Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.452
Filter
Add more filters

Publication year range
1.
Cell ; 187(9): 2124-2126, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670069

ABSTRACT

Many types of tumor cells alter metabolic pathways to meet their energy and biosynthetic demands for proliferation or stress adaptation. In this issue of Cell, Kong et al. find that the glycolytic metabolite methylglyoxal causes cancer-associated mutant single-base substitution features by inducing BRCA2 proteolysis, leading to functional haploinsufficiency of BRCA2.


Subject(s)
BRCA2 Protein , Glycolysis , Haploinsufficiency , Humans , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Pyruvaldehyde/metabolism , Mutation
2.
Cell ; 187(9): 2269-2287.e16, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38608703

ABSTRACT

Knudson's "two-hit" paradigm posits that carcinogenesis requires inactivation of both copies of an autosomal tumor suppressor gene. Here, we report that the glycolytic metabolite methylglyoxal (MGO) transiently bypasses Knudson's paradigm by inactivating the breast cancer suppressor protein BRCA2 to elicit a cancer-associated, mutational single-base substitution (SBS) signature in nonmalignant mammary cells or patient-derived organoids. Germline monoallelic BRCA2 mutations predispose to these changes. An analogous SBS signature, again without biallelic BRCA2 inactivation, accompanies MGO accumulation and DNA damage in Kras-driven, Brca2-mutant murine pancreatic cancers and human breast cancers. MGO triggers BRCA2 proteolysis, temporarily disabling BRCA2's tumor suppressive functions in DNA repair and replication, causing functional haploinsufficiency. Intermittent MGO exposure incites episodic SBS mutations without permanent BRCA2 inactivation. Thus, a metabolic mechanism wherein MGO-induced BRCA2 haploinsufficiency transiently bypasses Knudson's two-hit requirement could link glycolysis activation by oncogenes, metabolic disorders, or dietary challenges to mutational signatures implicated in cancer evolution.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Glycolysis , Pyruvaldehyde , Animals , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Mice , Humans , Female , Pyruvaldehyde/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Haploinsufficiency , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Mutation , DNA Damage , DNA Repair , Cell Line, Tumor
3.
Cell ; 184(4): 1064-1080.e20, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606977

ABSTRACT

Understanding the functional consequences of single-nucleotide variants is critical to uncovering the genetic underpinnings of diseases, but technologies to characterize variants are limiting. Here, we leverage CRISPR-Cas9 cytosine base editors in pooled screens to scalably assay variants at endogenous loci in mammalian cells. We benchmark the performance of base editors in positive and negative selection screens, identifying known loss-of-function mutations in BRCA1 and BRCA2 with high precision. To demonstrate the utility of base editor screens to probe small molecule-protein interactions, we screen against BH3 mimetics and PARP inhibitors, identifying point mutations that confer drug sensitivity or resistance. We also create a library of single guide RNAs (sgRNAs) predicted to generate 52,034 ClinVar variants in 3,584 genes and conduct screens in the presence of cellular stressors, identifying loss-of-function variants in numerous DNA damage repair genes. We anticipate that this screening approach will be broadly useful to readily and scalably functionalize genetic variants.


Subject(s)
Gene Editing , Genetic Variation , High-Throughput Nucleotide Sequencing , Alleles , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Base Sequence , Catalytic Domain , Cell Line, Tumor , Humans , Loss of Function Mutation , Mutagenesis/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Point Mutation/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Reproducibility of Results , Selection, Genetic , bcl-X Protein/genetics
4.
Annu Rev Biochem ; 88: 221-245, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30917004

ABSTRACT

Mutations in the BRCA1 and BRCA2 genes predispose afflicted individuals to breast, ovarian, and other cancers. The BRCA-encoded products form complexes with other tumor suppressor proteins and with the recombinase enzyme RAD51 to mediate chromosome damage repair by homologous recombination and also to protect stressed DNA replication forks against spurious nucleolytic attrition. Understanding how the BRCA tumor suppressor network executes its biological functions would provide the foundation for developing targeted cancer therapeutics, but progress in this area has been greatly hampered by the challenge of obtaining purified BRCA complexes for mechanistic studies. In this article, we review how recent effort begins to overcome this technical challenge, leading to functional and structural insights into the biochemical attributes of these complexes and the multifaceted roles that they fulfill in genome maintenance. We also highlight the major mechanistic questions that remain.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Gene Regulatory Networks , Rad51 Recombinase/genetics , Recombinational DNA Repair , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Replication , Female , Genome, Human , Genomic Instability , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
5.
Cell ; 173(7): 1755-1769.e22, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29754820

ABSTRACT

High-grade serous ovarian cancer (HGSC) exhibits extensive malignant clonal diversity with widespread but non-random patterns of disease dissemination. We investigated whether local immune microenvironment factors shape tumor progression properties at the interface of tumor-infiltrating lymphocytes (TILs) and cancer cells. Through multi-region study of 212 samples from 38 patients with whole-genome sequencing, immunohistochemistry, histologic image analysis, gene expression profiling, and T and B cell receptor sequencing, we identified three immunologic subtypes across samples and extensive within-patient diversity. Epithelial CD8+ TILs negatively associated with malignant diversity, reflecting immunological pruning of tumor clones inferred by neoantigen depletion, HLA I loss of heterozygosity, and spatial tracking between T cell and tumor clones. In addition, combinatorial prognostic effects of mutational processes and immune properties were observed, illuminating how specific genomic aberration types associate with immune response and impact survival. We conclude that within-patient spatial immune microenvironment variation shapes intraperitoneal malignant spread, provoking new evolutionary perspectives on HGSC clonal dispersion.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , CD8 Antigens/metabolism , Cluster Analysis , Female , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , Loss of Heterozygosity , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/classification , Ovarian Neoplasms/immunology , Polymorphism, Single Nucleotide , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Whole Genome Sequencing , Young Adult
6.
Cell ; 169(6): 979-981, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28575676

ABSTRACT

Humans with inherited heterozygous BRCA2 mutations have an increased risk of developing cancer; however, what triggers carcinogenesis in these individuals is unclear. Tan et al. find that environmental and metabolic aldehydes pose a threat to these individuals by promoting degradation of wild-type BRCA2 protein, thereby predisposing them to genomic instability and perhaps to cancer.


Subject(s)
Aldehydes , Mutation , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Genes, BRCA1 , Genomic Instability , Germ-Line Mutation , Heterozygote , Humans
7.
Cell ; 169(2): 183, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388401

ABSTRACT

Rucaparib is an inhibitor of nuclear poly (ADP-ribose) polymerases (inhibition of PARP-1 > PARP-2 > PARP-3), following a similar drug, Olaparib. It disrupts DNA repair and replication pathways (and possibly transcription), leading to selective killing of cancer cells with BRCA1/2 mutations. Rucaparib is approved for recurrent ovarian cancers with germline or somatic mutations in BRCA1/2.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Drug Approval , Female , Humans , Mutation , Ovarian Neoplasms/genetics
8.
Cell ; 169(6): 1105-1118.e15, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575672

ABSTRACT

Mutations truncating a single copy of the tumor suppressor, BRCA2, cause cancer susceptibility. In cells bearing such heterozygous mutations, we find that a cellular metabolite and ubiquitous environmental toxin, formaldehyde, stalls and destabilizes DNA replication forks, engendering structural chromosomal aberrations. Formaldehyde selectively depletes BRCA2 via proteasomal degradation, a mechanism of toxicity that affects very few additional cellular proteins. Heterozygous BRCA2 truncations, by lowering pre-existing BRCA2 expression, sensitize to BRCA2 haploinsufficiency induced by transient exposure to natural concentrations of formaldehyde. Acetaldehyde, an alcohol catabolite detoxified by ALDH2, precipitates similar effects. Ribonuclease H1 ameliorates replication fork instability and chromosomal aberrations provoked by aldehyde-induced BRCA2 haploinsufficiency, suggesting that BRCA2 inactivation triggers spontaneous mutagenesis during DNA replication via aberrant RNA-DNA hybrids (R-loops). These findings suggest a model wherein carcinogenesis in BRCA2 mutation carriers can be incited by compounds found pervasively in the environment and generated endogenously in certain tissues with implications for public health.


Subject(s)
BRCA2 Protein/genetics , Chromosome Aberrations/drug effects , Formaldehyde/toxicity , Genomic Instability/drug effects , Toxins, Biological/toxicity , DNA Damage , DNA Replication/drug effects , DNA-Binding Proteins/metabolism , Haploinsufficiency , HeLa Cells , Humans , MRE11 Homologue Protein , Proteome , Ribonuclease H/metabolism
9.
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
10.
Mol Cell ; 84(4): 659-674.e7, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266640

ABSTRACT

Inactivating mutations in the BRCA1 and BRCA2 genes impair DNA double-strand break (DSB) repair by homologous recombination (HR), leading to chromosomal instability and cancer. Importantly, BRCA1/2 deficiency also causes therapeutically targetable vulnerabilities. Here, we identify the dependency on the end resection factor EXO1 as a key vulnerability of BRCA1-deficient cells. EXO1 deficiency generates poly(ADP-ribose)-decorated DNA lesions during S phase that associate with unresolved DSBs and genomic instability in BRCA1-deficient but not in wild-type or BRCA2-deficient cells. Our data indicate that BRCA1/EXO1 double-deficient cells accumulate DSBs due to impaired repair by single-strand annealing (SSA) on top of their HR defect. In contrast, BRCA2-deficient cells retain SSA activity in the absence of EXO1 and hence tolerate EXO1 loss. Consistent with a dependency on EXO1-mediated SSA, we find that BRCA1-mutated tumors show elevated EXO1 expression and increased SSA-associated genomic scars compared with BRCA1-proficient tumors. Overall, our findings uncover EXO1 as a promising therapeutic target for BRCA1-deficient tumors.


Subject(s)
BRCA1 Protein , Neoplasms , Humans , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA Damage , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Homologous Recombination
11.
Cell ; 165(1): 35-44, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26997480

ABSTRACT

PD-1 immune checkpoint blockade provides significant clinical benefits for melanoma patients. We analyzed the somatic mutanomes and transcriptomes of pretreatment melanoma biopsies to identify factors that may influence innate sensitivity or resistance to anti-PD-1 therapy. We find that overall high mutational loads associate with improved survival, and tumors from responding patients are enriched for mutations in the DNA repair gene BRCA2. Innately resistant tumors display a transcriptional signature (referred to as the IPRES, or innate anti-PD-1 resistance), indicating concurrent up-expression of genes involved in the regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. Notably, mitogen-activated protein kinase (MAPK)-targeted therapy (MAPK inhibitor) induces similar signatures in melanoma, suggesting that a non-genomic form of MAPK inhibitor resistance mediates cross-resistance to anti-PD-1 therapy. Validation of the IPRES in other independent tumor cohorts defines a transcriptomic subset across distinct types of advanced cancer. These findings suggest that attenuating the biological processes that underlie IPRES may improve anti-PD-1 response in melanoma and other cancer types.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Melanoma/drug therapy , Neoplasm Metastasis/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , BRCA2 Protein/genetics , Humans , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Neoplasm Metastasis/genetics , Nivolumab , Transcriptome
12.
Mol Cell ; 83(16): 2925-2940.e8, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37499663

ABSTRACT

Homologous recombination (HR) is essential for error-free repair of DNA double-strand breaks, perturbed replication forks (RFs), and post-replicative single-stranded DNA (ssDNA) gaps. To initiate HR, the recombination mediator and tumor suppressor protein BRCA2 facilitates nucleation of RAD51 on ssDNA prior to stimulation of RAD51 filament growth by RAD51 paralogs. Although ssDNA binding by BRCA2 has been implicated in RAD51 nucleation, the function of double-stranded DNA (dsDNA) binding by BRCA2 remains unclear. Here, we exploit single-molecule (SM) imaging to visualize BRCA2-mediated RAD51 nucleation in real time using purified proteins. We report that BRCA2 nucleates and stabilizes RAD51 on ssDNA either directly or through an unappreciated diffusion-assisted delivery mechanism involving binding to and sliding along dsDNA, which requires the cooperative action of multiple dsDNA-binding modules in BRCA2. Collectively, our work reveals two distinct mechanisms of BRCA2-dependent RAD51 loading onto ssDNA, which we propose are critical for its diverse functions in maintaining genome stability and cancer suppression.


Subject(s)
BRCA2 Protein , Rad51 Recombinase , Humans , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , DNA-Binding Proteins/metabolism , DNA, Single-Stranded/genetics , DNA/metabolism , DNA Repair , Protein Binding
13.
Mol Cell ; 83(13): 2347-2356.e8, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37311462

ABSTRACT

Oncogenic mutations in isocitrate dehydrogenases 1 and 2 (IDH1/2) produce 2-hydroxyglutarate (2HG), which inhibits dioxygenases that modulate chromatin dynamics. The effects of 2HG have been reported to sensitize IDH tumors to poly-(ADP-ribose) polymerase (PARP) inhibitors. However, unlike PARP-inhibitor-sensitive BRCA1/2 tumors, which exhibit impaired homologous recombination, IDH-mutant tumors have a silent mutational profile and lack signatures associated with impaired homologous recombination. Instead, 2HG-producing IDH mutations lead to a heterochromatin-dependent slowing of DNA replication accompanied by increased replication stress and DNA double-strand breaks. This replicative stress manifests as replication fork slowing, but the breaks are repaired without a significant increase in mutation burden. Faithful resolution of replicative stress in IDH-mutant cells is dependent on poly-(ADP-ribosylation). Treatment with PARP inhibitors increases DNA replication but results in incomplete DNA repair. These findings demonstrate a role for PARP in the replication of heterochromatin and further validate PARP as a therapeutic target in IDH-mutant tumors.


Subject(s)
BRCA1 Protein , Neoplasms , Humans , BRCA1 Protein/genetics , Heterochromatin/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , BRCA2 Protein/genetics , Homologous Recombination/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Mutation , Isocitrate Dehydrogenase/genetics
14.
Mol Cell ; 82(20): 3781-3793.e7, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36099913

ABSTRACT

Germline mutations in the BRCA genes are associated with a higher risk of carcinogenesis, which is linked to an increased mutation rate and loss of the second unaffected BRCA allele (loss of heterozygosity, LOH). However, the mechanisms triggering mutagenesis are not clearly understood. The BRCA genes contain high numbers of repetitive DNA sequences. We detected replication forks stalling, DNA breaks, and deletions at these sites in haploinsufficient BRCA cells, thus identifying the BRCA genes as fragile sites. Next, we found that stalled forks are repaired by error-prone pathways, such as microhomology-mediated break-induced replication (MMBIR) in haploinsufficient BRCA1 breast epithelial cells. We detected MMBIR mutations in BRCA1 tumor cells and noticed deletions-insertions (>50 bp) at the BRCA1 genes in BRCA1 patients. Altogether, these results suggest that under stress, error-prone repair of stalled forks is upregulated and induces mutations, including complex genomic rearrangements at the BRCA genes (LOH), in haploinsufficient BRCA1 cells.


Subject(s)
BRCA1 Protein , DNA Replication , Humans , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Repair , Mutagenesis , Genes, BRCA1 , Loss of Heterozygosity , BRCA2 Protein/genetics , BRCA2 Protein/metabolism
15.
Mol Cell ; 82(18): 3382-3397.e7, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36002001

ABSTRACT

Aberrant replication causes cells lacking BRCA2 to enter mitosis with under-replicated DNA, which activates a repair mechanism known as mitotic DNA synthesis (MiDAS). Here, we identify genome-wide the sites where MiDAS reactions occur when BRCA2 is abrogated. High-resolution profiling revealed that these sites are different from MiDAS at aphidicolin-induced common fragile sites in that they map to genomic regions replicating in the early S-phase, which are close to early-firing replication origins, are highly transcribed, and display R-loop-forming potential. Both transcription inhibition in early S-phase and RNaseH1 overexpression reduced MiDAS in BRCA2-deficient cells, indicating that transcription-replication conflicts (TRCs) and R-loops are the source of MiDAS. Importantly, the MiDAS sites identified in BRCA2-deficient cells also represent hotspots for genomic rearrangements in BRCA2-mutated breast tumors. Thus, our work provides a mechanism for how tumor-predisposing BRCA2 inactivation links transcription-induced DNA damage with mitotic DNA repair to fuel the genomic instability characteristic of cancer cells.


Subject(s)
DNA Replication , Mitosis , Aphidicolin/pharmacology , BRCA2 Protein/genetics , Chromosome Fragile Sites/genetics , DNA/genetics , DNA Damage , Genomic Instability , Humans , Mitosis/genetics
16.
Mol Cell ; 82(13): 2363-2369, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35568026

ABSTRACT

Defects in DNA double-strand break repair are thought to render BRCA1 or BRCA2 (BRCA) mutant tumors selectively sensitive to PARP inhibitors (PARPis). Challenging this framework, BRCA and PARP1 share functions in DNA synthesis on the lagging strand. Thus, BRCA deficiency or "BRCAness" could reflect an inherent lagging strand problem that is vulnerable to drugs such as PARPi that also target the lagging strand, a combination that generates a toxic accumulation of replication gaps.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA Breaks, Double-Stranded , DNA Repair , Neoplasms , BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA , DNA Repair/genetics , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
17.
Mol Cell ; 82(19): 3553-3565.e5, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36070766

ABSTRACT

RAD51 and the breast cancer suppressor BRCA2 have critical functions in DNA double-strand (dsDNA) break repair by homologous recombination and the protection of newly replicated DNA from nucleolytic degradation. The recombination function of RAD51 requires its binding to single-stranded DNA (ssDNA), whereas binding to dsDNA is inhibitory. Using reconstituted MRE11-, EXO1-, and DNA2-dependent nuclease reactions, we show that the protective function of RAD51 unexpectedly depends on its binding to dsDNA. The BRC4 repeat of BRCA2 abrogates RAD51 binding to dsDNA and accordingly impairs the function of RAD51 in protection. The BRCA2 C-terminal RAD51-binding segment (TR2) acts in a dominant manner to overcome the effect of BRC4. Mechanistically, TR2 stabilizes RAD51 binding to dsDNA, even in the presence of BRC4, promoting DNA protection. Our data suggest that RAD51's dsDNA-binding capacity may have evolved together with its function in replication fork protection and provide a mechanistic basis for the DNA-protection function of BRCA2.


Subject(s)
DNA, Single-Stranded , Rad51 Recombinase , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA/genetics , DNA Breaks, Double-Stranded , DNA Repair , DNA Replication , DNA, Single-Stranded/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
18.
Nature ; 621(7977): 129-137, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37587346

ABSTRACT

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.


Subject(s)
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
19.
Mol Cell ; 81(5): 1074-1083.e5, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33453169

ABSTRACT

The RAD51 recombinase forms nucleoprotein filaments to promote double-strand break repair, replication fork reversal, and fork stabilization. The stability of these filaments is highly regulated, as both too little and too much RAD51 activity can cause genome instability. RADX is a single-strand DNA (ssDNA) binding protein that regulates DNA replication. Here, we define its mechanism of action. We find that RADX inhibits RAD51 strand exchange and D-loop formation activities. RADX directly and selectively interacts with ATP-bound RAD51, stimulates ATP hydrolysis, and destabilizes RAD51 nucleofilaments. The RADX interaction with RAD51, in addition to its ssDNA binding capability, is required to maintain replication fork elongation rates and fork stability. Furthermore, BRCA2 can overcome the RADX-dependent RAD51 inhibition. Thus, RADX functions in opposition to BRCA2 in regulating RAD51 nucleofilament stability to ensure the right level of RAD51 function during DNA replication.


Subject(s)
BRCA2 Protein/genetics , DNA Replication , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Rad51 Recombinase/genetics , Adenosine Triphosphate/metabolism , BRCA2 Protein/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hydrolysis , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , RNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Signal Transduction , Single Molecule Imaging , Red Fluorescent Protein
20.
Mol Cell ; 81(19): 4008-4025.e7, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34508659

ABSTRACT

BRCA1/2 mutant tumor cells display an elevated mutation burden, the etiology of which remains unclear. Here, we report that these cells accumulate ssDNA gaps and spontaneous mutations during unperturbed DNA replication due to repriming by the DNA primase-polymerase PRIMPOL. Gap accumulation requires the DNA glycosylase SMUG1 and is exacerbated by depletion of the translesion synthesis (TLS) factor RAD18 or inhibition of the error-prone TLS polymerase complex REV1-Polζ by the small molecule JH-RE-06. JH-RE-06 treatment of BRCA1/2-deficient cells results in reduced mutation rates and PRIMPOL- and SMUG1-dependent loss of viability. Through cellular and animal studies, we demonstrate that JH-RE-06 is preferentially toxic toward HR-deficient cancer cells. Furthermore, JH-RE-06 remains effective toward PARP inhibitor (PARPi)-resistant BRCA1 mutant cells and displays additive toxicity with crosslinking agents or PARPi. Collectively, these studies identify a protective and mutagenic role for REV1-Polζ in BRCA1/2 mutant cells and provide the rationale for using REV1-Polζ inhibitors to treat BRCA1/2 mutant tumors.


Subject(s)
DNA Breaks, Single-Stranded , DNA Primase/metabolism , DNA Replication , DNA, Neoplasm/biosynthesis , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/metabolism , Neoplasms/enzymology , Nucleotidyltransferases/metabolism , Recombinational DNA Repair , Animals , Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line, Tumor , DNA Primase/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Female , HEK293 Cells , Humans , Mice, Nude , Multifunctional Enzymes/genetics , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/genetics , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL