Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 127
1.
Nat Commun ; 15(1): 4430, 2024 May 24.
Article En | MEDLINE | ID: mdl-38789420

Histone H2AX plays a key role in DNA damage signalling in the surrounding regions of DNA double-strand breaks (DSBs). In response to DNA damage, H2AX becomes phosphorylated on serine residue 139 (known as γH2AX), resulting in the recruitment of the DNA repair effectors 53BP1 and BRCA1. Here, by studying resistance to poly(ADP-ribose) polymerase (PARP) inhibitors in BRCA1/2-deficient mammary tumours, we identify a function for γH2AX in orchestrating drug-induced replication fork degradation. Mechanistically, γH2AX-driven replication fork degradation is elicited by suppressing CtIP-mediated fork protection. As a result, H2AX loss restores replication fork stability and increases chemoresistance in BRCA1/2-deficient tumour cells without restoring homology-directed DNA repair, as highlighted by the lack of DNA damage-induced RAD51 foci. Furthermore, in the attempt to discover acquired genetic vulnerabilities, we find that ATM but not ATR inhibition overcomes PARP inhibitor (PARPi) resistance in H2AX-deficient tumours by interfering with CtIP-mediated fork protection. In summary, our results demonstrate a role for H2AX in replication fork biology in BRCA-deficient tumours and establish a function of H2AX separable from its classical role in DNA damage signalling and DSB repair.


BRCA1 Protein , BRCA2 Protein , DNA Replication , Drug Resistance, Neoplasm , Histones , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Female , Humans , Mice , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/deficiency , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , DNA Replication/drug effects , Drug Resistance, Neoplasm/genetics , Histones/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Mice, Nude
2.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38731844

More than 20% of metastatic prostate cancer carries genomic defects involving DNA damage repair pathways, mainly in homologous recombination repair-related genes. The recent approval of olaparib has paved the way to precision medicine for the treatment of metastatic prostate cancer with PARP inhibitors in this subset of patients, especially in the case of BRCA1 or BRCA2 pathogenic/likely pathogenic variants. In face of this new therapeutic opportunity, many issues remain unsolved. This narrative review aims to describe the relationship between homologous recombination repair deficiency and prostate cancer, the techniques used to determine homologous recombination repair status in prostate cancer, the crosstalk between homologous recombination repair and the androgen receptor pathway, the current evidence on PARP inhibitors activity in metastatic prostate cancer also in homologous recombination repair-proficient tumors, as well as emerging mechanisms of resistance to PARP inhibitors. The possibility of combination therapies including a PARP inhibitor is an attractive option, and more robust data are awaited from ongoing phase II and phase III trials outlined in this manuscript.


Poly(ADP-ribose) Polymerase Inhibitors , Prostatic Neoplasms , Recombinational DNA Repair , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , BRCA2 Protein/genetics , BRCA2 Protein/deficiency , Neoplasm Metastasis , BRCA1 Protein/genetics , BRCA1 Protein/deficiency , Phthalazines/therapeutic use , Phthalazines/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Piperazines
3.
Nature ; 621(7977): 129-137, 2023 Sep.
Article En | MEDLINE | ID: mdl-37587346

Homologous recombination (HR) deficiency is associated with DNA rearrangements and cytogenetic aberrations1. Paradoxically, the types of DNA rearrangements that are specifically associated with HR-deficient cancers only minimally affect chromosomal structure2. Here, to address this apparent contradiction, we combined genome-graph analysis of short-read whole-genome sequencing (WGS) profiles across thousands of tumours with deep linked-read WGS of 46 BRCA1- or BRCA2-mutant breast cancers. These data revealed a distinct class of HR-deficiency-enriched rearrangements called reciprocal pairs. Linked-read WGS showed that reciprocal pairs with identical rearrangement orientations gave rise to one of two distinct chromosomal outcomes, distinguishable only with long-molecule data. Whereas one (cis) outcome corresponded to the copying and pasting of a small segment to a distant site, a second (trans) outcome was a quasi-balanced translocation or multi-megabase inversion with substantial (10 kb) duplications at each junction. We propose an HR-independent replication-restart repair mechanism to explain the full spectrum of reciprocal pair outcomes. Linked-read WGS also identified single-strand annealing as a repair pathway that is specific to BRCA2 deficiency in human cancers. Integrating these features in a classifier improved discrimination between BRCA1- and BRCA2-deficient genomes. In conclusion, our data reveal classes of rearrangements that are specific to BRCA1 or BRCA2 deficiency as a source of cytogenetic aberrations in HR-deficient cells.


BRCA1 Protein , BRCA2 Protein , Chromosome Aberrations , DNA Repair , Neoplasms , Humans , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Chromosome Inversion , DNA Repair/genetics , Neoplasms/genetics , Translocation, Genetic/genetics , Homologous Recombination , Cytogenetic Analysis , Chromosome Aberrations/classification
4.
Nucleic Acids Res ; 49(22): 12836-12854, 2021 12 16.
Article En | MEDLINE | ID: mdl-34871413

Cockayne syndrome group B (CSB) protein has been implicated in the repair of a variety of DNA lesions that induce replication stress. However, little is known about its role at stalled replication forks. Here, we report that CSB is recruited to stalled forks in a manner dependent upon its T1031 phosphorylation by CDK. While dispensable for MRE11 association with stalled forks in wild-type cells, CSB is required for further accumulation of MRE11 at stalled forks in BRCA1/2-deficient cells. CSB promotes MRE11-mediated fork degradation in BRCA1/2-deficient cells. CSB possesses an intrinsic ATP-dependent fork reversal activity in vitro, which is activated upon removal of its N-terminal region that is known to autoinhibit CSB's ATPase domain. CSB functions similarly to fork reversal factors SMARCAL1, ZRANB3 and HLTF to regulate slowdown in fork progression upon exposure to replication stress, indicative of a role of CSB in fork reversal in vivo. Furthermore, CSB not only acts epistatically with MRE11 to facilitate fork restart but also promotes RAD52-mediated break-induced replication repair of double-strand breaks arising from cleavage of stalled forks by MUS81 in BRCA1/2-deficient cells. Loss of CSB exacerbates chemosensitivity in BRCA1/2-deficient cells, underscoring an important role of CSB in the treatment of cancer lacking functional BRCA1/2.


BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA Repair , DNA/genetics , MRE11 Homologue Protein/genetics , Poly-ADP-Ribose Binding Proteins/genetics , BRCA1 Protein/deficiency , BRCA1 Protein/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Cell Line , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Replication/genetics , HCT116 Cells , HEK293 Cells , Humans , MRE11 Homologue Protein/metabolism , Mutation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Interference
5.
Nucleic Acids Res ; 49(22): 12855-12869, 2021 12 16.
Article En | MEDLINE | ID: mdl-34871431

Understanding chemoresistance mechanisms in BRCA-deficient cells will allow for identification of biomarkers for predicting tumor response to therapy, as well as the design of novel therapeutic approaches targeting this chemoresistance. Here, we show that the protein MED12, a component of the Mediator transcription regulation complex, plays an unexpected role in regulating chemosensitivity in BRCA-deficient cells. We found that loss of MED12 confers resistance to cisplatin and PARP inhibitors in both BRCA1- and BRCA2-deficient cells, which is associated with restoration of both homologous recombination and replication fork stability. Surprisingly, MED12-controlled chemosensitivity does not involve a function of the Mediator complex, but instead reflects a distinct role of MED12 in suppression of the TGFß pathway. Importantly, we show that ectopic activation of the TGFß pathway is enough to overcome the fork protection and DNA repair defects of BRCA-mutant cells, resulting in chemoresistance. Our work identifies the MED12-TGFß module as an important regulator of genomic stability and chemosensitivity in BRCA-deficient cells.


BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Replication/genetics , Drug Resistance, Neoplasm/genetics , Mediator Complex/genetics , Transforming Growth Factor beta/genetics , Antineoplastic Agents/pharmacology , BRCA1 Protein/deficiency , BRCA1 Protein/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/pharmacology , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Repair , HeLa Cells , Humans , Mediator Complex/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , RNA Interference , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism
6.
Commun Biol ; 4(1): 1270, 2021 11 08.
Article En | MEDLINE | ID: mdl-34750509

PARP enzymes utilise NAD+ as a co-substrate for their enzymatic activity. Inhibition of PARP1 is synthetic lethal with defects in either BRCA1 or BRCA2. In order to assess whether other genes implicated in NAD+ metabolism were synthetic lethal with BRCA1 or BRCA2 gene defects, we carried out a genetic screen, which identified a synthetic lethality between BRCA1 and genetic inhibition of either of two sirtuin (SIRT) enzymes, SIRT1 or SIRT6. This synthetic lethal interaction was replicated using small-molecule SIRT inhibitors and was associated with replication stress and increased cellular PARylation, in contrast to the decreased PARylation associated with BRCA-gene/PARP inhibitor synthetic lethality. SIRT/BRCA1 synthetic lethality was reversed by genetic ablation of either PARP1 or the histone PARylation factor-coding gene HPF1, implicating PARP1/HPF1-mediated serine ADP-ribosylation as part of the mechanistic basis of this synthetic lethal effect. These observations suggest that PARP1/HPF1-mediated serine ADP-ribosylation, when driven by SIRT inhibition, can inadvertently inhibit the growth of BRCA-gene mutant cells.


BRCA1 Protein/genetics , BRCA2 Protein/genetics , Sirtuins/metabolism , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism
7.
Nat Commun ; 12(1): 4919, 2021 08 13.
Article En | MEDLINE | ID: mdl-34389725

BRCA1 or BRCA2 germline mutations predispose to breast, ovarian and other cancers. High-throughput sequencing of tumour genomes revealed that oncogene amplification and BRCA1/2 mutations are mutually exclusive in cancer, however the molecular mechanism underlying this incompatibility remains unknown. Here, we report that activation of ß-catenin, an oncogene of the WNT signalling pathway, inhibits proliferation of BRCA1/2-deficient cells. RNA-seq analyses revealed ß-catenin-induced discrete transcriptome alterations in BRCA2-deficient cells, including suppression of CDKN1A gene encoding the CDK inhibitor p21. This accelerates G1/S transition, triggering illegitimate origin firing and DNA damage. In addition, ß-catenin activation accelerates replication fork progression in BRCA2-deficient cells, which is critically dependent on p21 downregulation. Importantly, we find that upregulated p21 expression is essential for the survival of BRCA2-deficient cells and tumours. Thus, our work demonstrates that ß-catenin toxicity in cancer cells with compromised BRCA1/2 function is driven by transcriptional alterations that cause aberrant replication and inflict DNA damage.


BRCA1 Protein/genetics , BRCA2 Protein/genetics , Oncogenes/genetics , Transcription, Genetic/genetics , beta Catenin/genetics , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Female , Gene Expression Profiling/methods , HeLa Cells , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA-Seq/methods , beta Catenin/metabolism
8.
Cell Rep ; 36(5): 109478, 2021 08 03.
Article En | MEDLINE | ID: mdl-34348152

Oxidative stress is a ubiquitous cellular challenge implicated in aging, neurodegeneration, and cancer. By studying pathogenic mutations in the tumor suppressor BRCA2, we identify a general mechanism by which oxidative stress restricts mitochondrial (mt)DNA replication. BRCA2 inactivation induces R-loop accumulation in the mtDNA regulatory region and diminishes mtDNA replication initiation. In BRCA2-deficient cells, intracellular reactive oxygen species (ROS) are elevated, and ROS scavengers suppress the mtDNA defects. Conversely, wild-type cells exposed to oxidative stress by pharmacologic or genetic manipulation phenocopy these defects. Mechanistically, we find that 8-oxoguanine accumulation in mtDNA caused by oxidative stress suffices to impair recruitment of the mitochondrial enzyme RNaseH1 to sites of R-loop accrual, restricting mtDNA replication initiation. Thus, oxidative stress impairs RNaseH1 function to cripple mtDNA maintenance. Our findings highlight a molecular mechanism that links oxidative stress to mitochondrial dysfunction and is elicited by the inactivation of genes implicated in neurodegeneration and cancer.


BRCA2 Protein/deficiency , DNA, Mitochondrial/genetics , Oxidative Stress , Ribonuclease H/metabolism , BRCA2 Protein/metabolism , DNA Glycosylases/metabolism , DNA Helicases/metabolism , DNA Replication , DNA, Mitochondrial/chemistry , Female , Guanine/analogs & derivatives , Guanine/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondria/pathology , Multifunctional Enzymes/metabolism , R-Loop Structures , RNA Helicases/metabolism , RNA-Binding Proteins/metabolism
9.
Mol Cell ; 81(11): 2278-2289, 2021 06 03.
Article En | MEDLINE | ID: mdl-33984284

Agents that induce DNA damage can cure some cancers. However, the side effects of chemotherapy are severe because of the indiscriminate action of DNA-damaging agents on both healthy and cancerous cells. DNA repair pathway inhibition provides a less toxic and targeted alternative to chemotherapy. A compelling DNA repair target is the Fanconi anemia (FA) E3 ligase core complex due to its critical-and likely singular-role in the efficient removal of specific DNA lesions. FA pathway inactivation has been demonstrated to specifically kill some types of cancer cells without the addition of exogenous DNA damage, including cells that lack BRCA1, BRCA2, ATM, or functionally related genes. In this perspective, we discuss the genetic and biochemical evidence in support of the FA core complex as a compelling drug target for cancer therapy. In particular, we discuss the genetic, biochemical, and structural data that could rapidly advance our capacity to identify and implement the use of FA core complex inhibitors in the clinic.


Ataxia Telangiectasia Mutated Proteins/genetics , BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Repair/drug effects , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia/drug therapy , Ubiquitin-Protein Ligases/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/deficiency , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA Damage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group Proteins/antagonists & inhibitors , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy/methods , Morpholines/therapeutic use , Pyrones/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Synthetic Lethal Mutations , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/antagonists & inhibitors , Ubiquitins/genetics , Ubiquitins/metabolism
10.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article En | MEDLINE | ID: mdl-33922657

BRCA mutation, one of the most common types of mutations in breast and ovarian cancer, has been suggested to be synthetically lethal with depletion of RAD52. Pharmacologically inhibiting RAD52 specifically eradicates BRCA-deficient cancer cells. In this study, we demonstrated that curcumin, a plant polyphenol, sensitizes BRCA2-deficient cells to CPT-11 by impairing RAD52 recombinase in MCF7 cells. More specifically, in MCF7-siBRCA2 cells, curcumin reduced homologous recombination, resulting in tumor growth suppression. Furthermore, a BRCA2-deficient cell line, Capan1, became resistant to CPT-11 when BRCA2 was reintroduced. In vivo, xenograft model studies showed that curcumin combined with CPT-11 reduced the growth of BRCA2-knockout MCF7 tumors but not MCF7 tumors. In conclusion, our data indicate that curcumin, which has RAD52 inhibitor activity, is a promising candidate for sensitizing BRCA2-deficient cells to DNA damage-based cancer therapies.


BRCA2 Protein/deficiency , Breast Neoplasms/drug therapy , Curcumin/pharmacology , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , DNA Repair , Female , Humans , Irinotecan/pharmacology , Mice , Mice, Nude , Mutation , Topoisomerase I Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Dev Cell ; 56(4): 461-477.e7, 2021 02 22.
Article En | MEDLINE | ID: mdl-33621493

Homology-directed repair (HDR) safeguards DNA integrity under various forms of stress, but how HDR protects replicating genomes under extensive metabolic alterations remains unclear. Here, we report that besides stalling replication forks, inhibition of ribonucleotide reductase (RNR) triggers metabolic imbalance manifested by the accumulation of increased reactive oxygen species (ROS) in cell nuclei. This leads to a redox-sensitive activation of the ATM kinase followed by phosphorylation of the MRE11 nuclease, which in HDR-deficient settings degrades stalled replication forks. Intriguingly, nascent DNA degradation by the ROS-ATM-MRE11 cascade is also triggered by hypoxia, which elevates signaling-competent ROS and attenuates functional HDR without arresting replication forks. Under these conditions, MRE11 degrades daughter-strand DNA gaps, which accumulate behind active replisomes and attract error-prone DNA polymerases to escalate mutation rates. Thus, HDR safeguards replicating genomes against metabolic assaults by restraining mutagenic repair at aberrantly processed nascent DNA. These findings have implications for cancer evolution and tumor therapy.


DNA Replication , Genome, Human , Metabolism , Recombinational DNA Repair , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Cell Hypoxia , Cell Line, Tumor , DNA/metabolism , Humans , MRE11 Homologue Protein/metabolism , Models, Biological , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Polymerization , Reactive Oxygen Species/metabolism , Signal Transduction
12.
J Clin Invest ; 131(3)2021 02 01.
Article En | MEDLINE | ID: mdl-33529165

Germline mutations in BRCA1 and BRCA2 (BRCA1/2) genes considerably increase breast and ovarian cancer risk. Given that tumors with these mutations have elevated genomic instability, they exhibit relative vulnerability to certain chemotherapies and targeted treatments based on poly (ADP-ribose) polymerase (PARP) inhibition. However, the molecular mechanisms that influence cancer risk and therapeutic benefit or resistance remain only partially understood. BRCA1 and BRCA2 have also been implicated in the suppression of R-loops, triple-stranded nucleic acid structures composed of a DNA:RNA hybrid and a displaced ssDNA strand. Here, we report that loss of RNF168, an E3 ubiquitin ligase and DNA double-strand break (DSB) responder, remarkably protected Brca1-mutant mice against mammary tumorigenesis. We demonstrate that RNF168 deficiency resulted in accumulation of R-loops in BRCA1/2-mutant breast and ovarian cancer cells, leading to DSBs, senescence, and subsequent cell death. Using interactome assays, we identified RNF168 interaction with DHX9, a helicase involved in the resolution and removal of R-loops. Mechanistically, RNF168 directly ubiquitylated DHX9 to facilitate its recruitment to R-loop-prone genomic loci. Consequently, loss of RNF168 impaired DHX9 recruitment to R-loops, thereby abrogating its ability to resolve R-loops. The data presented in this study highlight a dependence of BRCA1/2-defective tumors on factors that suppress R-loops and reveal a fundamental RNF168-mediated molecular mechanism that governs cancer development and vulnerability.


BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA, Neoplasm/metabolism , Genomic Instability , Mammary Neoplasms, Animal/metabolism , Ovarian Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA, Neoplasm/genetics , Female , Genetic Loci , Humans , Mammary Neoplasms, Animal/genetics , Mice , Mice, Knockout , Ovarian Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
13.
Cells ; 9(9)2020 09 16.
Article En | MEDLINE | ID: mdl-32948057

Personalized medicine in treating pancreatic ductal adenocarcinoma (PDAC) is still in its infancy, albeit PDAC-related deaths are projected to rise over the next decade. Only recently, maintenance therapy with the PARP inhibitor olaparib showed improved progression-free survival in germline BRCA1/2-mutated PDAC patients after platinum-based induction for the first time. Transferability of such a concept to other DNA damage response (DDR) genes remains unclear. Here, we conducted a placebo-controlled, three-armed preclinical trial to evaluate the efficacy of multi-DDR interference (mDDRi) as maintenance therapy vs. continuous FOLFIRINOX treatment, implemented with orthotopically transplanted ATM-deficient PDAC cell lines. Kaplan-Meier analysis, cross-sectional imaging, histology, and in vitro analysis served as analytical readouts. Median overall survival was significantly longer in the mDDRi maintenance arm compared to the maintained FOLFIRINOX treatment. This survival benefit was mirrored in the highest DNA-damage load, accompanied by superior disease control and reduced metastatic burden. In vitro analysis suggests FOLFIRINOX-driven selection of invasive subclones, erased by subsequent mDDRi treatment. Collectively, this preclinical trial substantiates mDDRi in a maintenance setting as a novel therapeutic option and extends the concept to non-germline BRCA1/2-mutant PDAC.


Ataxia Telangiectasia Mutated Proteins/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Maintenance Chemotherapy/methods , Pancreatic Neoplasms/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ataxia Telangiectasia Mutated Proteins/deficiency , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Chromosomal Instability , DNA Damage , Female , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Irinotecan/therapeutic use , Leucovorin/therapeutic use , Mice , Mice, Nude , Middle Aged , Oxaliplatin/therapeutic use , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Poly(ADP-ribose) Polymerases , Survival Analysis , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
14.
Cell Death Dis ; 11(9): 812, 2020 09 26.
Article En | MEDLINE | ID: mdl-32980867

BRCA2 is crucial for repairing DNA double-strand breaks with high fidelity, and loss of BRCA2 increases the risks of developing breast and ovarian cancers. Herein, we show that BRCA2 is inactively mutated in 10% of gastric and 7% of colorectal adenocarcinomas, and that this inactivation is significantly correlated with microsatellite instability. Villin-driven Brca2 depletion promotes mouse gastrointestinal tumor formation when genome instability is increased. Whole-genome screening data showed that these BRCA2 monoallelic and biallelic mutant tumors were selectively inhibited by mitomycin C. Mechanistically, mitomycin C provoked double-strand breaks in cancer cells that often recruit wild-type BRCA2 for repair; the failure to repair double-strand breaks caused cell-cycle arrest at the S phase and p53-mediated cell apoptosis of BRCA2 monoallelic and biallelic mutant tumor cells. Our study unveils the role of BRCA2 loss in the development of gastrointestinal tumors and provides a potential therapeutic strategy to eliminate BRCA2 monoallelic and biallelic mutant tumors through mitomycin C.


BRCA2 Protein/deficiency , Gastrointestinal Neoplasms/genetics , Mitomycin/metabolism , Animals , Humans , Mice
15.
Mol Cell ; 80(2): 327-344.e8, 2020 10 15.
Article En | MEDLINE | ID: mdl-32966758

Stabilization of stalled replication forks is a prominent mechanism of PARP (Poly(ADP-ribose) Polymerase) inhibitor (PARPi) resistance in BRCA-deficient tumors. Epigenetic mechanisms of replication fork stability are emerging but remain poorly understood. Here, we report the histone acetyltransferase PCAF (p300/CBP-associated) as a fork-associated protein that promotes fork degradation in BRCA-deficient cells by acetylating H4K8 at stalled replication forks, which recruits MRE11 and EXO1. A H4K8ac binding domain within MRE11/EXO1 is required for their recruitment to stalled forks. Low PCAF levels, which we identify in a subset of BRCA2-deficient tumors, stabilize stalled forks, resulting in PARPi resistance in BRCA-deficient cells. Furthermore, PCAF activity is tightly regulated by ATR (ataxia telangiectasia and Rad3-related), which phosphorylates PCAF on serine 264 (S264) to limit its association and activity at stalled forks. Our results reveal PCAF and histone acetylation as critical regulators of fork stability and PARPi responses in BRCA-deficient cells, which provides key insights into targeting BRCA-deficient tumors and identifying epigenetic modulators of chemotherapeutic responses.


BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA Repair Enzymes/metabolism , DNA Replication , Exodeoxyribonucleases/metabolism , Histones/metabolism , MRE11 Homologue Protein/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , DNA Replication/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysine/metabolism , Models, Biological , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Binding/drug effects , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/genetics
16.
Nat Commun ; 11(1): 3839, 2020 07 31.
Article En | MEDLINE | ID: mdl-32737294

Chromatin regulates spatiotemporal gene expression during neurodevelopment, but it also mediates DNA damage repair essential to proliferating neural progenitor cells (NPCs). Here, we uncover molecularly dissociable roles for nucleosome remodeler Ino80 in chromatin-mediated transcriptional regulation and genome maintenance in corticogenesis. We find that conditional Ino80 deletion from cortical NPCs impairs DNA double-strand break (DSB) repair, triggering p53-dependent apoptosis and microcephaly. Using an in vivo DSB repair pathway assay, we find that Ino80 is selectively required for homologous recombination (HR) DNA repair, which is mechanistically distinct from Ino80 function in YY1-associated transcription. Unexpectedly, sensitivity to loss of Ino80-mediated HR is dependent on NPC division mode: Ino80 deletion leads to unrepaired DNA breaks and apoptosis in symmetric NPC-NPC divisions, but not in asymmetric neurogenic divisions. This division mode dependence is phenocopied following conditional deletion of HR gene Brca2. Thus, distinct modes of NPC division have divergent requirements for Ino80-dependent HR DNA repair.


ATPases Associated with Diverse Cellular Activities/genetics , BRCA2 Protein/genetics , Chromatin/chemistry , DNA-Binding Proteins/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics , Recombinational DNA Repair , ATPases Associated with Diverse Cellular Activities/deficiency , Animals , Apoptosis/genetics , BRCA2 Protein/deficiency , Cell Division , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/deficiency , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Neocortex/cytology , Neocortex/growth & development , Neocortex/metabolism , Neural Stem Cells/cytology , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
17.
Physiol Rep ; 8(13): e14481, 2020 07.
Article En | MEDLINE | ID: mdl-32638521

Mutations in the tumor suppressor gene BRCA2 (BReast CAncer susceptibility gene 2) predispose carriers to breast, ovarian, and other cancers. In response to DNA damage, BRCA2 participates in homology-directed DNA damage repair to maintain genome stability. Genome-wide association studies have identified an association between BRCA2 single nucleotide polymorphisms and plasma-lipid levels and lipid deregulation in humans. To date, DNA damage, apoptosis, and lipid deregulation are recognized as central pathways for endothelial dysfunction and atherosclerosis; however, the role of BRCA2 in endothelial dysfunction remains to be elucidated. To determine the role of BRCA2 in endothelial dysfunction, BRCA2 was silenced in human umbilical vein endothelial cells (ECs) and assessed for markers of DNA damage, apoptosis, and endothelial function following oxidized low-density lipoprotein (oxLDL) treatment. OxLDL was found to induce significant reactive oxygen species (ROS) production in BRCA2-silenced ECs. This increase in ROS production was associated with exacerbated DNA damage evidenced by increased expression and activation of DNA double-stranded break (DSB) marker γH2AX and reduced RAD51-foci formation-an essential regulator of DSB repair. Increased DSBs were associated with enhanced expression and activation of pro-apoptotic p53 and significant apoptosis in oxLDL-treated BRCA2-silenced ECs. Loss of BRCA2 in ECs was further associated with oxLDL-induced impaired tube-forming potential and eNOS expression. Collectively, the data reveals, for the first time, a novel role of BRCA2 as a regulator of EC survival and function in the setting of oxLDL treatment in vitro. Additionally, the data provide important clues regarding the potential susceptibility of BRCA2 mutation carriers to endothelial dysfunction, atherosclerosis, and other cardiovascular diseases.


Apoptosis , BRCA2 Protein/genetics , DNA Breaks, Double-Stranded , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Animals , BRCA2 Protein/deficiency , Humans , Lipoproteins, LDL/toxicity , Male , Mice , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism
18.
Proc Natl Acad Sci U S A ; 117(32): 19415-19424, 2020 08 11.
Article En | MEDLINE | ID: mdl-32719125

Synthetic lethality strategies for cancer therapy exploit cancer-specific genetic defects to identify targets that are uniquely essential to the survival of tumor cells. Here we show RAD27/FEN1, which encodes flap endonuclease 1 (FEN1), a structure-specific nuclease with roles in DNA replication and repair, and has the greatest number of synthetic lethal interactions with Saccharomyces cerevisiae genome instability genes, is a druggable target for an inhibitor-based approach to kill cancers with defects in homologous recombination (HR). The vulnerability of cancers with HR defects to FEN1 loss was validated by studies showing that small-molecule FEN1 inhibitors and FEN1 small interfering RNAs (siRNAs) selectively killed BRCA1- and BRCA2-defective human cell lines. Furthermore, the differential sensitivity to FEN1 inhibition was recapitulated in mice, where a small-molecule FEN1 inhibitor reduced the growth of tumors established from drug-sensitive but not drug-resistant cancer cell lines. FEN1 inhibition induced a DNA damage response in both sensitive and resistant cell lines; however, sensitive cell lines were unable to recover and replicate DNA even when the inhibitor was removed. Although FEN1 inhibition activated caspase to higher levels in sensitive cells, this apoptotic response occurred in p53-defective cells and cell killing was not blocked by a pan-caspase inhibitor. These results suggest that FEN1 inhibitors have the potential for therapeutically targeting HR-defective cancers such as those resulting from BRCA1 and BRCA2 mutations, and other genetic defects.


Antineoplastic Agents/pharmacology , Flap Endonucleases/antagonists & inhibitors , Homologous Recombination/drug effects , Neoplasms/genetics , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair/drug effects , DNA Replication/drug effects , Flap Endonucleases/genetics , Genomic Instability/genetics , Humans , Mice , Neoplasms/drug therapy , RNA, Small Interfering/pharmacology , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Small Molecule Libraries/pharmacology , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays
19.
J Cell Biol ; 219(6)2020 06 01.
Article En | MEDLINE | ID: mdl-32391871

BRCA1/2 help maintain genomic integrity by stabilizing stalled forks. Here, we identify the E3 ligase RFWD3 as an essential modulator of stalled fork stability in BRCA2-deficient cells and show that codepletion of RFWD3 rescues fork degradation, collapse, and cell sensitivity upon replication stress. Stalled forks in BRCA2-deficient cells accumulate phosphorylated and ubiquitinated replication protein A (ubq-pRPA), the latter of which is mediated by RFWD3. Generation of this intermediate requires SMARCAL1, suggesting that it depends on stalled fork reversal. We show that in BRCA2-deficient cells, rescuing fork degradation might not be sufficient to ensure fork repair. Depleting MRE11 in BRCA2-deficient cells does block fork degradation, but it does not prevent fork collapse and cell sensitivity in the presence of replication stress. No such ubq-pRPA intermediate is formed in BRCA1-deficient cells, and our results suggest that BRCA1 may function upstream of BRCA2 in the stalled fork repair pathway. Collectively, our data uncover a novel mechanism by which RFWD3 destabilizes forks in BRCA2-deficient cells.


BRCA2 Protein/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Replication Protein A/metabolism , Ubiquitin-Protein Ligases/metabolism , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Cell Cycle , Cell Line, Tumor , Cell Survival/genetics , DNA Damage/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication/drug effects , Humans , Hydroxyurea/pharmacology , MRE11 Homologue Protein/deficiency , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Mutation , Phosphorylation , RNA, Small Interfering , Rad51 Recombinase/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
20.
Nat Commun ; 11(1): 819, 2020 02 10.
Article En | MEDLINE | ID: mdl-32041954

Loss of functional BRCA1 protein leads to defects in DNA double-strand break (DSB) repair by homologous recombination (HR) and renders cells hypersensitive to poly (ADP-ribose) polymerase (PARP) inhibitors used to treat BRCA1/2-deficient cancers. However, upon chronic treatment of BRCA1-mutant cells with PARP inhibitors, resistant clones can arise via several mechanisms, including loss of 53BP1 or its downstream co-factors. Defects in the 53BP1 axis partially restore the ability of a BRCA1-deficient cell to form RAD51 filaments at resected DSBs in a PALB2- and BRCA2-dependent manner, and thereby repair DSBs by HR. Here we show that depleting 53BP1 in BRCA1-null cells restores PALB2 accrual at resected DSBs. Moreover, we demonstrate that PALB2 DSB recruitment in BRCA1/53BP1-deficient cells is mediated by an interaction between PALB2's chromatin associated motif (ChAM) and the nucleosome acidic patch region, which in 53BP1-expressing cells is bound by 53BP1's ubiquitin-directed recruitment (UDR) domain.


BRCA1 Protein/deficiency , Chromatin/metabolism , Fanconi Anemia Complementation Group N Protein/metabolism , Homologous Recombination , Tumor Suppressor p53-Binding Protein 1/deficiency , Amino Acid Motifs , BRCA2 Protein/deficiency , Cell Line , DNA Breaks, Double-Stranded , DNA Repair/genetics , Fanconi Anemia Complementation Group N Protein/chemistry , Fanconi Anemia Complementation Group N Protein/deficiency , Fanconi Anemia Complementation Group N Protein/genetics , Humans , Nucleosomes/metabolism
...