Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.744
Filter
1.
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
2.
Cell Physiol Biochem ; 58(5): 459-476, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39248186

ABSTRACT

BACKGROUND/AIMS: One of the treatments for breast cancer is surgical resection of the tumour and prevention of recurrence with postoperative radiotherapy. Unfortunately, radiotherapy is not always effective enough due to the low sensitivity of cancer cells to ionising radiation. This study aimed to evaluate the radiosensitising properties of resveratrol, piceatannol and polydatin on breast cancer cells, which differ in the presence of hormonal receptors on their surface. METHODS: The experimental part was carried out on triple-negative breast cancer cells (HCC38) and hormone-dependent cells (MCF7). The study assessed the level of cell death, changes in the expression of genes (BAX, BCL-2) and proteins related to the apoptosis process (CASPASE 3, 8 and P53), changes in the expression of antioxidant enzymes (CATALASE, SOD, GPx1/2) and NRF-2. Additionally, the expression level of RAD51 protein and histone H2AX, which are involved in DNA repair processes, was assessed. Statistical significance was evaluated by a two-way analysis of variance (ANOVA) followed by Tukey's post hoc test (p < 0.05). RESULTS: Ionising radiation in combination with resveratrol or piceatannol activates the apoptosis process by internal and external pathways. Greater sensitivity of MCF7 cells compared to HCC38 cells to ionising radiation in combination with resveratrol is associated with a weaker antioxidant response of cells and reduced intensity of DNA damage repair. DNA repair induced by ionising radiation occurs more effectively in HCC38 cells than in MCF7 cells. CONCLUSION: Resveratrol has the highest radiosensitising potential among the tested stilbene for cells of both lines. The effectiveness of ionizing radiation in combination with resveratrol (to a lesser extent with piceatannol) is more significant in MCF7 than in HCC38 cells.


Subject(s)
Apoptosis , Radiation, Ionizing , Radiation-Sensitizing Agents , Resveratrol , Stilbenes , Humans , Stilbenes/pharmacology , Resveratrol/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Female , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , MCF-7 Cells , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Breast Neoplasms/drug therapy , Histones/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Rad51 Recombinase/metabolism , Caspase 3/metabolism , Glucosides
3.
Nucleic Acids Res ; 52(17): 10490-10503, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39189448

ABSTRACT

Telomeric repeat containing RNA (TERRA) is a noncoding RNA that is transcribed from telomeres. Previous study showed that TERRA trans anneals by invading into the telomeric duplex to form an R-loop in mammalian cells. Here, we elucidate the molecular mechanism underlying TERRA recruitment and invasion into telomeres in the context of shelterin proteins, RAD51 and RNase H using single molecule (sm) assays. We demonstrate that TERRA trans annealing into telomeric DNA exhibits dynamic movement that is stabilized by TRF2. TERRA annealing to the telomeric duplex results in the formation of a stable triplex structure which differs from a conventional R-loop. We identified that the presence of a sub-telomeric DNA and a telomeric overhang in the form of a G-quadruplex significantly enhances TERRA annealing to telomeric duplex. We also demonstrate that RAD51-TERRA complex invades telomere duplex more efficiently than TERRA alone. Additionally, TRF2 increases TERRA affinity to telomeric duplex and protects it from RNase H digestion. In contrast, TRF1 represses TERRA annealing to telomeric duplex and fails to provide protection against RNase H digestion. Our findings provide an in-depth molecular mechanism underpinning TERRA recruitment and annealing to the telomere.


Subject(s)
Rad51 Recombinase , Ribonuclease H , Telomere , Telomeric Repeat Binding Protein 1 , Telomeric Repeat Binding Protein 2 , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Telomeric Repeat Binding Protein 2/genetics , Ribonuclease H/metabolism , Rad51 Recombinase/metabolism , Humans , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 1/genetics , G-Quadruplexes , DNA/metabolism , DNA/chemistry , DNA/genetics , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , R-Loop Structures , RNA, Untranslated/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/chemistry , Shelterin Complex/metabolism , Single Molecule Imaging
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167481, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39186963

ABSTRACT

Radiotherapy stands as an effective method in the clinical treatment of hepatocellular carcinoma (HCC) patients. However, both primary and acquired radioresistance limit its clinical application in HCC. Therefore, investigating the mechanism of radioresistance may provide other options for treating HCC. Based on single-cell RNA sequencing (scRNA-seq) and HCC transcriptome datasets, 227 feature genes with prognostic value were selected to establish the tSNE score. The tSNE score emerged as an independent prognostic factor for HCC and correlated with cell proliferation and radioresistance-related biological functions. UBAP2 was identified as the most relevant gene with the tSNE score, consistently elevated in human HCC samples, and positively associated with patient prognosis. Functionally, UBAP2 knockdown impeded HCC development and reduced radiation resistance in vitro and in vivo. The ectopic expression of SLC27A5 reversed the effects of UBAP2. Mechanically, we uncovered that UBAP2, through the ubiquitin-proteasome system, decreased the homologous recombination-related gene RAD51, not the non-homologous end-joining (NHEJ)-related gene CTIP, by degrading the antioncogene SLC27A5, thereby generating radioresistance in HCC. The findings recapitulated that UBAP2 promoted HCC progression and radioresistance via SLC27A5 stability mediated by the ubiquitin-proteasome pathway. It was also suggested that targeting the UBAP2/SLC27A5 axis could be a valuable radiosensitization strategy in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Radiation Tolerance , Ubiquitination , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/radiotherapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Liver Neoplasms/metabolism , Radiation Tolerance/genetics , Mice , Animals , Homologous Recombination , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Prognosis , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Cell Proliferation/genetics , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Mice, Nude , Male , Carrier Proteins
5.
Nat Commun ; 15(1): 7081, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39152168

ABSTRACT

DSS1, essential for BRCA2-RAD51 dependent homologous recombination (HR), associates with the helical domain (HD) and OB fold 1 (OB1) of the BRCA2 DSS1/DNA-binding domain (DBD) which is frequently targeted by cancer-associated pathogenic variants. Herein, we reveal robust ss/dsDNA binding abilities in HD-OB1 subdomains and find that DSS1 shuts down HD-OB1's DNA binding to enable ssDNA targeting of the BRCA2-RAD51 complex. We show that C-terminal helix mutations of DSS1, including the cancer-associated R57Q mutation, disrupt this DSS1 regulation and permit dsDNA binding of HD-OB1/BRCA2-DBD. Importantly, these DSS1 mutations impair BRCA2/RAD51 ssDNA loading and focus formation and cause decreased HR efficiency, destabilization of stalled forks and R-loop accumulation, and hypersensitize cells to DNA-damaging agents. We propose that DSS1 restrains the intrinsic dsDNA binding of BRCA2-DBD to ensure BRCA2/RAD51 targeting to ssDNA, thereby promoting optimal execution of HR, and potentially replication fork protection and R-loop suppression.


Subject(s)
BRCA2 Protein , DNA Replication , DNA, Single-Stranded , DNA , Homologous Recombination , Mutation , Rad51 Recombinase , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/chemistry , Humans , DNA/metabolism , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Homeostasis , Protein Binding , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Protein Domains , Cell Line, Tumor , DNA Damage , Proteasome Endopeptidase Complex
6.
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
7.
Mol Cell ; 84(17): 3237-3253.e6, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39178861

ABSTRACT

Homology search is a central step of DNA double-strand break (DSB) repair by homologous recombination (HR). How it operates in cells remains elusive. We developed a Hi-C-based methodology to map single-stranded DNA (ssDNA) contacts genome-wide in S. cerevisiae, which revealed two main homology search phases. Initial search conducted by short Rad51-ssDNA nucleoprotein filaments (NPFs) is confined in cis by cohesin-mediated chromatin loop folding. Progressive growth of stiff NPFs enables exploration of distant genomic sites. Long-range resection drives this transition from local to genome-wide search by increasing the probability of assembling extensive NPFs. DSB end-tethering promotes coordinated search by opposite NPFs. Finally, an autonomous genetic element on chromosome III engages the NPF, which stimulates homology search in its vicinity. This work reveals the mechanism of the progressive expansion of homology search that is orchestrated by chromatin organizers, long-range resection, end-tethering, and specialized genetic elements and that exploits the stiff NPF structure conferred by Rad51 oligomerization.


Subject(s)
DNA Breaks, Double-Stranded , DNA, Fungal , DNA, Single-Stranded , Rad51 Recombinase , Recombinational DNA Repair , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Chromatin/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cohesins
8.
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
9.
Cell Rep ; 43(8): 114594, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39116203

ABSTRACT

Homologous recombination (HR) plays an essential role in the repair of DNA double-strand breaks (DSBs), replication stress responses, and genome maintenance. However, unregulated HR during replication can impair genome duplication and compromise genome stability. The mechanisms underlying HR regulation during DNA replication are obscure. Here, we find that RTEL1 helicase, RAD51, and RAD51 paralogs are enriched at stalled replication sites. The absence of RTEL1 leads to an increase in the RAD51-mediated HR and fork reversal during replication and affects genome-wide replication, which can be rescued by co-depleting RAD51 and RAD51 paralogs. Interestingly, co-depletion of fork remodelers such as SMARCAL1/ZRANB3/HLTF/FBH1 and expression of HR-defective RAD51 mutants also rescues replication defects in RTEL1-deficient cells. The anti-recombinase function of RTEL1 during replication depends on its interaction with PCNA and helicase activity. Together, our data identify the role of RTEL1 helicase in restricting RAD51-mediated fork reversal and HR activity to facilitate error-free genome duplication.


Subject(s)
DNA Helicases , DNA Replication , Homologous Recombination , Rad51 Recombinase , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Humans , Proliferating Cell Nuclear Antigen/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Breaks, Double-Stranded , Genomic Instability
10.
Nat Commun ; 15(1): 6843, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39122671

ABSTRACT

Despite the potential of small molecules and recombinant proteins to enhance the efficiency of homology-directed repair (HDR), single-stranded DNA (ssDNA) donors, as currently designed and chemically modified, remain suboptimal for precise gene editing. Here, we screen the biased ssDNA binding sequences of DNA repair-related proteins and engineer RAD51-preferred sequences into HDR-boosting modules for ssDNA donors. Donors with these modules exhibit an augmented affinity for RAD51, thereby enhancing HDR efficiency across various genomic loci and cell types when cooperated with Cas9, nCas9, and Cas12a. By combining with an inhibitor of non-homologous end joining (NHEJ) or the HDRobust strategy, these modular ssDNA donors achieve up to 90.03% (median 74.81%) HDR efficiency. The HDR-boosting modules targeting an endogenous protein enable a chemical modification-free strategy to improve the efficacy of ssDNA donors for precise gene editing.


Subject(s)
DNA, Single-Stranded , Gene Editing , Rad51 Recombinase , Recombinational DNA Repair , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Humans , Gene Editing/methods , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , CRISPR-Cas Systems , HEK293 Cells , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , DNA End-Joining Repair
11.
Nat Commun ; 15(1): 7015, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147779

ABSTRACT

During meiosis, nucleoprotein filaments of the strand exchange proteins RAD51 and DMC1 are crucial for repairing SPO11-generated DNA double-strand breaks (DSBs) by homologous recombination (HR). A balanced activity of positive and negative RAD51/DMC1 regulators ensures proper recombination. Fidgetin-like 1 (FIGNL1) was previously shown to negatively regulate RAD51 in human cells. However, FIGNL1's role during meiotic recombination in mammals remains unknown. Here, we decipher the meiotic functions of FIGNL1 and FIGNL1 Interacting Regulator of Recombination and Mitosis (FIRRM) using male germline-specific conditional knock-out (cKO) mouse models. Both FIGNL1 and FIRRM are required for completing meiotic prophase in mouse spermatocytes. Despite efficient recruitment of DMC1 on ssDNA at meiotic DSB hotspots, the formation of late recombination intermediates is defective in Firrm cKO and Fignl1 cKO spermatocytes. Moreover, the FIGNL1-FIRRM complex limits RAD51 and DMC1 accumulation on intact chromatin, independently from the formation of SPO11-catalyzed DSBs. Purified human FIGNL1ΔN alters the RAD51/DMC1 nucleoprotein filament structure and inhibits strand invasion in vitro. Thus, this complex might regulate RAD51 and DMC1 association at sites of meiotic DSBs to promote proficient strand invasion and processing of recombination intermediates.


Subject(s)
Cell Cycle Proteins , DNA Breaks, Double-Stranded , DNA-Binding Proteins , Meiosis , Mice, Knockout , Rad51 Recombinase , Spermatocytes , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Animals , Male , Meiosis/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Humans , Mice , Spermatocytes/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Homologous Recombination , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA Damage , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Chromatin/metabolism , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics
12.
Mol Cell ; 84(16): 3026-3043.e11, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39178838

ABSTRACT

Abasic sites are DNA lesions repaired by base excision repair. Cleavage of unrepaired abasic sites in single-stranded DNA (ssDNA) can lead to chromosomal breakage during DNA replication. How rupture of abasic DNA is prevented remains poorly understood. Here, using cryoelectron microscopy (cryo-EM), Xenopus laevis egg extracts, and human cells, we show that RAD51 nucleofilaments specifically recognize and protect abasic sites, which increase RAD51 association rate to DNA. In the absence of BRCA2 or RAD51, abasic sites accumulate as a result of DNA base methylation, oxidation, and deamination, inducing abasic ssDNA gaps that make replicating DNA fibers sensitive to APE1. RAD51 assembled on abasic DNA prevents abasic site cleavage by the MRE11-RAD50 complex, suppressing replication fork breakage triggered by an excess of abasic sites or POLθ polymerase inhibition. Our study highlights the critical role of BRCA2 and RAD51 in safeguarding against unrepaired abasic sites in DNA templates stemming from base alterations, ensuring genomic stability.


Subject(s)
BRCA2 Protein , DNA Damage , DNA Repair , DNA Replication , DNA, Single-Stranded , Rad51 Recombinase , Xenopus laevis , Humans , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Animals , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Cryoelectron Microscopy , DNA Polymerase theta , DNA Methylation , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
13.
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
14.
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
15.
Cell Death Dis ; 15(7): 551, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085197

ABSTRACT

PLK1 is currently at the forefront of mitotic research and has emerged as a potential target for small cell lung cancer (SCLC) therapy. However, the factors influencing the efficacy of PLK1 inhibitors remain unclear. Herein, BRCA1 was identified as a key factor affecting the response of SCLC cells to BI-2536. Targeting AURKA with alisertib, at a non-toxic concentration, reduced the BI-2536-induced accumulation of BRCA1 and RAD51, leading to DNA repair defects and mitotic cell death in SCLC cells. In vivo experiments confirmed that combining BI-2536 with alisertib impaired DNA repair capacity and significantly delayed tumor growth. Additionally, GSEA analysis and loss- and gain-of-function assays demonstrated that MYC/MYCN signaling is crucial for determining the sensitivity of SCLC cells to BI-2536 and its combination with alisertib. The study further revealed a positive correlation between RAD51 expression and PLK1/AURKA expression, and a negative correlation with the IC50 values of BI-2536. Manipulating RAD51 expression significantly influenced the efficacy of BI-2536 and restored the MYC/MYCN-induced enhancement of BI-2536 sensitivity in SCLC cells. Our findings indicate that the BRCA1 and MYC/MYCN-RAD51 axes govern the response of small cell lung cancer to BI-2536 and its combination with alisertib. This study propose the combined use of BI-2536 and alisertib as a novel therapeutic strategy for the treatment of SCLC patients with MYC/MYCN activation.


Subject(s)
Azepines , BRCA1 Protein , Lung Neoplasms , Proto-Oncogene Proteins c-myc , Pyrimidines , Small Cell Lung Carcinoma , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Animals , Cell Line, Tumor , Azepines/pharmacology , Aurora Kinase A/metabolism , Aurora Kinase A/antagonists & inhibitors , Rad51 Recombinase/metabolism , Mice , Mice, Nude , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Polo-Like Kinase 1 , DNA Repair/drug effects , Female , Xenograft Model Antitumor Assays , Pteridines
16.
DNA Repair (Amst) ; 141: 103738, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084178

ABSTRACT

A key but often neglected component of genomic instability is the emergence of single-stranded DNA (ssDNA) gaps during DNA replication in the absence of functional homologous recombination (HR) proteins, such as RAD51 and BRCA1/2. Research in prokaryotes has shed light on the dual role of RAD51's bacterial ortholog, RecA, in HR and the protection of replication forks, emphasizing its essential role in preventing the formation of ssDNA gaps, which is vital for cellular viability. This phenomenon was corroborated in eukaryotic cells deficient in HR, where the formation of ssDNA gaps within newly synthesized DNA and their subsequent processing by the MRE11 nuclease were observed. Without functional HR proteins, cells employ alternative ssDNA gap-filling mechanisms to ensure survival, though this compensatory response can compromise genomic stability. A notable example is the involvement of the translesion synthesis (TLS) polymerase POLζ, along with the repair protein POLθ, in the suppression of replicative ssDNA gaps. Persistent ssDNA gaps may result in replication fork collapse, chromosomal anomalies, and cell death, which contribute to cancer progression and resistance to therapy. Elucidating the processes that avert ssDNA gaps and safeguard replication forks is critical for enhancing cancer treatment approaches by exploiting the vulnerabilities of cancer cells in these pathways.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA Replication , DNA, Single-Stranded , Rad51 Recombinase , Humans , Rad51 Recombinase/metabolism , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , DNA, Single-Stranded/metabolism , BRCA1 Protein/metabolism , Homologous Recombination , Genomic Instability , MRE11 Homologue Protein/metabolism , Animals , DNA Repair
17.
Toxicol In Vitro ; 100: 105901, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39029599

ABSTRACT

Hydroquinone (HQ) is one of benzene metabolites that can cause oxidative stress damage and Homologous recombination repair (HR). A good deal of reactive oxygen species (ROS) generated by oxidative stress can trigger apoptotic signaling pathways. The nuclear factor erythroid 2-related factor 2 (Nrf2) can regulate the cell response to oxidative stress damage. The aim of this study was to explore whether Nrf2 participate in HQ-induced apoptosis and its mechanism. The findings displayed that HQ triggered HR, promoted Nrf2 transfer into the cell nucleus and induced cell apoptosis, while Nrf2 deficient elevated cell apoptosis, attenuated the expression of PARP1 and RAD51. We also observed that Nrf2 deficient triggered Caspase-9. Thus, we speculated that Nrf2 might participate in HQ-induced cell apoptosis through Caspase-9 dependent pathways. Meanwhile, Nrf2 participated in HQ-induced DNA damage repair by regulating the level of PARP1 and RAD51.


Subject(s)
Apoptosis , DNA Damage , Hydroquinones , NF-E2-Related Factor 2 , Poly (ADP-Ribose) Polymerase-1 , Rad51 Recombinase , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Hydroquinones/toxicity , Apoptosis/drug effects , Humans , Cell Line , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Caspase 9/metabolism , DNA Repair/drug effects
19.
Nucleic Acids Res ; 52(16): 9695-9709, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39082275

ABSTRACT

Inappropriate homology-directed repair (HDR) of telomeres results in catastrophic telomere loss and aberrant chromosome fusions, leading to genome instability. We have previously shown that the TRF2-RAP1 heterodimer protects telomeres from engaging in aberrant telomere HDR. Cells lacking the basic domain of TRF2 and functional RAP1 display HDR-mediated telomere clustering, resulting in the formation of ultrabright telomeres (UTs) and massive chromosome fusions. Using purified proteins, we uncover three distinct molecular pathways that the TRF2-RAP1 heterodimer utilizes to protect telomeres from engaging in aberrant HDR. We show mechanistically that TRF2-RAP1 inhibits RAD51-initiated telomeric D-loop formation. Both the TRF2 basic domain and RAP1-binding to TRF2 are required to block RAD51-mediated homology search. TRF2 recruits the BLM helicase to telomeres through its TRFH domain to promote BLM-mediated unwinding of telomere D-loops. In addition, TRF2-RAP1 inhibits BLM-DNA2-mediated 5' telomere end resection, preventing the generation of 3' single-stranded telomere overhangs necessary for RAD51-dependent HDR. Importantly, cells expressing BLM mutants unable to interact with TRF2 accumulate telomere D-loops and UTs. Our findings uncover distinct molecular mechanisms coordinated by TRF2-RAP1 to protect telomeres from engaging in aberrant HDR.


Subject(s)
Rad51 Recombinase , RecQ Helicases , Recombinational DNA Repair , Shelterin Complex , Telomere-Binding Proteins , Telomere , Telomeric Repeat Binding Protein 2 , Telomeric Repeat Binding Protein 2/metabolism , Telomeric Repeat Binding Protein 2/genetics , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism , RecQ Helicases/genetics , Telomere/metabolism , Shelterin Complex/metabolism , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Humans , Protein Binding , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
20.
Molecules ; 29(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39064836

ABSTRACT

Genotoxic substances widely exist in the environment and the food supply, posing serious health risks due to their potential to induce DNA damage and cancer. Traditional genotoxicity assays, while valuable, are limited by insufficient sensitivity, specificity, and efficiency, particularly when applied to complex food matrices. This study introduces a multiparametric high-content analysis (HCA) for the detection of genotoxic substances in complex food matrices. The developed assay measures three genotoxic biomarkers, including γ-H2AX, p-H3, and RAD51, which enhances the sensitivity and accuracy of genotoxicity screening. Moreover, the assay effectively distinguishes genotoxic compounds with different modes of action, which not only offers a more comprehensive assessment of DNA damage and the cellular response to genotoxic stress but also provides new insights into the exploration of genotoxicity mechanisms. Notably, the five tested food matrices, including coffee, tea, pak choi, spinach, and tomato, were found not to interfere with the detection of these biomarkers under proper dilution ratios, validating the robustness and reliability of the assay for the screening of genotoxic compounds in the food industry. The integration of multiple biomarkers with HCA provides an efficient method for detecting and assessing genotoxic substances in the food supply, with potential applications in toxicology research and food safety.


Subject(s)
DNA Damage , Mutagenicity Tests , Mutagens , Mutagens/analysis , Mutagens/toxicity , Mutagenicity Tests/methods , Humans , Food Analysis/methods , Tea/chemistry , Biomarkers , Solanum lycopersicum/chemistry , Histones/metabolism , Histones/analysis , Coffee/chemistry , Spinacia oleracea/chemistry , Rad51 Recombinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL