Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 834
Filter
1.
Genes Dev ; 38(15-16): 755-771, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39231615

ABSTRACT

Premature telomere shortening or telomere instability is associated with a group of rare and heterogeneous diseases collectively known as telomere biology disorders (TBDs). Here we identified two unrelated individuals with clinical manifestations of TBDs and short telomeres associated with the identical monoallelic variant c.767A>G; Y256C in RPA2 Although the replication protein A2 (RPA2) mutant did not affect ssDNA binding and G-quadruplex-unfolding properties of RPA, the mutation reduced the affinity of RPA2 with the ubiquitin ligase RFWD3 and reduced RPA ubiquitination. Using engineered knock-in cell lines, we found an accumulation of RPA at telomeres that did not trigger ATR activation but caused short and dysfunctional telomeres. Finally, both patients acquired, in a subset of blood cells, somatic genetic rescue events in either POT1 genes or TERT promoters known to counteract the accelerated telomere shortening. Collectively, our study indicates that variants in RPA2 represent a novel genetic cause of TBDs. Our results further support the fundamental role of the RPA complex in regulating telomere length and stability in humans.


Subject(s)
Replication Protein A , Telomere-Binding Proteins , Telomere , Humans , Replication Protein A/genetics , Replication Protein A/metabolism , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Heterozygote , Male , Female , Shelterin Complex , Telomere Shortening/genetics , Mutation , Telomerase/genetics , Telomerase/metabolism , Ubiquitination/genetics , Ubiquitin-Protein Ligases/genetics
2.
Nat Commun ; 15(1): 7797, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242676

ABSTRACT

Ribosomal DNA (rDNA) encodes the ribosomal RNA genes and represents an intrinsically unstable genomic region. However, the underlying mechanisms and implications for genome integrity remain elusive. Here, we use Bloom syndrome (BS), a rare genetic disease characterized by DNA repair defects and hyper-unstable rDNA, as a model to investigate the mechanisms leading to rDNA instability. We find that in Bloom helicase (BLM) proficient cells, the homologous recombination (HR) pathway in rDNA resembles that in nuclear chromatin; it is initiated by resection, replication protein A (RPA) loading and BRCA2-dependent RAD51 filament formation. However, BLM deficiency compromises RPA-loading and BRCA1/2 recruitment to rDNA, but not RAD51 accumulation. RAD51 accumulates at rDNA despite depletion of long-range resection nucleases and rDNA damage results in micronuclei when BLM is absent. In summary, our findings indicate that rDNA is permissive to RAD51 accumulation in the absence of BLM, leading to micronucleation and potentially global genomic instability.


Subject(s)
DNA, Ribosomal , Genomic Instability , Rad51 Recombinase , RecQ Helicases , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Humans , RecQ Helicases/metabolism , RecQ Helicases/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , Homologous Recombination , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , DNA Repair
3.
J Struct Biol ; 216(3): 108115, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39117045

ABSTRACT

Human RAD52 protein binds DNA and is involved in genomic stability maintenance and several forms of DNA repair, including homologous recombination and single-strand annealing. Despite its importance, there are very few structural details about the variability of the RAD52 ring size and the RAD52 C-terminal protein-protein interaction domains. Even recent attempts to employ cryogenic electron microscopy (cryoEM) methods on full-length yeast and human RAD52 do not reveal interpretable structures for the C-terminal half that contains the replication protein A (RPA) and RAD51 binding domains. In this study, we employed the monodisperse purification of two RAD52 deletion constructs and small angle X-ray scattering (SAXS) to construct a structural model that includes RAD52's RPA binding domain. This model is of interest to DNA repair specialists as well as for drug development against HR-deficient cancers.


Subject(s)
Protein Binding , Rad52 DNA Repair and Recombination Protein , Replication Protein A , Scattering, Small Angle , Humans , Rad52 DNA Repair and Recombination Protein/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/chemistry , Replication Protein A/metabolism , Replication Protein A/chemistry , Replication Protein A/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , X-Ray Diffraction/methods , DNA Repair , Models, Molecular , Protein Domains
4.
DNA Repair (Amst) ; 142: 103741, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153403

ABSTRACT

PrimPol is a human DNA primase involved in DNA damage tolerance pathways by restarting DNA replication downstream of DNA lesions and non-canonical DNA structures. Activity and affinity to DNA relays on the interaction of PrimPol with replication protein A (RPA). In this work, we report that PrimPol has an intrinsic ability to copy DNA hairpins with a stem length of 5-9 base pairs (bp) but shows pronounced pausing of DNA synthesis. RPA greatly stimulates DNA synthesis across inverted DNA repeats by PrimPol. Moreover, deletion of the C-terminal RPA binding motif (RBM) facilitates DNA hairpin bypass and makes it independent of RPA. This work supports the idea that RBM is a negative regulator of PrimPol and its interaction with RPA is required to achieve the fully active state.


Subject(s)
DNA Primase , DNA Replication , DNA , Humans , DNA Primase/metabolism , DNA Primase/chemistry , DNA Primase/genetics , DNA/metabolism , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/chemistry , Replication Protein A/metabolism , Nucleic Acid Conformation , DNA-Directed DNA Polymerase/metabolism , Inverted Repeat Sequences , Protein Binding
5.
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
6.
Nat Commun ; 15(1): 7464, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39198528

ABSTRACT

RNase H1 has been acknowledged as an endoribonuclease specializing in the internal degradation of the RNA moiety within RNA-DNA hybrids, and its ribonuclease activity is indispensable in multifaceted aspects of nucleic acid metabolism. However, the molecular mechanism underlying RNase H1-mediated hybrid cleavage remains inadequately elucidated. Herein, using single-molecule approaches, we probe the dynamics of the hybrid cleavage by Saccharomyces cerevisiae RNase H1. Remarkably, a single RNase H1 enzyme displays 3'-to-5' exoribonuclease activity. The directional RNA degradation proceeds processively and yet discretely, wherein unwinding approximately 6-bp hybrids as a prerequisite for two consecutive 3-nt RNA excisions limits the overall rate within each catalytic cycle. Moreover, Replication Protein A (RPA) reinforces RNase H1's 3'-to-5' nucleolytic rate and processivity and stimulates its 5'-to-3' exoribonuclease activity. This stimulation is primarily realized through the pre-separation of the hybrids and consequently transfers RNase H1 to a bidirectional exoribonuclease, further potentiating its cleavage efficiency. These findings unveil unprecedented characteristics of an RNase and provide a dynamic view of RPA-enhanced processive hybrid cleavage by RNase H1.


Subject(s)
Exoribonucleases , RNA , Replication Protein A , Ribonuclease H , Saccharomyces cerevisiae , Ribonuclease H/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Exoribonucleases/metabolism , Exoribonucleases/genetics , RNA/metabolism , RNA/genetics , Replication Protein A/metabolism , DNA/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , RNA Stability , Nucleic Acid Hybridization
7.
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
8.
Nat Commun ; 15(1): 7197, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169038

ABSTRACT

Templated DNA repair that occurs during homologous recombination and replication stress relies on RAD51. RAD51 activity is positively regulated by BRCA2 and the RAD51 paralogs. The Shu complex is a RAD51 paralog-containing complex consisting of SWSAP1, SWS1, and SPIDR. We demonstrate that SWSAP1-SWS1 binds RAD51, maintains RAD51 filament stability, and enables strand exchange. Using single-molecule confocal fluorescence microscopy combined with optical tweezers, we show that SWSAP1-SWS1 decorates RAD51 filaments proficient for homologous recombination. We also find SWSAP1-SWS1 enhances RPA diffusion on ssDNA. Importantly, we show human sgSWSAP1 and sgSWS1 knockout cells are sensitive to pharmacological inhibition of PARP and APE1. Lastly, we identify cancer variants in SWSAP1 that alter Shu complex formation. Together, we show that SWSAP1-SWS1 stimulates RAD51-dependent high-fidelity repair and may be an important new cancer therapeutic target.


Subject(s)
DNA, Single-Stranded , Rad51 Recombinase , Replication Protein A , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Humans , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Replication Protein A/metabolism , Replication Protein A/genetics , DNA Repair , Protein Binding , Homologous Recombination , Single Molecule Imaging , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics
9.
Sci Rep ; 14(1): 19791, 2024 08 26.
Article in English | MEDLINE | ID: mdl-39187637

ABSTRACT

The ATR kinase responds to elevated levels of single-stranded DNA (ssDNA) to activate the G2/M checkpoint, regulate origin utilization, preserve fork stability, and allow DNA repair to ensure genome integrity. The intrinsic replication stress in cancer cells makes this pathway an attractive therapeutic target. The ssDNA that drives ATR signaling is sensed by the ssDNA-binding protein replication protein A (RPA), which acts as a platform for ATRIP recruitment and subsequent ATR activation by TopBP1. We have developed chemical RPA inhibitors (RPAi) that block RPA-ssDNA interactions (RPA-DBi) and RPA protein-protein interactions (RPA-PPIi); both activities are required for ATR activation. Here, we biochemically reconstitute the ATR kinase signaling pathway and demonstrate that RPA-DBi and RPA-PPIi abrogate ATR-dependent phosphorylation of target proteins with selectivity advantages over active site ATR inhibitors. We demonstrate that RPA post-translational modifications (PTMs) impact ATR kinase activation but do not alter sensitivity to RPAi. Specifically, phosphorylation of RPA32 and TopBP1 stimulate, while RPA70 acetylation does not affect ATR phosphorylation of target proteins. Collectively, this work reveals the RPAi mechanism of action to inhibit ATR signaling that can be regulated by RPA PTMs and offers insight into the anti-cancer activity of ATR pathway-targeted cancer therapeutics.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA, Single-Stranded , Protein Processing, Post-Translational , Replication Protein A , Signal Transduction , Replication Protein A/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Humans , Phosphorylation , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Protein Binding , Carrier Proteins , Nuclear Proteins
10.
Nucleic Acids Res ; 52(15): 8880-8896, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38967018

ABSTRACT

The simian virus 40 (SV40) replisome only encodes for its helicase; large T-antigen (L-Tag), while relying on the host for the remaining proteins, making it an intriguing model system. Despite being one of the earliest reconstituted eukaryotic systems, the interactions coordinating its activities and the identification of new factors remain largely unexplored. Herein, we in vitro reconstituted the SV40 replisome activities at the single-molecule level, including DNA unwinding by L-Tag and the single-stranded DNA-binding protein Replication Protein A (RPA), primer extension by DNA polymerase δ, and their concerted leading-strand synthesis. We show that RPA stimulates the processivity of L-Tag without altering its rate and that DNA polymerase δ forms a stable complex with L-Tag during leading-strand synthesis. Furthermore, similar to human and budding yeast Cdc45-MCM-GINS helicase, L-Tag uses the fork protection complex (FPC) and the mini-chromosome maintenance protein 10 (Mcm10) during synthesis. Hereby, we demonstrate that FPC increases this rate, and both FPC and Mcm10 increase the processivity by stabilizing stalled replisomes and increasing their chances of restarting synthesis. The detailed kinetics and novel factors of the SV40 replisome establish it as a closer mimic of the host replisome and expand its application as a model replication system.


Subject(s)
DNA Replication , Minichromosome Maintenance Proteins , Replication Protein A , Simian virus 40 , Simian virus 40/metabolism , Simian virus 40/genetics , Humans , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Replication Protein A/metabolism , DNA Polymerase III/metabolism , DNA Polymerase III/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , DNA, Viral/metabolism , DNA, Viral/genetics , Virus Replication , Single Molecule Imaging , Antigens, Polyomavirus Transforming/metabolism , Antigens, Polyomavirus Transforming/genetics , DNA, Single-Stranded/metabolism , DNA-Directed DNA Polymerase , Multienzyme Complexes
11.
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
12.
J Assist Reprod Genet ; 41(9): 2419-2439, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39023827

ABSTRACT

PURPOSE: Ovarian aging is closely related to a decrease in follicular reserve and oocyte quality. The precise molecular mechanisms underlying these reductions have yet to be fully elucidated. Herein, we examine spatiotemporal distribution of key proteins responsible for DNA double-strand break (DSB) repair in ovaries from early to older ages. Functional studies have shown that the γH2AX, RAD51, BRCA1, and RPA70 proteins play indispensable roles in HR-based repair pathway, while the KU80 and XRCC4 proteins are essential for successfully operating cNHEJ pathway. METHODS: Female Balb/C mice were divided into five groups as follows: Prepuberty (3 weeks old; n = 6), puberty (7 weeks old; n = 7), postpuberty (18 weeks old; n = 7), early aged (52 weeks old; n = 7), and late aged (60 weeks old; n = 7). The expression of DSB repair proteins, cellular senescence (ß-GAL) and apoptosis (cCASP3) markers was evaluated in the ovaries using immunohistochemistry. RESULT: ß-GAL and cCASP3 levels progressively increased from prepuberty to aged groups (P < 0.05). Notably, γH2AX levels varied in preantral and antral follicles among the groups (P < 0.05). In aged groups, RAD51, BRCA1, KU80, and XRCC4 levels increased (P < 0.05), while RPA70 levels decreased (P < 0.05) compared to the other groups. CONCLUSIONS: The observed alterations were primarily attributed to altered expression in oocytes and granulosa cells of the follicles and other ovarian cells. As a result, the findings indicate that these DSB repair proteins may play a role in the repair processes and even other related cellular events in ovarian cells from early to older ages.


Subject(s)
BRCA1 Protein , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins , Histones , Ku Autoantigen , Ovarian Follicle , Ovary , Rad51 Recombinase , Animals , Female , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Mice , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Repair/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/growth & development , Histones/genetics , Histones/metabolism , Ovary/metabolism , Ovary/growth & development , Oocytes/metabolism , Oocytes/growth & development , Aging/genetics , Aging/metabolism , Replication Protein A/metabolism , Replication Protein A/genetics , Mice, Inbred BALB C
13.
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 End-Joining Repair , DNA Polymerase beta/metabolism , DNA Polymerase beta/genetics , DNA Polymerase beta/chemistry , DNA Repair , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/chemistry , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Replication Protein A/metabolism , Replication Protein A/genetics
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL