Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 932
Filter
1.
BMC Bioinformatics ; 25(1): 236, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997639

ABSTRACT

BACKGROUND: Homologous recombination deficiency (HRD) stands as a clinical indicator for discerning responsive outcomes to platinum-based chemotherapy and poly ADP-ribose polymerase (PARP) inhibitors. One of the conventional approaches to HRD prognostication has generally centered on identifying deleterious mutations within the BRCA1/2 genes, along with quantifying the genomic scars, such as Genomic Instability Score (GIS) estimation with scarHRD. However, the scarHRD method has limitations in scenarios involving tumors bereft of corresponding germline data. Although several RNA-seq-based HRD prediction algorithms have been developed, they mainly support cohort-wise classification, thereby yielding HRD status without furnishing an analogous quantitative metric akin to scarHRD. This study introduces the expHRD method, which operates as a novel transcriptome-based framework tailored to n-of-1-style HRD scoring. RESULTS: The prediction model has been established using the elastic net regression method in the Cancer Genome Atlas (TCGA) pan-cancer training set. The bootstrap technique derived the HRD geneset for applying the expHRD calculation. The expHRD demonstrated a notable correlation with scarHRD and superior performance in predicting HRD-high samples. We also performed intra- and extra-cohort evaluations for clinical feasibility in the TCGA-OV and the Genomic Data Commons (GDC) ovarian cancer cohort, respectively. The innovative web service designed for ease of use is poised to extend the realms of HRD prediction across diverse malignancies, with ovarian cancer standing as an emblematic example. CONCLUSIONS: Our novel approach leverages the transcriptome data, enabling the prediction of HRD status with remarkable precision. This innovative method addresses the challenges associated with limited available data, opening new avenues for utilizing transcriptomics to inform clinical decisions.


Subject(s)
Homologous Recombination , Neoplasms , Transcriptome , Humans , Transcriptome/genetics , Homologous Recombination/genetics , Neoplasms/genetics , Algorithms , Female , Gene Expression Profiling/methods
2.
Fungal Genet Biol ; 173: 103910, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38897560

ABSTRACT

The homologous recombination strategy has a long history of editing Saccharomyces cerevisiae target genes. The application of CRISPR/Cas9 strategy to editing target genes in S. cerevisiae has also received a lot of attention in recent years. All findings seem to indicate that editing relevant target genes in S. cerevisiae is an extremely easy event. In this study, we systematically analyzed the advantages and disadvantages of homologous recombination (HR) strategy, CRISPR/Cas9 strategy, and CRISPR/Cas9 combined homology-mediated repair (CRISPR/Case9-HDR) strategy in knocking out BY4742 ade2. Our data showed that when the ade2 was knocked out by HR strategy, a large number of clones appeared to be off-target, and 10 %-80 % of the so-called knockout clones obtained were heteroclones. When the CRISPR/Cas9 strategy was applied, 60% of clones were off-target and the rest were all heteroclones. Interestingly, most of the cells were edited successfully, but at least 60 % of the clones were heteroclones, when the CRISPR/Cas9-HDR strategy was employed. Our results clearly showed that the emergence of heteroclone seems inevitable regardless of the strategies used for editing BY4742 ade2. Given the characteristics of BY4742 defective in ade2 showing red on the YPD plate, we attempted to build an efficient yeast gene editing strategy, in which the CRISPR/Cas9 combines homology-mediated repair template carrying an ade2 expression cassette, BY4742(ade2Δ0) as the start strain. We used this strategy to successfully achieve 100 % knockout efficiency of trp1, indicating that technical challenges of how to easily screen out pure knockout clones without color phenotype have been solved. Our data showed in this study not only establishes an efficient yeast gene knockout strategy with dual auxotrophy coupled red labeling but also provides new ideas and references for the knockout of target genes in the monokaryotic mycelium of macrofungi.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Homologous Recombination , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Gene Editing/methods , CRISPR-Cas Systems/genetics , Homologous Recombination/genetics , Genome, Fungal/genetics , Gene Knockout Techniques/methods , Saccharomyces cerevisiae Proteins/genetics
3.
Nucleic Acids Res ; 52(13): 7401-7413, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38869071

ABSTRACT

Homologous recombination (HR) is a key process for repairing DNA double strand breaks and for promoting genetic diversity. However, HR occurs unevenly across the genome, and certain genomic features can influence its activity. One such feature is the presence of guanine quadruplexes (G4s), stable secondary structures widely distributed throughout the genome. These G4s play essential roles in gene transcription and genome stability regulation. Especially, elevated G4 levels in cells deficient in the Bloom syndrome helicase (BLM) significantly enhance HR at G4 sites, potentially threatening genome stability. Here, we investigated the role of G4-binding protein Yin Yang-1 (YY1) in modulating HR at G4 sites in human cells. Our results show that YY1's binding to G4 structures suppresses sister chromatid exchange after BLM knockdown, and YY1's chromatin occupancy negatively correlates with the overall HR rate observed across the genome. By limiting RAD51 homolog 1 (RAD51) access, YY1 preferentially binds to essential genomic regions, shielding them from excessive HR. Our findings unveil a novel role of YY1-G4 interaction, revealing novel insights into cellular mechanisms involved in HR regulation.


Subject(s)
G-Quadruplexes , Homologous Recombination , Rad51 Recombinase , Sister Chromatid Exchange , YY1 Transcription Factor , Humans , YY1 Transcription Factor/metabolism , YY1 Transcription Factor/genetics , Homologous Recombination/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Sister Chromatid Exchange/genetics , RecQ Helicases/metabolism , RecQ Helicases/genetics , Chromatin/metabolism , Chromatin/genetics , DNA Breaks, Double-Stranded , Genomic Instability/genetics
4.
Nucleic Acids Res ; 52(13): 7687-7703, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38884202

ABSTRACT

RAD18 is an E3 ubiquitin ligase that prevents replication fork collapse by promoting DNA translesion synthesis and template switching. Besides this classical role, RAD18 has been implicated in homologous recombination; however, this function is incompletely understood. Here, we show that RAD18 is recruited to DNA lesions by monoubiquitination of histone H2A at K15 and counteracts accumulation of 53BP1. Super-resolution microscopy revealed that RAD18 localizes to the proximity of DNA double strand breaks and limits the distribution of 53BP1 to the peripheral chromatin nanodomains. Whereas auto-ubiquitination of RAD18 mediated by RAD6 inhibits its recruitment to DNA breaks, interaction with SLF1 promotes RAD18 accumulation at DNA breaks in the post-replicative chromatin by recognition of histone H4K20me0. Surprisingly, suppression of 53BP1 function by RAD18 is not involved in homologous recombination and rather leads to reduction of non-homologous end joining. Instead, we provide evidence that RAD18 promotes HR repair by recruiting the SMC5/6 complex to DNA breaks. Finally, we identified several new loss-of-function mutations in RAD18 in cancer patients suggesting that RAD18 could be involved in cancer development.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , DNA-Binding Proteins , Histones , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Chromatin/metabolism , Chromatin/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Histones/metabolism , Homologous Recombination/genetics , Recombinational DNA Repair , DNA Replication , DNA Repair , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA End-Joining Repair
5.
Nucleic Acids Res ; 52(12): 6994-7011, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38828775

ABSTRACT

The clinical success of PARP1/2 inhibitors (PARPi) prompts the expansion of their applicability beyond homologous recombination deficiency. Here, we demonstrate that the loss of the accessory subunits of DNA polymerase epsilon, POLE3 and POLE4, sensitizes cells to PARPi. We show that the sensitivity of POLE4 knockouts is not due to compromised response to DNA damage or homologous recombination deficiency. Instead, POLE4 loss affects replication speed leading to the accumulation of single-stranded DNA gaps behind replication forks upon PARPi treatment, due to impaired post-replicative repair. POLE4 knockouts elicit elevated replication stress signaling involving ATR and DNA-PK. We find POLE4 to act parallel to BRCA1 in inducing sensitivity to PARPi and counteracts acquired resistance associated with restoration of homologous recombination. Altogether, our findings establish POLE4 as a promising target to improve PARPi driven therapies and hamper acquired PARPi resistance.


Subject(s)
BRCA1 Protein , DNA Polymerase II , DNA Replication , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Humans , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Polymerase II/metabolism , DNA Polymerase II/genetics , DNA Replication/drug effects , DNA Damage , Cell Line, Tumor , Homologous Recombination/genetics , Homologous Recombination/drug effects , Drug Resistance, Neoplasm/genetics
6.
Nucleic Acids Res ; 52(11): 6518-6531, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38783157

ABSTRACT

Precise genomic editing through the combination of CRISPR/Cas systems and recombinant adeno-associated virus (rAAV)-delivered homology directed repair (HDR) donor templates represents a powerful approach. However, the challenge of effectively suppressing leaky transcription from the rAAV vector, a phenomenon associated to cytotoxicity, persists. In this study, we demonstrated substantial promoter activities of various homology arms and inverted terminal repeats (ITR). To address this issue, we identified a novel rAAV variant, Y704T, which not only yields high-vector quantities but also effectively suppresses in cis mRNA transcription driven by a robust promoter. The Y704T variant maintains normal functionality in receptor interaction, intracellular trafficking, nuclear entry, uncoating, and second-strand synthesis, while specifically exhibiting defects in transcription. Importantly, this inhibitory effect is found to be independent of ITR, promoter types, and RNA polymerases. Mechanistic studies unveiled the involvement of Valosin Containing Protein (VCP/p97) in capsid-mediated transcription repression. Remarkably, the Y704T variant delivers HDR donor templates without compromising DNA replication ability and homologous recombination efficiency. In summary, our findings enhance the understanding of capsid-regulated transcription and introduce novel avenues for the application of the rAAV-CRISPR/Cas9 system in human gene therapy.


Subject(s)
Dependovirus , Gene Editing , Homologous Recombination , Promoter Regions, Genetic , Dependovirus/genetics , Humans , Promoter Regions, Genetic/genetics , Gene Editing/methods , Homologous Recombination/genetics , HEK293 Cells , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid/metabolism , Mutation , Genetic Vectors/genetics , Transcription, Genetic , CRISPR-Cas Systems , Recombinational DNA Repair , Terminal Repeat Sequences/genetics , DNA Replication/genetics
7.
STAR Protoc ; 5(2): 103016, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38640065

ABSTRACT

Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.


Subject(s)
CRISPR-Cas Systems , Erythroid Cells , Gene Editing , Gene Knock-In Techniques , CRISPR-Cas Systems/genetics , Animals , Mice , Erythroid Cells/metabolism , Erythroid Cells/cytology , Gene Knock-In Techniques/methods , Gene Editing/methods , Transfection/methods , Cell Line , Homologous Recombination/genetics , Electroporation/methods
8.
Cell Cycle ; 23(4): 369-384, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38571319

ABSTRACT

Acetaldehyde, a chemical that can cause DNA damage and contribute to cancer, is prevalently present in our environment, e.g. in alcohol, tobacco, and food. Although aldehyde potentially promotes crosslinking reactions among biological substances including DNA, RNA, and protein, it remains unclear what types of DNA damage are caused by acetaldehyde and how they are repaired. In this study, we explored mechanisms involved in the repair of acetaldehyde-induced DNA damage by examining the cellular sensitivity to acetaldehyde in the collection of human TK6 mutant deficient in each genome maintenance system. Among the mutants, mismatch repair mutants did not show hypersensitivity to acetaldehyde, while mutants deficient in base and nucleotide excision repair pathways or homologous recombination (HR) exhibited higher sensitivity to acetaldehyde than did wild-type cells. We found that acetaldehyde-induced RAD51 foci representing HR intermediates were prolonged in HR-deficient cells. These results indicate a pivotal role of HR in the repair of acetaldehyde-induced DNA damage. These results suggest that acetaldehyde causes complex DNA damages that require various types of repair pathways. Mutants deficient in the removal of protein adducts from DNA ends such as TDP1-/- and TDP2-/- cells exhibited hypersensitivity to acetaldehyde. Strikingly, the double mutant deficient in both TDP1 and RAD54 showed similar sensitivity to each single mutant. This epistatic relationship between TDP1-/- and RAD54-/- suggests that the protein-DNA adducts generated by acetaldehyde need to be removed for efficient repair by HR. Our study would help understand the molecular mechanism of the genotoxic and mutagenic effects of acetaldehyde.


Subject(s)
Acetaldehyde , DNA Damage , DNA Repair , Homologous Recombination , Acetaldehyde/toxicity , Humans , Homologous Recombination/drug effects , Homologous Recombination/genetics , DNA Repair/drug effects , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Mutation/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Line
9.
Nucleic Acids Res ; 52(10): 5698-5719, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38587186

ABSTRACT

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Subject(s)
Chromatin , DNA Repair , DNA-Binding Proteins , Immunity , Transcription Factors , Humans , Cell Line, Tumor , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Homologous Recombination/genetics , Immunity/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/immunology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Trans-Activators , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Nucleic Acids Res ; 52(10): 5774-5791, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38597669

ABSTRACT

RAD51 filament is crucial for the homology-dependent repair of DNA double-strand breaks and stalled DNA replication fork protection. Positive and negative regulators control RAD51 filament assembly and disassembly. RAD51 is vital for genome integrity but excessive accumulation of RAD51 on chromatin causes genome instability and growth defects. However, the detailed mechanism underlying RAD51 disassembly by negative regulators and the physiological consequence of abnormal RAD51 persistence remain largely unknown. Here, we report the role of the human AAA+ ATPase FIGNL1 in suppressing a novel type of RAD51-mediated genome instability. FIGNL1 knockout human cells were defective in RAD51 dissociation after replication fork restart and accumulated ultra-fine chromosome bridges (UFBs), whose formation depends on RAD51 rather than replication fork stalling. FIGNL1 suppresses homologous recombination intermediate-like UFBs generated between sister chromatids at genomic loci with repeated sequences such as telomeres and centromeres. These data suggest that RAD51 persistence per se induces the formation of unresolved linkage between sister chromatids resulting in catastrophic genome instability. FIGNL1 facilitates post-replicative disassembly of RAD51 filament to suppress abnormal recombination intermediates and UFBs. These findings implicate FIGNL1 as a key factor required for active RAD51 removal after processing of stalled replication forks, which is essential to maintain genome stability.


Subject(s)
Genomic Instability , Rad51 Recombinase , Humans , Chromatids/metabolism , Chromatids/genetics , DNA Replication/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Cell Line , Gene Knockout Techniques
11.
PLoS Genet ; 20(4): e1011250, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683763

ABSTRACT

Accurate repair of DNA double-strand breaks (DSBs) is essential for the maintenance of genome integrity, as failure to repair DSBs can result in cell death. The cell has evolved two main mechanisms for DSB repair: non-homologous end-joining (NHEJ) and homology-directed repair (HDR), which includes single-strand annealing (SSA) and homologous recombination (HR). While certain factors like age and state of the chromatin are known to influence DSB repair pathway choice, the roles of developmental stage, tissue type, and sex have yet to be elucidated in multicellular organisms. To examine the influence of these factors, DSB repair in various embryonic developmental stages, larva, and adult tissues in Drosophila melanogaster was analyzed through molecular analysis of the DR-white assay using Tracking across Indels by DEcomposition (TIDE). The proportion of HR repair was highest in tissues that maintain the canonical (G1/S/G2/M) cell cycle and suppressed in both terminally differentiated and polyploid tissues. To determine the impact of sex on repair pathway choice, repair in different tissues in both males and females was analyzed. When molecularly examining tissues containing mostly somatic cells, males and females demonstrated similar proportions of HR and NHEJ. However, when DSB repair was analyzed in male and female premeiotic germline cells utilizing phenotypic analysis of the DR-white assay, there was a significant decrease in HR in females compared to males. This study describes the impact of development, tissue-specific cycling profile, and, in some cases, sex on DSB repair outcomes, underscoring the complexity of repair in multicellular organisms.


Subject(s)
DNA Breaks, Double-Stranded , Drosophila melanogaster , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Male , DNA Repair/genetics , DNA End-Joining Repair/genetics , Recombinational DNA Repair , Homologous Recombination/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Cell Cycle/genetics
12.
JAMA Netw Open ; 7(4): e247811, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38648056

ABSTRACT

Importance: RAD51C and RAD51D are involved in DNA repair by homologous recombination. Germline pathogenic variants (PVs) in these genes are associated with an increased risk of ovarian and breast cancer. Understanding the homologous recombination deficiency (HRD) status of tumors from patients with germline PVs in RAD51C/D could guide therapeutic decision-making and improve survival. Objective: To characterize the clinical and tumor characteristics of germline RAD51C/D PV carriers, including the evaluation of HRD status. Design, Setting, and Participants: This retrospective cohort study included 91 index patients plus 90 relatives carrying germline RAD51C/D PV (n = 181) in Spanish hospitals from January 1, 2014, to December 31, 2021. Genomic and functional HRD biomarkers were assessed in untreated breast and ovarian tumor samples (n = 45) from June 2022 to February 2023. Main Outcomes and Measures: Clinical and pathologic characteristics were assessed using descriptive statistics. Genomic HRD by genomic instability scores, functional HRD by RAD51, and gene-specific loss of heterozygosity were analyzed. Associations between HRD status and tumor subtype, age at diagnosis, and gene-specific loss of heterozygosity in RAD51C/D were investigated using logistic regression or the t test. Results: A total of 9507 index patients were reviewed, and 91 patients (1.0%) were found to carry a PV in RAD51C/D; 90 family members with a germline PV in RAD51C/D were also included. A total of 157 of carriers (86.7%) were women and 181 (55.8%) had received a diagnosis of cancer, mainly breast cancer or ovarian cancer. The most prevalent PVs were c.1026+5_1026+7del (11 of 56 [19.6%]) and c.709C>T (9 of 56 [16.1%]) in RAD51C and c.694C>T (20 of 35 [57.1%]) in RAD51D. In untreated breast cancer and ovarian cancer, the prevalence of functional and genomic HRD was 55.2% (16 of 29) and 61.1% (11 of 18) for RAD51C, respectively, and 66.7% (6 of 9) and 90.0% (9 of 10) for RAD51D. The concordance between HRD biomarkers was 91%. Tumors with the same PV displayed contrasting HRD status, and age at diagnosis did not correlate with the occurrence of HRD. All breast cancers retaining the wild-type allele were estrogen receptor positive and lacked HRD. Conclusions and Relevance: In this cohort study of germline RAD51C/D breast cancer and ovarian cancer, less than 70% of tumors displayed functional HRD, and half of those that did not display HRD were explained by retention of the wild-type allele, which was more frequent among estrogen receptor-positive breast cancers. Understanding which tumors are associated with RAD51C/D and HRD is key to identify patients who can benefit from targeted therapies, such as PARP (poly [adenosine diphosphate-ribose] polymerase) inhibitors.


Subject(s)
Breast Neoplasms , Germ-Line Mutation , Homologous Recombination , Ovarian Neoplasms , Rad51 Recombinase , Adult , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/epidemiology , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Homologous Recombination/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/epidemiology , Prevalence , Retrospective Studies , Spain/epidemiology , Rad51 Recombinase/genetics
13.
Trends Genet ; 40(6): 467-470, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38494375

ABSTRACT

DNA repair through homologous recombination (HR) is of vital importance for maintaining genome stability and preventing tumorigenesis. RAD51 is the core component of HR, catalyzing the strand invasion and homology search. Here, we highlight recent findings on FIRRM and FIGNL1 as regulators of the dynamics of RAD51.


Subject(s)
Homologous Recombination , Rad51 Recombinase , Homologous Recombination/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Humans , DNA Repair/genetics , Genomic Instability/genetics , Animals
14.
Nucleic Acids Res ; 52(9): 5138-5151, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38554108

ABSTRACT

Recycling of parental histones is an important step in epigenetic inheritance. During DNA replication, DNA polymerase epsilon subunit DPB3/DPB4 and DNA replication helicase subunit MCM2 are involved in the transfer of parental histones to the leading and lagging strands, respectively. Single Dpb3 deletion (dpb3Δ) or Mcm2 mutation (mcm2-3A), which each disrupts one parental histone transfer pathway, leads to the other's predominance. However, the biological impact of the two histone transfer pathways on chromatin structure and DNA repair remains elusive. In this study, we used budding yeast Saccharomyces cerevisiae to determine the genetic and epigenetic outcomes from disruption of parental histone H3-H4 tetramer transfer. We found that a dpb3Δ mcm2-3A double mutant did not exhibit the asymmetric parental histone patterns caused by a single dpb3Δ or mcm2-3A mutation, suggesting that the processes by which parental histones are transferred to the leading and lagging strands are independent. Surprisingly, the frequency of homologous recombination was significantly lower in dpb3Δ, mcm2-3A and dpb3Δ mcm2-3A mutants, likely due to the elevated levels of free histones detected in the mutant cells. Together, these findings indicate that proper transfer of parental histones during DNA replication is essential for maintaining chromatin structure and that lower homologous recombination activity due to parental histone transfer defects is detrimental to cells.


Subject(s)
DNA Replication , Histones , Homologous Recombination , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Histones/metabolism , Histones/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Homologous Recombination/genetics , DNA Replication/genetics , Mutation , Chromatin/metabolism , Chromatin/genetics , DNA Polymerase II/metabolism , DNA Polymerase II/genetics , Epigenesis, Genetic , DNA Repair
15.
JCO Precis Oncol ; 8: e2300483, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38427930

ABSTRACT

PURPOSE: To meet the urgent need for accessible homologous recombination-deficient (HRD) test options, we validated a laboratory-developed test (LDT) and a functional RAD51 assay to assess patients with ovarian cancer and predict the clinical benefit of poly(ADP-ribose) polymerase inhibitor therapy. METHODS: Optimization of the LDT cutoff and validation on the basis of samples from 91 patients enrolled in the ENGOT-ov24/NSGO-AVANOVA1&2 trial (ClinicalTrials.gov identifier: NCT02354131), previously subjected to commercial CDx HRD testing (CDx). RAD51 foci analysis was performed and tumors with ≥five foci/nucleus were classified as RAD51-positive (homologous recombination-proficient). RESULTS: The optimal LDT cutoff is 54. Comparing CDx genome instability score and LDT HRD scores show a Spearman's correlation of rho = 0.764 (P < .0001). Cross-tabulation analysis shows that the sensitivity of the LDT HRD score is 86% and of the LDT HRD status is 91.8% (Fisher's exact test P < .001). Survival analysis on progression-free survival (PFS) of LDT-assessed patients show a Cox regression P < .05. RAD51 assays show a correlation between low RAD51 foci detection (<20% RAD51+ cells) and significantly prolonged PFS (P < .001). CONCLUSION: The robust concordance between the open standard LDT and the CDx, especially the correlation with PFS, warrants future validation and implementation of the open standard LDT for HRD testing in diagnostic settings.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Homologous Recombination/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Progression-Free Survival , Rad51 Recombinase/genetics
16.
Proc Natl Acad Sci U S A ; 121(12): e2312820121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38478689

ABSTRACT

Meiotic recombination shows broad variations across species and along chromosomes and is often suppressed at and around genomic regions determining sexual compatibility such as mating type loci in fungi. Here, we show that the absence of Spo11-DSBs and meiotic recombination on Lakl0C-left, the chromosome arm containing the sex locus of the Lachancea kluyveri budding yeast, results from the absence of recruitment of the two chromosome axis proteins Red1 and Hop1, essential for proper Spo11-DSBs formation. Furthermore, cytological observation of spread pachytene meiotic chromosomes reveals that Lakl0C-left does not undergo synapsis. However, we show that the behavior of Lakl0C-left is independent of its particularly early replication timing and is not accompanied by any peculiar chromosome structure as detectable by Hi-C in this yet poorly studied yeast. Finally, we observed an accumulation of heterozygous mutations on Lakl0C-left and a sexual dimorphism of the haploid meiotic offspring, supporting a direct effect of this absence of meiotic recombination on L. kluyveri genome evolution and fitness. Because suppression of meiotic recombination on sex chromosomes is widely observed across eukaryotes, the mechanism for recombination suppression described here may apply to other species, with the potential to impact sex chromosome evolution.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomycetales , Chromosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomycetales/genetics , Saccharomycetales/metabolism , Homologous Recombination/genetics , Meiosis/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
17.
JCO Precis Oncol ; 8: e2300348, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38513168

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase inhibitors (PARPi) have shown promising clinical results in the treatment of ovarian cancer. Analysis of biomarker subgroups consistently revealed higher benefits for patients with homologous recombination deficiency (HRD). The test that is most often used for the detection of HRD in clinical studies is the Myriad myChoice assay. However, other assays can also be used to assess biomarkers, which are indicative of HRD, genomic instability (GI), and BRCA1/2 mutation status. Many of these assays have high potential to be broadly applied in clinical routine diagnostics in a time-effective decentralized manner. Here, we compare the performance of a multitude of alternative assays in comparison with Myriad myChoice in high-grade serous ovarian cancer (HGSOC). METHODS: DNA from HGSOC samples was extracted from formalin-fixed paraffin-embedded tissue blocks of cases previously run with the Myriad myChoice assay, and GI was measured by multiple molecular assays (CytoSNP, AmoyDx, Illumina TSO500 HRD, OncoScan, NOGGO GISv1, QIAseq HRD Panel and whole genome sequencing), applying different bioinformatics algorithms. RESULTS: Application of different assays to assess GI, including Myriad myChoice, revealed high concordance of the generated scores ranging from very substantial to nearly perfect fit, depending on the assay and bioinformatics pipelines applied. Interlaboratory comparison of assays also showed high concordance of GI scores. CONCLUSION: Assays for GI assessment not only show a high concordance with each other but also in correlation with Myriad myChoice. Thus, almost all of the assays included here can be used effectively to assess HRD-associated GI in the clinical setting. This is important as PARPi treatment on the basis of these tests is compliant with European Medicines Agency approvals, which are methodologically not test-bound.


Subject(s)
BRCA1 Protein , Ovarian Neoplasms , Humans , Female , BRCA1 Protein/genetics , Mutation , BRCA2 Protein/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Genomic Instability/genetics , Homologous Recombination/genetics
19.
Genes (Basel) ; 15(2)2024 01 26.
Article in English | MEDLINE | ID: mdl-38397152

ABSTRACT

Genomic instability is one of the well-established hallmarks of cancer. The homologous recombination repair (HRR) pathway plays a critical role in correcting the double-stranded breaks (DSB) due to DNA damage in human cells. Traditionally, the BRCA1/2 genes in the HRR pathway have been tested for their association with breast cancer. However, defects in the HRR pathway (HRD, also termed 'BRCAness'), which has up to 50 genes, have been shown to be involved in tumorigenesis and treatment susceptibility to poly-ADP ribose polymerase inhibitors (PARPis), platinum-based chemotherapy, and immune checkpoint inhibitors (ICIs). A reliable consensus on HRD scores is yet to be established. Emerging evidence suggests that only a subset of breast cancer patients benefit from ICI-based immunotherapy. Currently, albeit with limitations, the expression of programmed death-ligand 1 (PDL1) and tumor mutational burden (TMB) are utilized as biomarkers to predict the favorable outcomes of ICI therapy in breast cancer patients. Preclinical studies demonstrate an interplay between the HRR pathway and PDL1 expression. In this review, we outline the current understanding of the role of HRD in genomic instability leading to breast tumorigenesis and delineate outcomes from various clinical trials. Furthermore, we discuss potential strategies for combining HRD-targeted therapy with immunotherapy to achieve the best healthcare outcomes in breast cancer patients.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , BRCA1 Protein/genetics , Homologous Recombination/genetics , BRCA2 Protein/genetics , Genomic Instability , Genomics , Immunotherapy , Carcinogenesis
20.
Mol Cell ; 84(3): 409-410, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38307000

ABSTRACT

In this issue of Molecular Cell, Lim et al.1 reveal new insights into the distinct roles of BRCA2 in coping with DNA breaks, highlighting homologous recombination as the pivotal function that affects tumorigenesis and therapy response.


Subject(s)
DNA Replication , Rad51 Recombinase , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA Breaks , DNA Repair , Homologous Recombination/genetics , Rad51 Recombinase/genetics , Humans , Animals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL