impaired replication fork movement and S-phase progression, endogenous checkpoint activation, chromosome segmentation, and multinucleation. In contrast to SLX4, SLX1, the nuclease subunit of the SLX1-SLX4 structure-selective nuclease, plays no role in the replication-related phenotypes associated with SLX4/MUS81 and GEN1 depletion. These observations demonstrate that the SLX1-SLX4 nuclease and the SLX4 scaffold play divergent roles in the maintenance of genome integrity in human cells.


Subject(s)
Genomic Instability/physiology , Mitosis/physiology , Anaphase , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Centromere/metabolism , Chromosome Aberrations , Chromosomes/enzymology , DNA Breaks , Genomic Instability/genetics , HeLa Cells , Humans , Indoles/metabolism , Micronuclei, Chromosome-Defective , Mitosis/genetics , Recombinases/metabolism , Replication Origin/genetics
18.
Mol Cell ; 52(2): 234-47, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24076221

ABSTRACT

Holliday junctions (HJs) are four-way DNA intermediates that form during homologous recombination, and their efficient resolution is essential for chromosome segregation. Here, we show that three structure-selective endonucleases, namely SLX1-SLX4, MUS81-EME1, and GEN1, define two pathways of HJ resolution in human cells. One pathway is mediated by GEN1, whereas SLX1-SLX4 and MUS81-EME1 provide a second and genetically distinct pathway (SLX-MUS). Cells depleted for SLX-MUS or GEN1 pathway proteins exhibit severe defects in chromosome segregation and reduced survival. In response to CDK-mediated phosphorylation, SLX1-SLX4 and MUS81-EME1 associate at the G2/M transition to form a stable SLX-MUS holoenzyme, which can be reconstituted in vitro. Biochemical studies show that SLX-MUS is a HJ resolvase that coordinates the active sites of two distinct endonucleases during HJ resolution. This cleavage reaction is more efficient and orchestrated than that mediated by SLX1-SLX4 alone, which exhibits a potent nickase activity that acts promiscuously upon DNA secondary structures.


Subject(s)
DNA, Cruciform , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/metabolism , Recombinases/metabolism , Base Sequence , Cell Line, Transformed , DNA Repair , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Endonucleases/genetics , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/genetics , HeLa Cells , Holliday Junction Resolvases/genetics , Holliday Junction Resolvases/metabolism , Humans , Immunoblotting , Models, Genetic , Oligonucleotides/genetics , Oligonucleotides/metabolism , Protein Binding , RNA Interference , Recombinases/genetics , Sister Chromatid Exchange , Substrate Specificity
19.
Proc Natl Acad Sci U S A ; 114(3): 443-450, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049850

ABSTRACT

Genetic recombination provides an important mechanism for the repair of DNA double-strand breaks. Homologous pairing and strand exchange lead to the formation of DNA intermediates, in which sister chromatids or homologous chromosomes are covalently linked by four-way Holliday junctions (HJs). Depending on the type of recombination reaction that takes place, intermediates may have single or double HJs, and their resolution is essential for proper chromosome segregation. In mitotic cells, double HJs are primarily dissolved by the BLM helicase-TopoisomeraseIIIα-RMI1-RMI2 (BTR) complex, whereas single HJs (and double HJs that have escaped the attention of BTR) are resolved by structure-selective endonucleases known as HJ resolvases. These enzymes are ubiquitous in nature, because they are present in bacteriophage, bacteria, archaea, and simple and complex eukaryotes. The human HJ resolvase GEN1 is a member of the XPG/Rad2 family of 5'-flap endonucleases. Biochemical studies of GEN1 revealed that it cleaves synthetic DNA substrates containing a single HJ by a mechanism similar to that shown by the prototypic HJ resolvase, Escherichia coli RuvC protein, but it is unclear whether these substrates fully recapitulate the properties of recombination intermediates that arise within a physiological context. Here, we show that GEN1 efficiently cleaves both single and double HJs contained within large recombination intermediates. Moreover, we find that GEN1 exhibits a weak sequence preference for incision between two G residues that reside in a T-rich region of DNA. These results contrast with those obtained with RuvC, which exhibits a strict requirement for the consensus sequence 5'-A/TTTG/C-3'.


Subject(s)
DNA, Cruciform/genetics , DNA, Cruciform/metabolism , Holliday Junction Resolvases/metabolism , Base Sequence , DNA Repair , DNA, Cruciform/chemistry , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Holliday Junction Resolvases/chemistry , Homologous Recombination , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL