Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nature ; 621(7978): 415-422, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37674080

ABSTRACT

DNA double-strand breaks (DSBs) are deleterious lesions that challenge genome integrity. To mitigate this threat, human cells rely on the activity of multiple DNA repair machineries that are tightly regulated throughout the cell cycle1. In interphase, DSBs are mainly repaired by non-homologous end joining and homologous recombination2. However, these pathways are completely inhibited in mitosis3-5, leaving the fate of mitotic DSBs unknown. Here we show that DNA polymerase theta6 (Polθ) repairs mitotic DSBs and thereby maintains genome integrity. In contrast to other DSB repair factors, Polθ function is activated in mitosis upon phosphorylation by Polo-like kinase 1 (PLK1). Phosphorylated Polθ is recruited by a direct interaction with the BRCA1 C-terminal domains of TOPBP1 to mitotic DSBs, where it mediates joining of broken DNA ends. Loss of Polθ leads to defective repair of mitotic DSBs, resulting in a loss of genome integrity. This is further exacerbated in cells that are deficient in homologous recombination, where loss of mitotic DSB repair by Polθ results in cell death. Our results identify mitotic DSB repair as the underlying cause of synthetic lethality between Polθ and homologous recombination. Together, our findings reveal the critical importance of mitotic DSB repair in the maintenance of genome integrity.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Directed DNA Polymerase , Mitosis , Protein Serine-Threonine Kinases , Humans , BRCA1 Protein/metabolism , Cell Cycle Proteins/metabolism , Cell Death/genetics , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Synthetic Lethal Mutations , DNA Polymerase theta , Polo-Like Kinase 1
2.
EMBO J ; 42(20): e110844, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37661798

ABSTRACT

Homologous recombination (HR) is a prominent DNA repair pathway maintaining genome integrity. Mutations in many HR genes lead to cancer predisposition. Paradoxically, the implication of the pivotal HR factor RAD51 on cancer development remains puzzling. Particularly, no RAD51 mouse models are available to address the role of RAD51 in aging and carcinogenesis in vivo. We engineered a mouse model with an inducible dominant-negative form of RAD51 (SMRad51) that suppresses RAD51-mediated HR without stimulating alternative mutagenic repair pathways. We found that in vivo expression of SMRad51 led to replicative stress, systemic inflammation, progenitor exhaustion, premature aging and reduced lifespan, but did not trigger tumorigenesis. Expressing SMRAD51 in a breast cancer predisposition mouse model (PyMT) decreased the number and the size of tumors, revealing an anti-tumor activity of SMRAD51. We propose that these in vivo phenotypes result from chronic endogenous replication stress caused by HR decrease, which preferentially targets progenitors and tumor cells. Our work underlines the importance of RAD51 activity for progenitor cell homeostasis, preventing aging and more generally for the balance between cancer and aging.


Subject(s)
Neoplasms , Rad51 Recombinase , Animals , Mice , Aging/genetics , Carcinogenesis/genetics , Cell Transformation, Neoplastic , DNA Damage , DNA Repair , Homologous Recombination , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
4.
Mol Cell ; 61(1): 15-26, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26687679

ABSTRACT

The end joining of distant DNA double-strand ends (DSEs) can produce potentially deleterious rearrangements. We show that depletion of cohesion complex proteins specifically stimulates the end joining (both C-NHEJ and A-EJ) of distant, but not close, I-SceI-induced DSEs in S/G2 phases. At the genome level, whole-exome sequencing showed that ablation of RAD21 or Sororin produces large chromosomal rearrangements (translocation, duplication, deletion). Moreover, cytogenetic analysis showed that RAD21 silencing leads to the formation of chromosome fusions synergistically with replication stress, which generates distant single-ended DSEs. These data reveal a role for the cohesin complex in protecting against genome rearrangements arising from the ligation of distant DSEs in S/G2 phases (both long-range DSEs and those that are only a few kilobases apart), while keeping end joining fully active for close DSEs. Therefore, this role likely involves limitation of DSE motility specifically in S phase, rather than inhibition of the end-joining machinery itself.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA Replication , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosome Aberrations , DNA-Binding Proteins , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , G2 Phase Cell Cycle Checkpoints , Gene Rearrangement , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Interference , S Phase Cell Cycle Checkpoints , Time Factors , Transfection , Cohesins
5.
Nucleic Acids Res ; 50(5): 2651-2666, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35137208

ABSTRACT

Selection of the appropriate DNA double-strand break (DSB) repair pathway is decisive for genetic stability. It is proposed to act according to two steps: 1-canonical nonhomologous end-joining (C-NHEJ) versus resection that generates single-stranded DNA (ssDNA) stretches; 2-on ssDNA, gene conversion (GC) versus nonconservative single-strand annealing (SSA) or alternative end-joining (A-EJ). Here, we addressed the mechanisms by which RAD51 regulates this second step, preventing nonconservative repair in human cells. Silencing RAD51 or BRCA2 stimulated both SSA and A-EJ, but not C-NHEJ, validating the two-step model. Three different RAD51 dominant-negative forms (DN-RAD51s) repressed GC and stimulated SSA/A-EJ. However, a fourth DN-RAD51 repressed SSA/A-EJ, although it efficiently represses GC. In living cells, the three DN-RAD51s that stimulate SSA/A-EJ failed to load efficiently onto damaged chromatin and inhibited the binding of endogenous RAD51, while the fourth DN-RAD51, which inhibits SSA/A-EJ, efficiently loads on damaged chromatin. Therefore, the binding of RAD51 to DNA, rather than its ability to promote GC, is required for SSA/A-EJ inhibition by RAD51. We showed that RAD51 did not limit resection of endonuclease-induced DSBs, but prevented spontaneous and RAD52-induced annealing of complementary ssDNA in vitro. Therefore, RAD51 controls the selection of the DSB repair pathway, protecting genome integrity from nonconservative DSB repair through ssDNA occupancy, independently of the promotion of CG.


Subject(s)
DNA Breaks, Double-Stranded , Rad51 Recombinase , Chromatin , DNA End-Joining Repair , DNA Repair , DNA, Single-Stranded/genetics , Humans , Rad51 Recombinase/metabolism
6.
Nucleic Acids Res ; 49(20): 11728-11745, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718776

ABSTRACT

Canonical non-homologous end-joining (cNHEJ) is the prominent mammalian DNA double-strand breaks (DSBs) repair pathway operative throughout the cell cycle. Phosphorylation of Ku70 at ser27-ser33 (pKu70) is induced by DNA DSBs and has been shown to regulate cNHEJ activity, but the underlying mechanism remained unknown. Here, we established that following DNA damage induction, Ku70 moves from nucleoli to the sites of damage, and once linked to DNA, it is phosphorylated. Notably, the novel emanating functions of pKu70 are evidenced through the recruitment of RNA Pol II and concomitant formation of phospho-53BP1 foci. Phosphorylation is also a prerequisite for the dynamic release of Ku70 from the repair complex through neddylation-dependent ubiquitylation. Although the non-phosphorylable ala-Ku70 form does not compromise the formation of the NHEJ core complex per se, cells expressing this form displayed constitutive and stress-inducible chromosomal instability. Consistently, upon targeted induction of DSBs by the I-SceI meganuclease into an intrachromosomal reporter substrate, cells expressing pKu70, rather than ala-Ku70, are protected against the joining of distal DNA ends. Collectively, our results underpin the essential role of pKu70 in the orchestration of DNA repair execution in living cells and substantiated the way it paves the maintenance of genome stability.


Subject(s)
DNA End-Joining Repair , Ku Autoantigen/metabolism , Cell Line , Cell Line, Tumor , DNA Damage , Humans , Phosphorylation , Protein Binding , RNA Polymerase II/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
7.
Nucleic Acids Res ; 45(21): 12325-12339, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29036662

ABSTRACT

Double strand breaks (DSBs) are one of the most toxic lesions to cells. DSB repair by the canonical non-homologous end-joining (C-EJ) pathway involves minor, if any, processing of the broken DNA-ends, whereas the initiation of DNA resection channels the broken-ends toward DNA repair pathways using various lengths of homology. Mechanisms that control the resection initiation are thus central to the regulation to the choice of DSB repair pathway. Therefore, understanding the mechanisms which regulate the initiation of DNA end-resection is of prime importance. Our findings reveal that poly(ADP-ribose) polymerase 2 (PARP2) is involved in DSBR pathway choice independently of its PAR synthesis activity. We show that PARP2 favors repair by homologous recombination (HR), single strand annealing (SSA) and alternative-end joining (A-EJ) rather than the C-EJ pathway and increases the deletion sizes at A-EJ junctions. We demonstrate that PARP2 specifically limits the accumulation of the resection barrier factor 53BP1 at DNA damage sites, allowing efficient CtIP-dependent DNA end-resection. Collectively, we have identified a new PARP2 function, independent of its PAR synthesis activity, which directs DSBs toward resection-dependent repair pathways.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Poly(ADP-ribose) Polymerases/physiology , Tumor Suppressor p53-Binding Protein 1/metabolism , BRCA1 Protein/metabolism , Carrier Proteins/metabolism , Cell Line , DNA End-Joining Repair , Endodeoxyribonucleases , Humans , Nuclear Proteins/metabolism , Recombinational DNA Repair
8.
PLoS Genet ; 12(10): e1006230, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27798638

ABSTRACT

DNA double-strand breaks (DSB) are very harmful lesions that can generate genome rearrangements. In this study, we used intrachromosomal reporters to compare both the efficiency and accuracy of end-joining occurring with close (34 bp apart) vs. distant DSBs (3200 bp apart) in human fibroblasts. We showed that a few kb between two intrachromosomal I-SceI-induced DSBs are sufficient to foster deletions and capture/insertions at the junction scar. Captured sequences are mostly coupled to deletions and can be partial duplications of the reporter (i.e., sequences adjacent to the DSB) or insertions of ectopic chromosomal sequences (ECS). Interestingly, silencing 53BP1 stimulates capture/insertions with distant but not with close double-strand ends (DSEs), although deletions were stimulated in both case. This shows that 53BP1 protects both close and distant DSEs from degradation and that the association of unprotection with distance between DSEs favors ECS capture. Reciprocally, silencing CtIP lessens ECS capture both in control and 53BP1-depleted cells. We propose that close ends are immediately/rapidly tethered and ligated, whereas distant ends first require synapsis of the distant DSEs prior to ligation. This "spatio-temporal" gap gives time and space for CtIP to initiate DNA resection, suggesting an involvement of single-stranded DNA tails for ECS capture. We therefore speculate that the resulting single-stranded DNA copies ECS through microhomology-mediated template switching.


Subject(s)
Carrier Proteins/genetics , DNA Breaks, Double-Stranded , Nuclear Proteins/genetics , Recombination, Genetic , Tumor Suppressor p53-Binding Protein 1/genetics , Chromosome Pairing/genetics , DNA End-Joining Repair/genetics , DNA, Single-Stranded/genetics , Endodeoxyribonucleases , Fibroblasts , Gene Silencing , Genome, Human , Humans
9.
Nucleic Acids Res ; 44(4): 1657-68, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26578561

ABSTRACT

Repair of DNA double-strand breaks occurs in a chromatin context that needs to be modified and remodeled to allow suitable access to the different DNA repair machineries. Of particular importance for the maintenance of genetic stability is the tight control of error-prone pathways, such as the alternative End Joining pathway. Here, we show that the chromatin remodeler p400 ATPase is a brake to the use of alternative End Joining. Using specific intracellular reporter susbstrates we observed that p400 depletion increases the frequency of alternative End Joining events, and generates large deletions following repair of double-strand breaks. This increase of alternative End Joining events is largely dependent on CtIP-mediated resection, indicating that it is probably related to the role of p400 in late steps of homologous recombination. Moreover, p400 depletion leads to the recruitment of poly(ADP) ribose polymerase (PARP) and DNA ligase 3 at DNA double-strand breaks, driving to selective killing by PARP inhibitors. All together these results show that p400 acts as a brake to prevent alternative End Joining-dependent genetic instability and underline its potential value as a clinical marker.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Chromatin/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , Humans , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage
10.
Nucleic Acids Res ; 42(9): 5616-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24598253

ABSTRACT

The repair of toxic double-strand breaks (DSB) is critical for the maintenance of genome integrity. The major mechanisms that cope with DSB are: homologous recombination (HR) and classical or alternative nonhomologous end joining (C-NHEJ versus A-EJ). Because these pathways compete for the repair of DSB, the choice of the appropriate repair pathway is pivotal. Among the mechanisms that influence this choice, deoxyribonucleic acid (DNA) end resection plays a critical role by driving cells to HR, while accurate C-NHEJ is suppressed. Furthermore, end resection promotes error-prone A-EJ. Increasing evidence define Poly(ADP-ribose) polymerase 3 (PARP3, also known as ARTD3) as an important player in cellular response to DSB. In this work, we reveal a specific feature of PARP3 that together with Ku80 limits DNA end resection and thereby helps in making the choice between HR and NHEJ pathways. PARP3 interacts with and PARylates Ku70/Ku80. The depletion of PARP3 impairs the recruitment of YFP-Ku80 to laser-induced DNA damage sites and induces an imbalance between BRCA1 and 53BP1. Both events result in compromised accurate C-NHEJ and a concomitant increase in DNA end resection. Nevertheless, HR is significantly reduced upon PARP3 silencing while the enhanced end resection causes mutagenic deletions during A-EJ. As a result, the absence of PARP3 confers hypersensitivity to anti-tumoral drugs generating DSB.


Subject(s)
Cell Cycle Proteins/physiology , DNA End-Joining Repair , Poly(ADP-ribose) Polymerases/physiology , Recombinational DNA Repair , Antigens, Nuclear/metabolism , Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Protein Transport , Replication Protein A/metabolism , Tumor Suppressor p53-Binding Protein 1
11.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38081641

ABSTRACT

Homologous recombination (HR) is a DNA repair mechanism of double-strand breaks and blocked replication forks, involving a process of homology search leading to the formation of synaptic intermediates that are regulated to ensure genome integrity. RAD51 recombinase plays a central role in this mechanism, supported by its RAD52 and BRCA2 partners. If the mediator function of BRCA2 to load RAD51 on RPA-ssDNA is well established, the role of RAD52 in HR is still far from understood. We used transmission electron microscopy combined with biochemistry to characterize the sequential participation of RPA, RAD52, and BRCA2 in the assembly of the RAD51 filament and its activity. Although our results confirm that RAD52 lacks a mediator activity, RAD52 can tightly bind to RPA-coated ssDNA, inhibit the mediator activity of BRCA2, and form shorter RAD51-RAD52 mixed filaments that are more efficient in the formation of synaptic complexes and D-loops, resulting in more frequent multi-invasions as well. We confirm the in situ interaction between RAD51 and RAD52 after double-strand break induction in vivo. This study provides new molecular insights into the formation and regulation of presynaptic and synaptic intermediates by BRCA2 and RAD52 during human HR.


Subject(s)
Rad51 Recombinase , Replication Protein A , Humans , Replication Protein A/genetics , Replication Protein A/metabolism , Rad51 Recombinase/genetics , DNA, Single-Stranded/genetics , DNA Repair/genetics , Homologous Recombination/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism
12.
Cells ; 12(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-37190078

ABSTRACT

Homologous recombination (HR), an evolutionary conserved pathway, plays a paramount role(s) in genome plasticity. The pivotal HR step is the strand invasion/exchange of double-stranded DNA by a homologous single-stranded DNA (ssDNA) covered by RAD51. Thus, RAD51 plays a prime role in HR through this canonical catalytic strand invasion/exchange activity. The mutations in many HR genes cause oncogenesis. Surprisingly, despite its central role in HR, the invalidation of RAD51 is not classified as being cancer prone, constituting the "RAD51 paradox". This suggests that RAD51 exercises other noncanonical roles that are independent of its catalytic strand invasion/exchange function. For example, the binding of RAD51 on ssDNA prevents nonconservative mutagenic DNA repair, which is independent of its strand exchange activity but relies on its ssDNA occupancy. At the arrested replication forks, RAD51 plays several noncanonical roles in the formation, protection, and management of fork reversal, allowing for the resumption of replication. RAD51 also exhibits noncanonical roles in RNA-mediated processes. Finally, RAD51 pathogenic variants have been described in the congenital mirror movement syndrome, revealing an unexpected role in brain development. In this review, we present and discuss the different noncanonical roles of RAD51, whose presence does not automatically result in an HR event, revealing the multiple faces of this prominent actor in genomic plasticity.


Subject(s)
DNA Repair , Rad51 Recombinase , DNA/metabolism , DNA Replication , DNA, Single-Stranded , DNA-Binding Proteins/metabolism , Rad51 Recombinase/genetics , Humans , Animals
13.
Cell Death Differ ; 30(5): 1349-1365, 2023 05.
Article in English | MEDLINE | ID: mdl-36869180

ABSTRACT

Cells are inevitably challenged by low-level/endogenous stresses that do not arrest DNA replication. Here, in human primary cells, we discovered and characterized a noncanonical cellular response that is specific to nonblocking replication stress. Although this response generates reactive oxygen species (ROS), it induces a program that prevents the accumulation of premutagenic 8-oxoguanine in an adaptive way. Indeed, replication stress-induced ROS (RIR) activate FOXO1-controlled detoxification genes such as SEPP1, catalase, GPX1, and SOD2. Primary cells tightly control the production of RIR: They are excluded from the nucleus and are produced by the cellular NADPH oxidases DUOX1/DUOX2, whose expression is controlled by NF-κB, which is activated by PARP1 upon replication stress. In parallel, inflammatory cytokine gene expression is induced through the NF-κB-PARP1 axis upon nonblocking replication stress. Increasing replication stress intensity accumulates DNA double-strand breaks and triggers the suppression of RIR by p53 and ATM. These data underline the fine-tuning of the cellular response to stress that protects genome stability maintenance, showing that primary cells adapt their responses to replication stress severity.


Subject(s)
NADPH Oxidases , NF-kappa B , Humans , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Cytokines/genetics , Genomic Instability
14.
Cancer Gene Ther ; 30(8): 1144-1155, 2023 08.
Article in English | MEDLINE | ID: mdl-37248434

ABSTRACT

The tumor suppressor gene WWOX is localized in an unstable chromosomal region and its expression is decreased or absent in several types of cancer. A low expression of WWOX is associated with a poor prognosis in breast cancer (BC). It has recently been shown that WWOX contributes to genome stability through its role in the DNA damage response (DDR). In breast cancer cells, WWOX inhibits homologous recombination (HR), and thus promotes the repair of DNA double-stranded breaks (DSBs) by non-homologous end joining (NHEJ). The fine-tuning modulation of HR activity is crucial. Its under or overstimulation inducing genome alterations that can induce cancer. MERIT40 is a positive regulator of the DDR. This protein is indispensable for the function of the multi-protein complex BRCA1-A, which suppresses excessive HR activity. MERIT40 also recruits Tankyrase, a positive regulator of HR, to the DSBs to stimulate DNA repair. Here, we identified MERIT40 as a new molecular partner of WWOX. We demonstrated that WWOX inhibited excessive HR activity induced by overexpression of MERIT40. We showed that WWOX impaired the MERIT40-Tankyrase interaction preventing the role of the complex on DSBs. Furthermore, we found that MERIT40 is overexpressed in BC and that this overexpression is associated to a poor prognosis. These results strongly suggest that WWOX, through its interaction with MERIT40, prevents the deleterious impact of excessive HR on BC development by inhibiting MERIT40-Tankyrase association. This inhibitory effect of WWOX would oppose MERIT40-dependent BC development.


Subject(s)
Breast Neoplasms , Homologous Recombination , Female , Humans , Breast Neoplasms/genetics , DNA Breaks, Double-Stranded , DNA Repair , Tankyrases/genetics , Tankyrases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase/genetics , WW Domain-Containing Oxidoreductase/metabolism
15.
EMBO Rep ; 10(8): 887-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19557000

ABSTRACT

Ataxia telangiectasia mutated (ATM), the deficiency of which causes a severe neurodegenerative disease, is a crucial mediator for the DNA damage response (DDR). As neurons have high rates of transcription that require topoisomerase I (TOP1), we investigated whether TOP1 cleavage complexes (TOP1cc)-which are potent transcription-blocking lesions-also produce transcription-dependent DNA double-strand breaks (DSBs) with ATM activation. We show the induction of DSBs and DDR activation in post-mitotic primary neurons and lymphocytes treated with camptothecin, with the induction of nuclear DDR foci containing activated ATM, gamma-H2AX (phosphorylated histone H2AX), activated CHK2 (checkpoint kinase 2), MDC1 (mediator of DNA damage checkpoint 1) and 53BP1 (p53 binding protein 1). The DSB-ATM-DDR pathway was suppressed by inhibiting transcription and gamma-H2AX signals were reduced by RNase H1 transfection, which removes transcription-mediated R-loops. Thus, we propose that Top1cc produce transcription arrests with R-loop formation and generate DSBs that activate ATM in post-mitotic cells.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alpha-Amanitin/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Camptothecin/pharmacology , Cells, Cultured , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Flow Cytometry , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Rats , Ribonuclease H/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Tumor Suppressor p53-Binding Protein 1
16.
NAR Cancer ; 3(2): zcab016, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34316706

ABSTRACT

Genetic instability is a hallmark of cancer cells. Homologous recombination (HR) plays key roles in genome stability and variability due to its roles in DNA double-strand break and interstrand crosslink repair, and in the protection and resumption of arrested replication forks. HR deficiency leads to genetic instability, and, as expected, many HR genes are downregulated in cancer cells. The link between HR deficiency and cancer predisposition is exemplified by familial breast and ovarian cancers and by some subgroups of Fanconi anaemia syndromes. Surprisingly, although RAD51 plays a pivotal role in HR, i.e., homology search and in strand exchange with a homologous DNA partner, almost no inactivating mutations of RAD51 have been associated with cancer predisposition; on the contrary, overexpression of RAD51 is associated with a poor prognosis in different types of tumours. Taken together, these data highlight the fact that RAD51 differs from its HR partners with regard to cancer susceptibility and expose what we call the 'RAD51 paradox'. Here, we catalogue the dysregulations of HR genes in human pathologies, including cancer and Fanconi anaemia or congenital mirror movement syndromes, and we discuss the RAD51 paradox.

17.
Sci Transl Med ; 13(587)2021 03 31.
Article in English | MEDLINE | ID: mdl-33790022

ABSTRACT

The development and survival of cancer cells require adaptive mechanisms to stress. Such adaptations can confer intrinsic vulnerabilities, enabling the selective targeting of cancer cells. Through a pooled in vivo short hairpin RNA (shRNA) screen, we identified the adenosine triphosphatase associated with diverse cellular activities (AAA-ATPase) valosin-containing protein (VCP) as a top stress-related vulnerability in acute myeloid leukemia (AML). We established that AML was the most responsive disease to chemical inhibition of VCP across a panel of 16 cancer types. The sensitivity to VCP inhibition of human AML cell lines, primary patient samples, and syngeneic and xenograft mouse models of AML was validated using VCP-directed shRNAs, overexpression of a dominant-negative VCP mutant, and chemical inhibition. By combining mass spectrometry-based analysis of the VCP interactome and phospho-signaling studies, we determined that VCP is important for ataxia telangiectasia mutated (ATM) kinase activation and subsequent DNA repair through homologous recombination in AML. A second-generation VCP inhibitor, CB-5339, was then developed and characterized. Efficacy and safety of CB-5339 were validated in multiple AML models, including syngeneic and patient-derived xenograft murine models. We further demonstrated that combining DNA-damaging agents, such as anthracyclines, with CB-5339 treatment synergizes to impair leukemic growth in an MLL-AF9-driven AML murine model. These studies support the clinical testing of CB-5339 as a single agent or in combination with standard-of-care DNA-damaging chemotherapy for the treatment of AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Adenosine Triphosphatases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Repair , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Valosin Containing Protein
18.
Proc Natl Acad Sci U S A ; 104(52): 20902-7, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18093953

ABSTRACT

XRCC4-null mice have a more severe phenotype than KU80-null mice. Here, we address whether this difference in phenotype is connected to nonhomologous end-joining (NHEJ). We used intrachromosomal substrates to monitor NHEJ of two distal double-strand breaks (DSBs) targeted by I-SceI, in living cells. In xrcc4-defective XR-1 cells, a residual but significant end-joining process exists, which primarily uses microhomologies distal from the DSB. However, NHEJ efficiency was strongly reduced in xrcc4-defective XR-1 cells versus complemented cells, contrasting with KU-deficient xrs6 cells, which showed levels of end-joining similar to those of complemented cells. Nevertheless, sequence analysis of the repair junctions indicated that the accuracy of end-joining was strongly affected in both xrcc4-deficient and KU-deficient cells. More specifically, these data showed that the KU80/XRCC4 pathway is conservative and not intrinsically error-prone but can accommodate non-fully complementary ends at the cost of limited mutagenesis.


Subject(s)
Antigens, Nuclear/genetics , Antigens, Nuclear/physiology , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Animals , CD4-Positive T-Lymphocytes/metabolism , CHO Cells , Cricetinae , Cricetulus , Flow Cytometry , Genetic Complementation Test , Ku Autoantigen , Mice , Mice, Transgenic , Models, Genetic , Phenotype
19.
Clin Cancer Res ; 14(20): 6449-55, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927284

ABSTRACT

PURPOSE: Ecteinascidin 743 (Et743; trabectedin, Yondelis) has recently been approved in Europe for the treatment of soft tissue sarcomas and is undergoing clinical trials for other solid tumors. Et743 selectively targets cells proficient for TC-NER, which sets it apart from other DNA alkylating agents. In the present study, we examined the effects of Et743 on RNA Pol II. EXPERIMENTAL DESIGN AND RESULTS: We report that Et743 induces the rapid and massive degradation of transcribing Pol II in various cancer cell lines and normal fibroblasts. Pol II degradation was abrogated by the proteasome inhibitor MG132 and was dependent on TC-NER. Cockayne syndrome (CS) cells and xeroderma pigmentosum (XP) cells (XPD, XPA, XPG, and XPF) were defective in Pol II degradation, whereas XPC cells whose defect is limited to global genome NER in nontranscribing regions were proficient for Pol II degradation. Complementation of the CSB and XPD cells restored Pol II degradation. We also show that cells defective for the VHL complex were defective in Pol II degradation and that complementation of those cells restores Pol II degradation. Moreover, VHL deficiency rendered cells resistant to Et743-induced cell death, a similar effect to that of TC-NER deficiency. CONCLUSION: These results suggest that both TC-NER-induced and VHL-mediated Pol II degradation play a role in cell killing by Et743.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair/drug effects , Dioxoles/pharmacology , Neoplasms/enzymology , RNA Polymerase II/metabolism , Tetrahydroisoquinolines/pharmacology , Transcription, Genetic/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cockayne Syndrome/enzymology , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Cysteine Proteinase Inhibitors/pharmacology , DNA Damage/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Genetic Complementation Test , Humans , Leupeptins/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Poly-ADP-Ribose Binding Proteins , RNA Polymerase II/genetics , Sarcoma/enzymology , Sarcoma/genetics , Sarcoma/pathology , Trabectedin , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism , von Hippel-Lindau Disease/enzymology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
20.
Mol Cancer Res ; 17(1): 54-69, 2019 01.
Article in English | MEDLINE | ID: mdl-30257991

ABSTRACT

BRCA1 mutations have been identified that increase the risk of developing hereditary breast and ovarian cancers. Genetic screening is now offered to patients with a family history of cancer, to adapt their treatment and the management of their relatives. However, a large number of BRCA1 variants of uncertain significance (VUS) are detected. To better understand the significance of these variants, a high-throughput structural and functional analysis was performed on a large set of BRCA1 VUS. Information on both cellular localization and homology-directed DNA repair (HR) capacity was obtained for 78 BRCT missense variants in the UMD-BRCA1 database and measurement of the structural stability and phosphopeptide-binding capacities was performed for 42 mutated BRCT domains. This extensive and systematic analysis revealed that most characterized causal variants affect BRCT-domain solubility in bacteria and all impair BRCA1 HR activity in cells. Furthermore, binding to a set of 5 different phosphopeptides was tested: all causal variants showed phosphopeptide-binding defects and no neutral variant showed such defects. A classification is presented on the basis of mutated BRCT domain solubility, phosphopeptide-binding properties, and VUS HR capacity. These data suggest that HR-defective variants, which present, in addition, BRCT domains either insoluble in bacteria or defective for phosphopeptide binding, lead to an increased cancer risk. Furthermore, the data suggest that variants with a WT HR activity and whose BRCT domains bind with a WT affinity to the 5 phosphopeptides are neutral. The case of variants with WT HR activity and defective phosphopeptide binding should be further characterized, as this last functional defect might be sufficient per se to lead to tumorigenesis. IMPLICATIONS: The analysis of the current study on BRCA1 structural and functional defects on cancer risk and classification presented may improve clinical interpretation and therapeutic selection.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Phosphopeptides/genetics , Phosphopeptides/metabolism , Animals , Breast Neoplasms/pathology , Female , Genetic Predisposition to Disease , Genetic Testing , Homologous Recombination , Humans , Mice , Models, Molecular , Mutation, Missense , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL