Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Mol Cell ; 84(4): 640-658.e10, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266639

ABSTRACT

The Bloom syndrome helicase BLM interacts with topoisomerase IIIα (TOP3A), RMI1, and RMI2 to form the BTR complex, which dissolves double Holliday junctions and DNA replication intermediates to promote sister chromatid disjunction before cell division. In its absence, structure-specific nucleases like the SMX complex (comprising SLX1-SLX4, MUS81-EME1, and XPF-ERCC1) can cleave joint DNA molecules instead, but cells deficient in both BTR and SMX are not viable. Here, we identify a negative genetic interaction between BLM loss and deficiency in the BRCA1-BARD1 tumor suppressor complex. We show that this is due to a previously overlooked role for BARD1 in recruiting SLX4 to resolve DNA intermediates left unprocessed by BLM in the preceding interphase. Consequently, cells with defective BLM and BRCA1-BARD1 accumulate catastrophic levels of chromosome breakage and micronucleation, leading to cell death. Thus, we reveal mechanistic insights into SLX4 recruitment to DNA lesions, with potential clinical implications for treating BRCA1-deficient tumors.


Subject(s)
DNA-Binding Proteins , Recombinases , Humans , DNA/genetics , DNA Repair , DNA Replication , DNA, Cruciform , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Recombinases/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism
2.
Mol Cell ; 83(10): 1640-1658.e9, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37059091

ABSTRACT

SLX4, disabled in the Fanconi anemia group P, is a scaffolding protein that coordinates the action of structure-specific endonucleases and other proteins involved in the replication-coupled repair of DNA interstrand cross-links. Here, we show that SLX4 dimerization and SUMO-SIM interactions drive the assembly of SLX4 membraneless compartments in the nucleus called condensates. Super-resolution microscopy reveals that SLX4 forms chromatin-bound clusters of nanocondensates. We report that SLX4 compartmentalizes the SUMO-RNF4 signaling pathway. SENP6 and RNF4 regulate the assembly and disassembly of SLX4 condensates, respectively. SLX4 condensation per se triggers the selective modification of proteins by SUMO and ubiquitin. Specifically, SLX4 condensation induces ubiquitylation and chromatin extraction of topoisomerase 1 DNA-protein cross-links. SLX4 condensation also induces the nucleolytic degradation of newly replicated DNA. We propose that the compartmentalization of proteins by SLX4 through site-specific interactions ensures the spatiotemporal control of protein modifications and nucleolytic reactions during DNA repair.


Subject(s)
DNA Repair , Ubiquitin , Ubiquitination , Ubiquitin/metabolism , DNA/metabolism , Chromatin
3.
Genes Dev ; 34(19-20): 1392-1405, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32883681

ABSTRACT

TRF1 facilitates the replication of telomeric DNA in part by recruiting the BLM helicase, which can resolve G-quadruplexes on the lagging-strand template. Lagging-strand telomeres lacking TRF1 or BLM form fragile telomeres-structures that resemble common fragile sites (CFSs)-but how they are formed is not known. We report that analogous to CFSs, fragile telomeres in BLM-deficient cells involved double-strand break (DSB) formation, in this case by the SLX4/SLX1 nuclease. The DSBs were repaired by POLD3/POLD4-dependent break-induced replication (BIR), resulting in fragile telomeres containing conservatively replicated DNA. BIR also promoted fragile telomere formation in cells with FokI-induced telomeric DSBs and in alternative lengthening of telomeres (ALT) cells, which have spontaneous telomeric damage. BIR of telomeric DSBs competed with PARP1-, LIG3-, and XPF-dependent alternative nonhomologous end joining (alt-NHEJ), which did not generate fragile telomeres. Collectively, these findings indicate that fragile telomeres can arise from BIR-mediated repair of telomeric DSBs.


Subject(s)
Chromosome Fragile Sites/genetics , DNA Breaks, Double-Stranded , DNA Replication , RecQ Helicases/genetics , RecQ Helicases/metabolism , Telomere/pathology , Animals , Cell Line , Cells, Cultured , DNA Repair , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Fibroblasts , Humans , Mice , Recombinases/genetics , Recombinases/metabolism
4.
Mol Cell ; 76(1): 27-43.e11, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31447390

ABSTRACT

Cancer cells acquire unlimited proliferative capacity by either re-expressing telomerase or inducing alternative lengthening of telomeres (ALT), which relies on telomere recombination. Here, we show that ALT recombination requires coordinate regulation of the SMX and BTR complexes to ensure the appropriate balance of resolution and dissolution activities at recombining telomeres. Critical to this control is SLX4IP, which accumulates at ALT telomeres and interacts with SLX4, XPF, and BLM. Loss of SLX4IP increases ALT-related phenotypes, which is incompatible with cell growth following concomitant loss of SLX4. Inactivation of BLM is sufficient to rescue telomere aggregation and the synthetic growth defect in this context, suggesting that SLX4IP favors SMX-dependent resolution by antagonizing promiscuous BLM activity during ALT recombination. Finally, we show that SLX4IP is inactivated in a subset of ALT-positive osteosarcomas. Collectively, our findings uncover an SLX4IP-dependent regulatory mechanism critical for telomere maintenance in ALT cancer cells.


Subject(s)
Bone Neoplasms/enzymology , Carrier Proteins/metabolism , Osteosarcoma/enzymology , RecQ Helicases/metabolism , Telomere Homeostasis , Telomere/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carrier Proteins/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mice, Knockout , Mice, SCID , Osteosarcoma/genetics , Osteosarcoma/pathology , Protein Binding , Protein Interaction Domains and Motifs , RecQ Helicases/genetics , Recombinases/genetics , Recombinases/metabolism , Signal Transduction , Telomere/genetics , Telomere/pathology
5.
Mol Cell ; 75(2): 238-251.e5, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348879

ABSTRACT

BRCT domains support myriad protein-protein interactions involved in genome maintenance. Although di-BRCT recognition of phospho-proteins is well known to support the genotoxic response, whether multi-BRCT domains can acquire distinct structures and functions is unclear. Here we present the tetra-BRCT structures from the conserved yeast protein Rtt107 in free and ligand-bound forms. The four BRCT repeats fold into a tetrahedral structure that recognizes unmodified ligands using a bi-partite mechanism, suggesting repeat origami enabling function acquisition. Functional studies show that Rtt107 binding of partner proteins of diverse activities promotes genome replication and stability in both distinct and concerted manners. A unified theme is that tetra- and di-BRCT domains of Rtt107 collaborate to recruit partner proteins to chromatin. Our work thus illustrates how a master regulator uses two types of BRCT domains to recognize distinct genome factors and direct them to chromatin for constitutive genome protection.


Subject(s)
Genomic Instability/genetics , Nuclear Proteins/ultrastructure , Protein Interaction Domains and Motifs/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/genetics , Chromatin/genetics , DNA Damage/genetics , Ligands , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Domains/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
6.
Mol Cell ; 75(4): 859-874.e4, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31351878

ABSTRACT

Homologous recombination (HR) is essential for high-fidelity DNA repair during mitotic proliferation and meiosis. Yet, context-specific modifications must tailor the recombination machinery to avoid (mitosis) or enforce (meiosis) the formation of reciprocal exchanges-crossovers-between recombining chromosomes. To obtain molecular insight into how crossover control is achieved, we affinity purified 7 DNA-processing enzymes that channel HR intermediates into crossovers or noncrossovers from vegetative cells or cells undergoing meiosis. Using mass spectrometry, we provide a global characterization of their composition and reveal mitosis- and meiosis-specific modules in the interaction networks. Functional analyses of meiosis-specific interactors of MutLγ-Exo1 identified Rtk1, Caf120, and Chd1 as regulators of crossing-over. Chd1, which transiently associates with Exo1 at the prophase-to-metaphase I transition, enables the formation of MutLγ-dependent crossovers through its conserved ability to bind and displace nucleosomes. Thus, rewiring of the HR network, coupled to chromatin remodeling, promotes context-specific control of the recombination outcome.


Subject(s)
Crossing Over, Genetic/physiology , Meiosis/physiology , Mitosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Mass Spectrometry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
Genes Dev ; 33(3-4): 221-235, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30692206

ABSTRACT

Approximately 15% of cancers use homologous recombination for alternative lengthening of telomeres (ALT). How the initiating genomic lesions invoke homology-directed telomere synthesis remains enigmatic. Here, we show that distinct dependencies exist for telomere synthesis in response to replication stress or DNA double-strand breaks (DSBs). RAD52 deficiency reduced spontaneous telomeric DNA synthesis and replication stress-associated recombination in G2, concomitant with telomere shortening and damage. However, viability and proliferation remained unaffected, suggesting that alternative telomere recombination mechanisms compensate in the absence of RAD52. In agreement, RAD52 was dispensable for DSB-induced telomere synthesis. Moreover, a targeted CRISPR screen revealed that loss of the structure-specific endonuclease scaffold SLX4 reduced the proliferation of RAD52-null ALT cells. While SLX4 was dispensable for RAD52-mediated ALT telomere synthesis in G2, combined SLX4 and RAD52 loss resulted in elevated telomere loss, unresolved telomere recombination intermediates, and mitotic infidelity. These findings establish that RAD52 and SLX4 mediate distinct postreplicative DNA repair processes that maintain ALT telomere stability and cancer cell viability.


Subject(s)
Rad52 DNA Repair and Recombination Protein/metabolism , Recombinases/metabolism , Telomere Homeostasis/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , Gene Knockout Techniques , Genomic Instability/genetics , HEK293 Cells , HeLa Cells , Humans , Interphase , Rad52 DNA Repair and Recombination Protein/genetics , Recombinases/genetics
8.
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
9.
Semin Cell Dev Biol ; 113: 57-64, 2021 05.
Article in English | MEDLINE | ID: mdl-32912640

ABSTRACT

TopBP1/Rad4/Dpb11 is an essential eukaryotic protein with important roles in DNA replication, DNA repair, DNA damage checkpoint activation, and chromosome segregation. TopBP1 serves as a scaffold to assemble protein complexes in a phosphorylation-dependent manner via its multiple BRCT-repeats. Recently, it has become clear that TopBP1 is repurposed to scaffold different processes dependent on cell cycle regulated changes in phosphorylation of client proteins. Here we review the functions of human TopBP1 in maintaining genome integrity during mitosis.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Genomics/methods , Mitosis/genetics , Nuclear Proteins/genetics , Humans
10.
J Proteome Res ; 22(6): 1660-1681, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37071664

ABSTRACT

The DNA repair scaffold SLX4 has pivotal roles in cellular processes that maintain genome stability, most notably homologous recombination. Germline mutations in SLX4 are associated with Fanconi anemia, a disease characterized by chromosome instability and cancer susceptibility. The role of mammalian SLX4 in homologous recombination depends critically on binding and activating structure-selective endonucleases, namely SLX1, MUS81-EME1, and XPF-ERCC1. Increasing evidence indicates that cells rely on distinct SLX4-dependent complexes to remove DNA lesions in specific regions of the genome. Despite our understanding of SLX4 as a scaffold for DNA repair proteins, a detailed repertoire of SLX4 interactors has never been reported. Here, we provide a comprehensive map of the human SLX4 interactome using proximity-dependent biotin identification (BioID) and affinity purification coupled to mass spectrometry (AP-MS). We identified 221 unique high-confidence interactors, of which the vast majority represent novel SLX4-binding proteins. Network analysis of these hits revealed pathways with known involvement of SLX4, such as DNA repair, and several emerging pathways of interest, including RNA metabolism and chromatin remodeling. In summary, the comprehensive SLX4 interactome we report here provides a deeper understanding of how SLX4 functions in DNA repair while revealing new cellular processes that may involve SLX4.


Subject(s)
DNA Repair , DNA-Binding Proteins , Animals , Humans , DNA-Binding Proteins/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , DNA/genetics , Homologous Recombination , Mammals/genetics , Mammals/metabolism , Recombinases/chemistry , Recombinases/genetics , Recombinases/metabolism
11.
Reprod Domest Anim ; 58(5): 679-687, 2023 May.
Article in English | MEDLINE | ID: mdl-36880652

ABSTRACT

Cattle-yak, the hybrid offspring of yak (Bos grunniens) and cattle (Bos taurus), serves as a unique model to dissect the molecular mechanisms underlying reproductive isolation. While female cattle-yaks are fertile, the males are completely sterile due to spermatogenic arrest at the meiosis stage and massive germ cell apoptosis. Interestingly, meiotic defects are partially rescued in the testes of backcrossed offspring. The genetic basis of meiotic defects in male cattle-yak remains unclear. Structure-specific endonuclease subunit (SLX4) participates in meiotic double-strand break (DSB) formation in mice, and its deletion results in defects in spermatogenesis. In the present study, we examined the expression patterns of SLX4 in the testes of yak, cattle-yak, and backcrossed offspring to investigate its potential roles in hybrid sterility. The results showed that the relative abundances of SLX4 mRNA and protein were significantly reduced in the testis of cattle-yak. The results of immunohistochemistry revealed that SLX4 was predominately expressed in spermatogonia and spermatocytes. Chromosome spreading experiments showed that SLX4 was significantly decreased in the pachytene spermatocytes of cattle-yak compared with yak and backcrossed offspring. These findings suggest that SLX4 expression was dysregulated in the testis of cattle-yak, potentially resulting in the failure of crossover formation and collapses of meiosis in hybrid males.


Subject(s)
Cattle Diseases , Infertility, Male , Animals , Cattle , Female , Male , Mice , Cattle Diseases/metabolism , Infertility, Male/veterinary , Spermatocytes , Spermatogenesis/genetics , Spermatogonia , Testis/metabolism , Recombinases/metabolism
12.
EMBO J ; 36(19): 2907-2919, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28877996

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomere lengthening pathway that predominates in aggressive tumors of mesenchymal origin; however, the underlying mechanism of telomere synthesis is not fully understood. Here, we show that the BLM-TOP3A-RMI (BTR) dissolvase complex is required for ALT-mediated telomere synthesis. We propose that recombination intermediates formed during strand invasion are processed by the BTR complex, initiating rapid and extensive POLD3-dependent telomere synthesis followed by dissolution, with no overall exchange of telomeric DNA. This process is counteracted by the SLX4-SLX1-ERCC4 complex, which promotes resolution of the recombination intermediate, resulting in telomere exchange in the absence of telomere extension. Our data are consistent with ALT being a conservative DNA replication process, analogous to break-induced replication, which is dependent on BTR and counteracted by SLX4 complex-mediated resolution events.


Subject(s)
DNA Replication/genetics , RecQ Helicases/physiology , Recombinases/physiology , Recombination, Genetic/genetics , Telomere Homeostasis/genetics , Cells, Cultured , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/physiology , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/physiology , Humans , Multienzyme Complexes/metabolism , Multienzyme Complexes/physiology , RecQ Helicases/metabolism , Recombinases/metabolism , Telomere/metabolism
13.
FEMS Yeast Res ; 21(2)2021 03 18.
Article in English | MEDLINE | ID: mdl-33606011

ABSTRACT

SLX4 is a scaffold to coordinate the action of structure-specific endonucleases that are required for homologous recombination and DNA repair. In view of ScSLX4 functions in the maintenance and stability of the genome in Saccharomyces cerevisiae, we have explored the roles of CaSLX4 in Candida albicans. Here, we constructed slx4Δ/Δ mutant and found that it exhibited increased sensitivity to the DNA damaging agent, methyl methanesulfonate (MMS) but not the DNA replication inhibitor, hydroxyurea (HU). Accordingly, RT-qPCR and western blotting analysis revealed the activation of SLX4 expression in response to MMS. The deletion of SLX4 resulted in a defect in the recovery from MMS-induced filamentation to yeast form and re-entry into the cell cycle. Like many other DNA repair genes, SLX4 expression was activated by the checkpoint kinase Rad53 under MMS-induced DNA damage. In addition, SLX4 was not required for the inactivation of the DNA damage checkpoint, as indicated by normal phosphorylation of Rad53 in slx4Δ/Δ cells. Therefore, our results demonstrate SLX4 plays an important role in cell recovery from MMS-induced DNA damage in C. albicans.


Subject(s)
Candida albicans/drug effects , Candida albicans/genetics , DNA Damage/drug effects , Endodeoxyribonucleases/genetics , Fungal Proteins/genetics , Methyl Methanesulfonate/pharmacology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Checkpoints/drug effects , Endodeoxyribonucleases/metabolism , Fungal Proteins/metabolism , Hydroxyurea/pharmacology , Phosphorylation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
Genes Dev ; 27(20): 2259-73, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24142876

ABSTRACT

The ATR-CHK1 axis stabilizes stalled replication forks and prevents their collapse into DNA double-strand breaks (DSBs). Here, we show that fork collapse in Atr-deleted cells is mediated through the combined effects the sumo targeted E3-ubiquitin ligase RNF4 and activation of the AURKA-PLK1 pathway. As indicated previously, Atr-deleted cells exhibited a decreased ability to restart DNA replication following fork stalling in comparison with control cells. However, suppression of RNF4, AURKA, or PLK1 returned the reinitiation of replication in Atr-deleted cells to near wild-type levels. In RNF4-depleted cells, this rescue directly correlated with the persistence of sumoylation of chromatin-bound factors. Notably, RNF4 repression substantially suppressed the accumulation of DSBs in ATR-deficient cells, and this decrease in breaks was enhanced by concomitant inhibition of PLK1. DSBs resulting from ATR inhibition were also observed to be dependent on the endonuclease scaffold protein SLX4, suggesting that RNF4 and PLK1 either help activate the SLX4 complex or make DNA replication fork structures accessible for subsequent SLX4-dependent cleavage. Thus, replication fork collapse following ATR inhibition is a multistep process that disrupts replisome function and permits cleavage of the replication fork.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , Chromatin/metabolism , DNA Breaks, Double-Stranded , Mice , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Recombinases/metabolism , Sumoylation , Transcription Factors/genetics , Ubiquitin-Protein Ligases , Polo-Like Kinase 1
15.
EMBO J ; 34(12): 1704-17, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25896509

ABSTRACT

In response to DNA damage, checkpoint signalling protects genome integrity at the cost of repressing cell cycle progression and DNA replication. Mechanisms for checkpoint down-regulation are therefore necessary for proper cellular proliferation. We recently uncovered a phosphatase-independent mechanism for dampening checkpoint signalling, where the checkpoint adaptor Rad9 is counteracted by the repair scaffolds Slx4-Rtt107. Here, we establish the molecular requirements for this new mode of checkpoint regulation. We engineered a minimal multi-BRCT-domain (MBD) module that recapitulates the action of Slx4-Rtt107 in checkpoint down-regulation. MBD mimics the damage-induced Dpb11-Slx4-Rtt107 complex by synergistically interacting with lesion-specific phospho-sites in Ddc1 and H2A. We propose that efficient recruitment of Dpb11-Slx4-Rtt107 or MBD via a cooperative 'two-site-docking' mechanism displaces Rad9. MBD also interacts with the Mus81 nuclease following checkpoint dampening, suggesting a spatio-temporal coordination of checkpoint signalling and DNA repair via a combinatorial mode of BRCT-domains interactions.


Subject(s)
Cell Cycle Checkpoints/physiology , DNA Damage/physiology , Models, Biological , Nuclear Proteins/metabolism , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Cycle Proteins/metabolism , Electrophoresis, Gel, Pulsed-Field , Immunoprecipitation , Nuclear Proteins/genetics , Protein Engineering/methods , Protein Structure, Tertiary , Recombinant Proteins/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics
16.
EMBO J ; 34(16): 2182-97, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26113155

ABSTRACT

Obstructions to replication fork progression, referred to collectively as DNA replication stress, challenge genome stability. In Saccharomyces cerevisiae, cells lacking RTT107 or SLX4 show genome instability and sensitivity to DNA replication stress and are defective in the completion of DNA replication during recovery from replication stress. We demonstrate that Slx4 is recruited to chromatin behind stressed replication forks, in a region that is spatially distinct from that occupied by the replication machinery. Slx4 complex formation is nucleated by Mec1 phosphorylation of histone H2A, which is recognized by the constitutive Slx4 binding partner Rtt107. Slx4 is essential for recruiting the Mec1 activator Dpb11 behind stressed replication forks, and Slx4 complexes are important for full activity of Mec1. We propose that Slx4 complexes promote robust checkpoint signaling by Mec1 by stably recruiting Dpb11 within a discrete domain behind the replication fork, during DNA replication stress.


Subject(s)
DNA Replication , DNA, Fungal/metabolism , Endodeoxyribonucleases/metabolism , Protein Multimerization , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Cell Cycle Proteins , Histones , Intracellular Signaling Peptides and Proteins , Nuclear Proteins , Protein Binding , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
17.
Trends Biochem Sci ; 39(9): 409-19, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25131815

ABSTRACT

Holliday junctions (HJs) are four-stranded DNA intermediates that arise during the recombinational repair of DNA double-strand breaks (DSBs). Their timely removal is crucial for faithful chromosome segregation and genome stability. In mammalian cells, HJs are processed by the BTR (BLM-topoisomerase IIIα-RMI1-RMI2) complex, the SLX-MUS (SLX1-SLX4-MUS81-EME1) complex, and the GEN1 resolvase. Recent studies have linked the deficiency of one or more of these enzymes to perturbed DNA replication, impaired crosslink repair, chromosomal instability, and defective mitoses, coupled with the transmission of widespread DNA damage and high levels of mortality. We review these key advances and how they have cemented the status of HJ-processing enzymes as guardians of genome integrity and viability in mammalian cells.


Subject(s)
DNA Replication , DNA, Cruciform/metabolism , Genomic Instability , Holliday Junction Resolvases/metabolism , Animals , DNA Damage , Humans , Recombination, Genetic
18.
Chromosoma ; 126(1): 45-58, 2017 02.
Article in English | MEDLINE | ID: mdl-27165041

ABSTRACT

Homologous recombination-mediated DNA repair is essential for maintaining genome integrity. It is a multi-step process that involves resection of DNA ends, strand invasion, DNA synthesis and/or DNA end ligation, and finally, the processing of recombination intermediates such as Holliday junctions or other joint molecules. Over the last 15 years, it has been established that the Slx4 protein plays key roles in the processing of recombination intermediates, functioning as a scaffold to coordinate the action of structure-specific endonucleases. Recent work in budding yeast has uncovered unexpected roles for Slx4 in the initial step of DNA-end resection and in the modulation of DNA damage checkpoint signaling. Here we review these latest findings and discuss the emerging role of yeast Slx4 as an important coordinator of DNA damage signaling responses and a regulator of multiple steps in homologous recombination-mediated repair.


Subject(s)
Cell Cycle Checkpoints , Endodeoxyribonucleases/metabolism , Homologous Recombination , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , DNA Repair , Endodeoxyribonucleases/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
19.
EMBO Rep ; 16(4): 512-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25722289

ABSTRACT

SUMOylation plays important roles in the DNA damage response. However, whether it is important for interstrand crosslink repair remains unknown. We report that the SLX4 nuclease scaffold protein is regulated by SUMOylation. We have identified three SUMO interaction motifs (SIMs) in SLX4, mutating all of which abrogated the binding of SLX4 to SUMO-2 and covalent SLX4 SUMOylation. An SLX4 mutant lacking functional SIMs is not recruited to PML nuclear bodies nor stabilized at laser-induced DNA damage sites. Additionally, we elucidated a novel role for PARylation in the recruitment of SLX4 to sites of DNA damage. Combined, our results uncover how SLX4 is regulated by post-translational modifications.


Subject(s)
DNA Repair , DNA/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Recombinases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Binding Sites , Cell Line, Tumor , DNA/genetics , DNA Damage , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Protein Stability , Protein Transport , Recombinases/genetics , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation
20.
J Cell Sci ; 127(Pt 13): 2811-7, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24794496

ABSTRACT

Defects in SLX4, a scaffold for DNA repair nucleases, result in Fanconi anemia (FA), due to the defective repair of inter-strand DNA crosslinks (ICLs). Some FA patients have an SLX4 deletion removing two tandem UBZ4-type ubiquitin-binding domains that are implicated in protein recruitment to sites of DNA damage. Here, we show that human SLX4 is recruited to sites of ICL induction but that the UBZ-deleted form of SLX4 in cells from FA patients is not. SLX4 recruitment does not require either the ubiquitylation of FANCD2 or the E3 ligases RNF8, RAD18 and BRCA1. We show that the first (UBZ-1) but not the second UBZ domain of SLX4 binds to ubiquitin polymers, with a preference for K63-linked chains. Furthermore, UBZ-1 is required for SLX4 recruitment to ICL sites and for efficient ICL repair in murine fibroblasts. The SLX4 UBZ-2 domain does not bind to ubiquitin in vitro or contribute to ICL repair, but it is required for the resolution of Holliday junctions in vivo. These data shed light on SLX4 recruitment, and they point to the existence of currently unidentified ubiquitylated ligands and E3 ligases that are crucial for ICL repair.


Subject(s)
Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Recombinases/genetics , Ubiquitin/metabolism , Animals , Binding Sites , DNA/genetics , DNA/metabolism , DNA Repair , Humans , Mice , Protein Structure, Tertiary , Recombinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL