Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.575
Filter
1.
Proc Natl Acad Sci U S A ; 121(34): e2402262121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145931

ABSTRACT

Homologous recombination (HR) is essential for the maintenance of genome stability. During HR, Replication Protein A (RPA) rapidly coats the 3'-tailed single-strand DNA (ssDNA) generated by end resection. Then, the ssDNA-bound RPA must be timely replaced by Rad51 recombinase to form Rad51 nucleoprotein filaments that drive homology search and HR repair. How cells regulate Rad51 assembly dynamics and coordinate RPA and Rad51 actions to ensure proper HR remains poorly understood. Here, we identified that Rtt105, a Ty1 transposon regulator, acts to stimulate Rad51 assembly and orchestrate RPA and Rad51 actions during HR. We found that Rtt105 interacts with Rad51 in vitro and in vivo and restrains the adenosine 5' triphosphate (ATP) hydrolysis activity of Rad51. We showed that Rtt105 directly stimulates dynamic Rad51-ssDNA assembly, strand exchange, and D-loop formation in vitro. Notably, we found that Rtt105 physically regulates the binding of Rad51 and RPA to ssDNA via different motifs and that both regulations are necessary and epistatic in promoting Rad51 nucleation, strand exchange, and HR repair. Consequently, disrupting either of the interactions impaired HR and conferred DNA damage sensitivity, underscoring the importance of Rtt105 in orchestrating the actions of Rad51 and RPA. Our work reveals additional layers of mechanisms regulating Rad51 filament dynamics and the coordination of HR.


Subject(s)
DNA, Single-Stranded , Rad51 Recombinase , Recombinational DNA Repair , Replication Protein A , Saccharomyces cerevisiae Proteins , Rad51 Recombinase/metabolism , Replication Protein A/metabolism , Replication Protein A/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Protein Binding
2.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125953

ABSTRACT

Targeting CDC20 can enhance the radiosensitivity of tumor cells, but the function and mechanism of CDC20 on DNA damage repair response remains vague. To examine that issue, tumor cell lines, including KYSE200, KYSE450, and HCT116, were utilized to detect the expression, function, and underlying mechanism of CDC20 in radio-chemoresistance. Western blot and immunofluorescence staining were employed to confirm CDC20 expression and location, and radiation could upregulate the expression of CDC20 in the cell nucleus. The homologous recombination (HR) and non-homologous end joining (NHEJ) reporter gene systems were utilized to explore the impact of CDC20 on DNA damage repair, indicating that CDC20 could promote HR repair and radio/chemo-resistance. In the early stages of DNA damage, CDC20 stabilizes the RPA1 protein through protein-protein interactions, activating the ATR-mediated signaling cascade, thereby aiding in genomic repair. In the later stages, CDC20 assists in the subsequent steps of damage repair by the ubiquitin-mediated degradation of RPA1. CCK-8 and colony formation assay were used to detect the function of CDC20 in cell vitality and proliferation, and targeting CDC20 can exacerbate the increase in DNA damage levels caused by cisplatin or etoposide. A tumor xenograft model was conducted in BALB/c-nu/nu mice to confirm the function of CDC20 in vivo, confirming the in vitro results. In conclusion, this study provides further validation of the potential clinical significance of CDC20 as a strategy to overcome radio-chemoresistance via uncovering a novel role of CDC20 in regulating RPA1 during DNA damage repair.


Subject(s)
Cdc20 Proteins , DNA Damage , Drug Resistance, Neoplasm , Radiation Tolerance , Replication Protein A , Humans , Animals , Replication Protein A/metabolism , Replication Protein A/genetics , Mice , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Drug Resistance, Neoplasm/genetics , Cdc20 Proteins/metabolism , Cdc20 Proteins/genetics , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude , DNA Repair/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Cisplatin/pharmacology , HCT116 Cells , Gene Expression Regulation, Neoplastic/drug effects
3.
Oncogene ; 43(32): 2475-2489, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38961202

ABSTRACT

The preferential response to PARP inhibitors (PARPis) in BRCA-deficient and Schlafen 11 (SLFN11)-expressing ovarian cancers has been documented, yet the underlying molecular mechanisms remain unclear. As the accumulation of single-strand DNA (ssDNA) gaps behind replication forks is key for the lethality effect of PARPis, we investigated the combined effects of SLFN11 expression and BRCA deficiency on PARPi sensitivity and ssDNA gap formation in human cancer cells. PARPis increased chromatin-bound RPA2 and ssDNA gaps in SLFN11-expressing cells and even more in cells with BRCA1 or BRCA2 deficiency. SLFN11 was co-localized with chromatin-bound RPA2 under PARPis treatment, with enhanced recruitment in BRCA2-deficient cells. Notably, the chromatin-bound SLFN11 under PARPis did not block replication, contrary to its function under replication stress. SLFN11 recruitment was attenuated by the inactivation of MRE11. Hence, under PARPi treatment, MRE11 expression and BRCA deficiency lead to ssDNA gaps behind replication forks, where SLFN11 binds and increases their accumulation. As ovarian cancer patients who responded (progression-free survival >2 years) to olaparib maintenance therapy had a significantly higher SLFN11-positivity than short-responders (<6 months), our findings provide a mechanistic understanding of the favorable responses to PARPis in SLFN11-expressing and BRCA-deficient tumors. It highlight the clinical implications of SLFN11.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA Replication , DNA, Single-Stranded , MRE11 Homologue Protein , Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , DNA Replication/drug effects , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Chromatin/metabolism , Phthalazines/pharmacology
4.
J Biol Chem ; 300(7): 107461, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876299

ABSTRACT

Theta-mediated end joining (TMEJ) is critical for survival of cancer cells when other DNA double-stranded break repair pathways are impaired. Human DNA polymerase theta (Pol θ) can extend ssDNA oligonucleotides, but little is known about preferred substrates and mechanism. We show that Pol θ can extend both ssDNA and RNA substrates by unimolecular stem-loop synthesis initiated by only two 3' terminal base pairs. Given sufficient time, Pol θ uses alternative pairing configurations that greatly expand the repertoire of sequence outcomes. Further primer-template adjustments yield low-fidelity outcomes when the nucleotide pool is imbalanced. Unimolecular stem-loop synthesis competes with bimolecular end joining, even when a longer terminal microhomology for end joining is available. Both reactions are partially suppressed by the ssDNA-binding protein replication protein A. Protein-primer grasp residues that are specific to Pol θ are needed for rapid stem-loop synthesis. The ability to perform stem-loop synthesis from a minimally paired primer is rare among human DNA polymerases, but we show that human DNA polymerases Pol η and Pol λ can catalyze related reactions. Using purified human Pol θ, we reconstituted in vitro TMEJ incorporating an insertion arising from a stem-loop extension. These activities may help explain TMEJ repair events that include inverted repeat sequences.


Subject(s)
DNA Polymerase theta , DNA-Directed DNA Polymerase , Humans , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/chemistry , DNA End-Joining Repair , DNA Repair , Replication Protein A/metabolism , Replication Protein A/genetics , DNA Polymerase beta/metabolism , DNA Polymerase beta/genetics , DNA Polymerase beta/chemistry
5.
Nucleic Acids Res ; 52(13): 7650-7664, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38842913

ABSTRACT

DNA polymerase δ (pol δ) holoenzymes, comprised of pol δ and the processivity sliding clamp, PCNA, carry out DNA synthesis during lagging strand replication, initiation of leading strand replication, and the major DNA damage repair and tolerance pathways. Pol δ holoenzymes are assembled at primer/template (P/T) junctions and initiate DNA synthesis in a stepwise process involving the major single strand DNA (ssDNA)-binding protein complex, RPA, the processivity sliding clamp loader, RFC, PCNA and pol δ. During this process, the interactions of RPA, RFC and pol δ with a P/T junction all significantly overlap. A burning issue that has yet to be resolved is how these overlapping interactions are accommodated during this process. To address this, we design and utilize novel, ensemble FRET assays that continuously monitor the interactions of RPA, RFC, PCNA and pol δ with DNA as pol δ holoenzymes are assembled and initiate DNA synthesis. Results from the present study reveal that RPA remains engaged with P/T junctions throughout this process and the RPA•DNA complexes dynamically re-organize to allow successive binding of RFC and pol δ. These results have broad implications as they highlight and distinguish the functional consequences of dynamic RPA•DNA interactions in RPA-dependent DNA metabolic processes.


Subject(s)
DNA Polymerase III , DNA Replication , DNA , Proliferating Cell Nuclear Antigen , Replication Protein A , Replication Protein C , Templates, Genetic , Replication Protein A/metabolism , DNA Polymerase III/metabolism , DNA Polymerase III/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Holoenzymes/metabolism , DNA/metabolism , DNA/biosynthesis , Replication Protein C/metabolism , Replication Protein C/genetics , DNA Primers/genetics , Fluorescence Resonance Energy Transfer , Humans
6.
Anal Chem ; 96(25): 10443-10450, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38864271

ABSTRACT

Due to their ability to selectively target pathogen-specific nucleic acids, CRISPR-Cas systems are increasingly being employed as diagnostic tools. "One-pot" assays that combine nucleic acid amplification and CRISPR-Cas systems (NAAT-CRISPR-Cas) in a single step have emerged as one of the most popular CRISPR-Cas biosensing formats. However, operational simplicity comes at a cost, with one-pot assays typically being less sensitive than corresponding two-step NAAT-CRISPR-Cas assays and often failing to detect targets at low concentrations. It is thought that these performance reductions result from the competition between the two enzymatic processes driving the assay, namely, Cas-mediated cis-cleavage and polymerase-mediated amplification of the target DNA. Herein, we describe a novel one-pot RPA-Cas12a assay that circumvents this issue by leveraging in situ complexation of the target-specific sgRNA and Cas12a to purposefully limit the concentration of active Cas12a during the early stages of the assay. Using a clinically relevant assay against a DNA target for HPV-16, we show how this in situ format reduces competition between target cleavage and amplification and engenders significant improvements in detection limit when compared to the traditional one-pot assay format, even in patient-derived samples. Finally, to gain further insight into the assay, we use experimental data to formulate a mechanistic model describing the competition between the Cas enzyme and nucleic acid amplification. These findings suggest that purposefully limiting cis-cleavage rates of Cas proteins is a viable strategy for improving the performance of one-pot NAAT-CRISPR-Cas assays.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , CRISPR-Cas Systems/genetics , CRISPR-Associated Proteins/metabolism , RNA, Guide, CRISPR-Cas Systems/metabolism , Humans , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Nucleic Acid Amplification Techniques , Replication Protein A/metabolism , Biosensing Techniques/methods
7.
Sci China Life Sci ; 67(6): 1089-1105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842635

ABSTRACT

Histone H3 Lys36 (H3K36) methylation and its associated modifiers are crucial for DNA double-strand break (DSB) repair, but the mechanism governing whether and how different H3K36 methylation forms impact repair pathways is unclear. Here, we unveil the distinct roles of H3K36 dimethylation (H3K36me2) and H3K36 trimethylation (H3K36me3) in DSB repair via non-homologous end joining (NHEJ) or homologous recombination (HR). Yeast cells lacking H3K36me2 or H3K36me3 exhibit reduced NHEJ or HR efficiency. yKu70 and Rfa1 bind H3K36me2- or H3K36me3-modified peptides and chromatin, respectively. Disrupting these interactions impairs yKu70 and Rfa1 recruitment to damaged H3K36me2- or H3K36me3-rich loci, increasing DNA damage sensitivity and decreasing repair efficiency. Conversely, H3K36me2-enriched intergenic regions and H3K36me3-enriched gene bodies independently recruit yKu70 or Rfa1 under DSB stress. Importantly, human KU70 and RPA1, the homologs of yKu70 and Rfa1, exclusively associate with H3K36me2 and H3K36me3 in a conserved manner. These findings provide valuable insights into how H3K36me2 and H3K36me3 regulate distinct DSB repair pathways, highlighting H3K36 methylation as a critical element in the choice of DSB repair pathway.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Histones , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Humans , Methylation , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Homologous Recombination , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Repair , Chromatin/metabolism , Chromatin/genetics
8.
PLoS Genet ; 20(5): e1011148, 2024 May.
Article in English | MEDLINE | ID: mdl-38776358

ABSTRACT

The helicase MCM and the ribonucleotide reductase RNR are the complexes that provide the substrates (ssDNA templates and dNTPs, respectively) for DNA replication. Here, we demonstrate that MCM interacts physically with RNR and some of its regulators, including the kinase Dun1. These physical interactions encompass small subpopulations of MCM and RNR, are independent of the major subcellular locations of these two complexes, augment in response to DNA damage and, in the case of the Rnr2 and Rnr4 subunits of RNR, depend on Dun1. Partial disruption of the MCM/RNR interactions impairs the release of Rad52 -but not RPA-from the DNA repair centers despite the lesions are repaired, a phenotype that is associated with hypermutagenesis but not with alterations in the levels of dNTPs. These results suggest that a specifically regulated pool of MCM and RNR complexes plays non-canonical roles in genetic stability preventing persistent Rad52 centers and hypermutagenesis.


Subject(s)
Cell Cycle Proteins , DNA Damage , DNA Repair , DNA Replication , Genomic Instability , Rad52 DNA Repair and Recombination Protein , Ribonucleotide Reductases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA Replication/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA Damage/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , DNA Repair/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism
9.
Sci Adv ; 10(21): eadk8908, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781342

ABSTRACT

DNA replication is a vulnerable cellular process, and its deregulation leads to genomic instability. Here, we demonstrate that chromobox protein homolog 3 (CBX3) binds replication protein A 32-kDa subunit (RPA2) and regulates RPA2 retention at stalled replication forks. CBX3 is recruited to stalled replication forks by RPA2 and inhibits ring finger and WD repeat domain 3 (RFWD3)-facilitated replication restart. Phosphorylation of CBX3 at serine-95 by casein kinase 2 (CK2) kinase augments cadherin 1 (CDH1)-mediated CBX3 degradation and RPA2 dynamics at stalled replication forks, which permits replication fork restart. Increased expression of CBX3 due to gene amplification or CK2 inhibitor treatment sensitizes prostate cancer cells to poly(ADP-ribose) polymerase (PARP) inhibitors while inducing replication stress and DNA damage. Our work reveals CBX3 as a key regulator of RPA2 function and DNA replication, suggesting that CBX3 could serve as an indicator for targeted therapy of cancer using PARP inhibitors.


Subject(s)
Casein Kinase II , DNA Replication , Poly(ADP-ribose) Polymerase Inhibitors , Replication Protein A , Humans , Casein Kinase II/metabolism , Casein Kinase II/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Replication Protein A/metabolism , Replication Protein A/genetics , Cell Line, Tumor , Proteolysis , DNA Damage , Phosphorylation , Chromosomal Proteins, Non-Histone
10.
Nucleic Acids Res ; 52(11): 6317-6332, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38613387

ABSTRACT

Telomerase is the enzyme that lengthens telomeres and is tightly regulated by a variety of means to maintain genome integrity. Several DNA helicases function at telomeres, and we previously found that the Saccharomyces cerevisiae helicases Hrq1 and Pif1 directly regulate telomerase. To extend these findings, we are investigating the interplay between helicases, single-stranded DNA (ssDNA) binding proteins (ssBPs), and telomerase. The yeast ssBPs Cdc13 and RPA differentially affect Hrq1 and Pif1 helicase activity, and experiments to measure helicase disruption of Cdc13/ssDNA complexes instead revealed that Cdc13 can exchange between substrates. Although other ssBPs display dynamic binding, this was unexpected with Cdc13 due to the reported in vitro stability of the Cdc13/telomeric ssDNA complex. We found that the DNA exchange by Cdc13 occurs rapidly at physiological temperatures, requires telomeric repeat sequence DNA, and is affected by ssDNA length. Cdc13 truncations revealed that the low-affinity binding site (OB1), which is distal from the high-affinity binding site (OB3), is required for this intermolecular dynamic DNA exchange (DDE). We hypothesize that DDE by Cdc13 is the basis for how Cdc13 'moves' at telomeres to alternate between modes where it regulates telomerase activity and assists in telomere replication.


Subject(s)
DNA Helicases , DNA, Single-Stranded , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Telomere-Binding Proteins , Telomere , Binding Sites , DNA Helicases/metabolism , DNA, Fungal/metabolism , DNA, Fungal/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Protein Binding , RecQ Helicases , Replication Protein A/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
11.
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
12.
Sci Rep ; 14(1): 9550, 2024 04 25.
Article in English | MEDLINE | ID: mdl-38664461

ABSTRACT

DNA double-strand breaks (DSBs) activate DNA damage responses (DDRs) in both mitotic and meiotic cells. A single-stranded DNA (ssDNA) binding protein, Replication protein-A (RPA) binds to the ssDNA formed at DSBs to activate ATR/Mec1 kinase for the response. Meiotic DSBs induce homologous recombination monitored by a meiotic DDR called the recombination checkpoint that blocks the pachytene exit in meiotic prophase I. In this study, we further characterized the essential role of RPA in the maintenance of the recombination checkpoint during Saccharomyces cerevisiae meiosis. The depletion of an RPA subunit, Rfa1, in a recombination-defective dmc1 mutant, fully alleviates the pachytene arrest with the persistent unrepaired DSBs. RPA depletion decreases the activity of a meiosis-specific CHK2 homolog, Mek1 kinase, which in turn activates the Ndt80 transcriptional regulator for pachytene exit. These support the idea that RPA is a sensor of ssDNAs for the activation of meiotic DDR. Rfa1 depletion also accelerates the prophase I delay in the zip1 mutant defective in both chromosome synapsis and the recombination, consistent with the notion that the accumulation of ssDNAs rather than defective synapsis triggers prophase I delay in the zip1 mutant.


Subject(s)
DNA Breaks, Double-Stranded , Meiosis , Replication Protein A , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Replication Protein A/metabolism , Replication Protein A/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Recombination, Genetic , Homologous Recombination , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 1/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
13.
BMC Biol ; 22(1): 101, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685010

ABSTRACT

BACKGROUND: CRISPR-Cas9 genome editing often induces unintended, large genomic rearrangements, posing potential safety risks. However, there are no methods for mitigating these risks. RESULTS: Using long-read individual-molecule sequencing (IDMseq), we found the microhomology-mediated end joining (MMEJ) DNA repair pathway plays a predominant role in Cas9-induced large deletions (LDs). We targeted MMEJ-associated genes genetically and/or pharmacologically and analyzed Cas9-induced LDs at multiple gene loci using flow cytometry and long-read sequencing. Reducing POLQ levels or activity significantly decreases LDs, while depleting or overexpressing RPA increases or reduces LD frequency, respectively. Interestingly, small-molecule inhibition of POLQ and delivery of recombinant RPA proteins also dramatically promote homology-directed repair (HDR) at multiple disease-relevant gene loci in human pluripotent stem cells and hematopoietic progenitor cells. CONCLUSIONS: Our findings reveal the contrasting roles of RPA and POLQ in Cas9-induced LD and HDR, suggesting new strategies for safer and more precise genome editing.


Subject(s)
CRISPR-Cas Systems , DNA End-Joining Repair , Gene Editing , Humans , Gene Editing/methods , DNA Breaks , Recombinational DNA Repair , Sequence Deletion , DNA Polymerase theta , Replication Protein A/metabolism , Replication Protein A/genetics
14.
PLoS Biol ; 22(3): e3002552, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38502677

ABSTRACT

Impediments in replication fork progression cause genomic instability, mutagenesis, and severe pathologies. At stalled forks, RPA-coated single-stranded DNA (ssDNA) activates the ATR kinase and directs fork remodeling, 2 key early events of the replication stress response. RFWD3, a recently described Fanconi anemia (FA) ubiquitin ligase, associates with RPA and promotes its ubiquitylation, facilitating late steps of homologous recombination (HR). Intriguingly, RFWD3 also regulates fork progression, restart and stability via poorly understood mechanisms. Here, we used proteomics to identify putative RFWD3 substrates during replication stress in human cells. We show that RFWD3 interacts with and ubiquitylates the SMARCAL1 DNA translocase directly in vitro and following DNA damage in vivo. SMARCAL1 ubiquitylation does not trigger its subsequent proteasomal degradation but instead disengages it from RPA thereby regulating its function at replication forks. Proper regulation of SMARCAL1 by RFWD3 at stalled forks protects them from excessive MUS81-mediated cleavage in response to UV irradiation, thereby limiting DNA replication stress. Collectively, our results identify RFWD3-mediated SMARCAL1 ubiquitylation as a novel mechanism that modulates fork remodeling to avoid genome instability triggered by aberrant fork processing.


Subject(s)
DNA Replication , DNA, Single-Stranded , Humans , DNA, Single-Stranded/genetics , DNA Replication/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , Protein Binding , Ubiquitination , DNA Damage , Genomic Instability , DNA Helicases/genetics , DNA Helicases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
J Radiat Res ; 65(3): 263-271, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38461549

ABSTRACT

Ionizing radiation (IR)-induced double-strand breaks (DSBs) are primarily repaired by non-homologous end joining or homologous recombination (HR) in human cells. DSB repair requires adenosine-5'-triphosphate (ATP) for protein kinase activities in the multiple steps of DSB repair, such as DNA ligation, chromatin remodeling, and DNA damage signaling via protein kinase and ATPase activities. To investigate whether low ATP culture conditions affect the recruitment of repair proteins at DSB sites, IR-induced foci were examined in the presence of ATP synthesis inhibitors. We found that p53 binding protein 1 foci formation was modestly reduced under low ATP conditions after IR, although phosphorylated histone H2AX and mediator of DNA damage checkpoint 1 foci formation were not impaired. Next, we examined the foci formation of breast cancer susceptibility gene I (BRCA1), replication protein A (RPA) and radiation 51 (RAD51), which are HR factors, in G2 phase cells following IR. Interestingly, BRCA1 and RPA foci in the G2 phase were significantly reduced under low ATP conditions compared to that under normal culture conditions. Notably, RAD51 foci were drastically impaired under low ATP conditions. These results suggest that HR does not effectively progress under low ATP conditions; in particular, ATP shortages impair downstream steps in HR, such as RAD51 loading. Taken together, these results suggest that the maintenance of cellular ATP levels is critical for DNA damage response and HR progression after IR.


Subject(s)
Adenosine Triphosphate , BRCA1 Protein , Homologous Recombination , Rad51 Recombinase , Radiation, Ionizing , Humans , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Homologous Recombination/radiation effects , Rad51 Recombinase/metabolism , BRCA1 Protein/metabolism , DNA Breaks, Double-Stranded/radiation effects , Replication Protein A/metabolism , Cell Line, Tumor , Intracellular Space/metabolism , Intracellular Space/radiation effects , DNA Repair , Histones/metabolism
16.
Nat Commun ; 15(1): 2210, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472229

ABSTRACT

The ATR-CHK1 DNA damage response pathway becomes activated by the exposure of RPA-coated single-stranded DNA (ssDNA) that forms as an intermediate during DNA damage and repair, and as a part of the replication stress response. Here, we identify ZNF827 as a component of the ATR-CHK1 kinase pathway. We demonstrate that ZNF827 is a ssDNA binding protein that associates with RPA through concurrent binding to ssDNA intermediates. These interactions are dependent on two clusters of C2H2 zinc finger motifs within ZNF827. We find that ZNF827 accumulates at stalled forks and DNA damage sites, where it activates ATR and promotes the engagement of homologous recombination-mediated DNA repair. Additionally, we demonstrate that ZNF827 depletion inhibits replication initiation and sensitizes cancer cells to the topoisomerase inhibitor topotecan, revealing ZNF827 as a therapeutic target within the DNA damage response pathway.


Subject(s)
Protein Kinases , Signal Transduction , Protein Kinases/metabolism , Phosphorylation , Replication Protein A/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , DNA Replication , DNA Damage , DNA, Single-Stranded , DNA Repair
17.
Genes (Basel) ; 15(2)2024 01 27.
Article in English | MEDLINE | ID: mdl-38397158

ABSTRACT

Human Replication Protein A (RPA) was historically discovered as one of the six components needed to reconstitute simian virus 40 DNA replication from purified components. RPA is now known to be involved in all DNA metabolism pathways that involve single-stranded DNA (ssDNA). Heterotrimeric RPA comprises several domains connected by flexible linkers and is heavily regulated by post-translational modifications (PTMs). The structure of RPA has been challenging to obtain. Various structural methods have been applied, but a complete understanding of RPA's flexible structure, its function, and how it is regulated by PTMs has yet to be obtained. This review will summarize recent literature concerning how RPA is phosphorylated in the cell cycle, the structural analysis of RPA, DNA and protein interactions involving RPA, and how PTMs regulate RPA activity and complex formation in double-strand break repair. There are many holes in our understanding of this research area. We will conclude with perspectives for future research on how RPA PTMs control double-strand break repair in the cell cycle.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA , Replication Protein A , Humans , DNA/metabolism , DNA Repair/genetics , DNA, Single-Stranded , Phosphorylation , Replication Protein A/metabolism
18.
J Mol Biol ; 436(6): 168491, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38360091

ABSTRACT

Replication Protein A (RPA) is asingle strandedDNA(ssDNA)binding protein that coordinates diverse DNA metabolic processes including DNA replication, repair, and recombination. RPA is a heterotrimeric protein with six functional oligosaccharide/oligonucleotide (OB) domains and flexible linkers. Flexibility enables RPA to adopt multiple configurations andis thought to modulate its function. Here, usingsingle moleculeconfocal fluorescencemicroscopy combinedwith optical tweezers and coarse-grained molecular dynamics simulations, we investigated the diffusional migration of single RPA molecules on ssDNA undertension.The diffusioncoefficientDis the highest (20,000nucleotides2/s) at 3pNtension and in 100 mMKCl and markedly decreases whentensionor salt concentrationincreases. We attribute the tension effect to intersegmental transfer which is hindered by DNA stretching and the salt effect to an increase in binding site size and interaction energy of RPA-ssDNA. Our integrative study allowed us to estimate the size and frequency of intersegmental transfer events that occur through transient bridging of distant sites on DNA by multiple binding sites on RPA. Interestingly, deletion of RPA trimeric core still allowed significant ssDNA binding although the reduced contact area made RPA 15-fold more mobile. Finally, we characterized the effect of RPA crowding on RPA migration. These findings reveal how the high affinity RPA-ssDNA interactions are remodeled to yield access, a key step in several DNA metabolic processes.


Subject(s)
DNA, Single-Stranded , Replication Protein A , DNA Replication , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Protein Binding/genetics , Replication Protein A/chemistry , Replication Protein A/genetics , Replication Protein A/metabolism
19.
Nucleic Acids Res ; 52(7): 3794-3809, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38340339

ABSTRACT

Meiotic recombination is initiated by programmed double-strand breaks (DSBs). Studies in Saccharomyces cerevisiae have shown that, following rapid resection to generate 3' single-stranded DNA (ssDNA) tails, one DSB end engages a homolog partner chromatid and is extended by DNA synthesis, whereas the other end remains associated with its sister. Then, after regulated differentiation into crossover- and noncrossover-fated types, the second DSB end participates in the reaction by strand annealing with the extended first end, along both pathways. This second-end capture is dependent on Rad52, presumably via its known capacity to anneal two ssDNAs. Here, using physical analysis of DNA recombination, we demonstrate that this process is dependent on direct interaction of Rad52 with the ssDNA binding protein, replication protein A (RPA). Furthermore, the absence of this Rad52-RPA joint activity results in a cytologically-prominent RPA spike, which emerges from the homolog axes at sites of crossovers during the pachytene stage of the meiotic prophase. Our findings suggest that this spike represents the DSB end of a broken chromatid caused by either the displaced leading DSB end or the second DSB end, which has been unable to engage with the partner homolog-associated ssDNA. These and other results imply a close correspondence between Rad52-RPA roles in meiotic recombination and mitotic DSB repair.


Subject(s)
Crossing Over, Genetic , DNA Breaks, Double-Stranded , Meiosis , Rad52 DNA Repair and Recombination Protein , Replication Protein A , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Meiosis/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Recombination, Genetic , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Homologous Recombination/genetics
20.
Nucleic Acids Res ; 52(9): 5067-5087, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38416570

ABSTRACT

CSB (Cockayne syndrome group B) and SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent, regulator of chromatin, subfamily A-like 1) are DNA translocases that belong to the SNF2 helicase family. They both are enriched at stalled replication forks. While SMARCAL1 is recruited by RPA32 to stalled forks, little is known about whether RPA32 also regulates CSB's association with stalled forks. Here, we report that CSB directly interacts with RPA, at least in part via a RPA32C-interacting motif within the N-terminal region of CSB. Modeling of the CSB-RPA32C interaction suggests that CSB binds the RPA32C surface previously shown to be important for binding of UNG2 and SMARCAL1. We show that this interaction is necessary for promoting fork slowing and fork degradation in BRCA2-deficient cells but dispensable for mediating restart of stalled forks. CSB competes with SMARCAL1 for RPA32 at stalled forks and acts non-redundantly with SMARCAL1 to restrain fork progression in response to mild replication stress. In contrast to CSB stimulated restart of stalled forks, SMARCAL1 inhibits restart of stalled forks in BRCA2-deficient cells, likely by suppressing BIR-mediated repair of collapsed forks. Loss of CSB leads to re-sensitization of SMARCAL1-depleted BRCA2-deficient cells to chemodrugs, underscoring a role of CSB in targeted cancer therapy.


Subject(s)
BRCA2 Protein , DNA Helicases , DNA Repair Enzymes , DNA Replication , Poly-ADP-Ribose Binding Proteins , Replication Protein A , DNA Helicases/metabolism , DNA Helicases/genetics , Humans , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Protein Binding , Cell Line, Tumor , DNA Repair
SELECTION OF CITATIONS
SEARCH DETAIL