Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 30(10): 1456-1467, 2023 10.
Article in English | MEDLINE | ID: mdl-37696958

ABSTRACT

The extent and efficacy of DNA end resection at DNA double-strand breaks (DSB) determine the repair pathway choice. Here we describe how the 53BP1-associated protein DYNLL1 works in tandem with the Shieldin complex to protect DNA ends. DYNLL1 is recruited to DSBs by 53BP1, where it limits end resection by binding and disrupting the MRE11 dimer. The Shieldin complex is recruited to a fraction of 53BP1-positive DSBs hours after DYNLL1, predominantly in G1 cells. Shieldin localization to DSBs depends on MRE11 activity and is regulated by the interaction of DYNLL1 with MRE11. BRCA1-deficient cells rendered resistant to PARP inhibitors by the loss of Shieldin proteins can be resensitized by the constitutive association of DYNLL1 with MRE11. These results define the temporal and functional dynamics of the 53BP1-centric DNA end resection factors in cells.


Subject(s)
BRCA1 Protein , DNA Breaks, Double-Stranded , BRCA1 Protein/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , DNA/metabolism , DNA End-Joining Repair , Cell Nucleus/metabolism , DNA Repair
2.
bioRxiv ; 2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37034578

ABSTRACT

Extent and efficacy of DNA end resection at DNA double strand break (DSB)s determines the choice of repair pathway. Here we describe how the 53BP1 associated protein DYNLL1 works in tandem with Shieldin and the CST complex to protect DNA ends. DYNLL1 is recruited to DSBs by 53BP1 where it limits end resection by binding and disrupting the MRE11 dimer. The Shieldin complex is recruited to a fraction of 53BP1-positive DSBs hours after DYNLL1 predominantly in the G1 cells. Shieldin localization to DSBs is dependent on MRE11 activity and is regulated by the interaction of DYNLL1 with MRE11. BRCA1-deficient cells rendered resistant to PARP inhibitors by the loss of Shieldin proteins can be re-sensitized by the constitutive association of DYNLL1 with MRE11. These results define the temporal and functional dynamics of the 53BP1-centric DNA end resection factors in cells.

3.
Cell Rep ; 40(9): 111297, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044844

ABSTRACT

A critical determinant of DNA repair pathway choice is REV7, an adaptor that binds to various DNA repair proteins through its C-terminal seatbelt domain. The REV7 seatbelt binds to either REV3, activating translesion synthesis, or to SHLD3, activating non-homologous end joining (NHEJ) repair. Recent studies have identified another REV7 seatbelt-binding protein, CHAMP1 (chromosome alignment-maintaining phosphoprotein 1), though its possible role in DNA repair is unknown. Here, we show that binding of CHAMP1 to REV7 activates homologous recombination (HR) repair. Mechanistically, CHAMP1 binds directly to REV7 and reduces the level of the Shieldin complex, causing an increase in double-strand break end resection. CHAMP1 also interacts with POGZ in a heterochromatin complex further promoting HR repair. Importantly, in human tumors, CHAMP1 overexpression promotes HR, confers poly (ADP-ribose) polymerase inhibitor resistance, and correlates with poor prognosis. Thus, by binding to either SHLD3 or CHAMP1 through its seatbelt, the REV7 protein can promote either NHEJ or HR repair, respectively.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Mad2 Proteins , Recombinational DNA Repair , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA End-Joining Repair , DNA Repair/genetics , Homologous Recombination , Humans , Mad2 Proteins/metabolism , Phosphoproteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recombinational DNA Repair/genetics , Transposases/metabolism
4.
Cancer Cell ; 40(9): 957-972.e10, 2022 09 12.
Article in English | MEDLINE | ID: mdl-35985342

ABSTRACT

Diffuse midline glioma (DMG) is a uniformly fatal pediatric cancer driven by oncohistones that do not readily lend themselves to drug development. To identify druggable targets for DMG, we conducted a genome-wide CRISPR screen that reveals a DMG selective dependency on the de novo pathway for pyrimidine biosynthesis. This metabolic vulnerability reflects an elevated rate of uridine/uracil degradation that depletes DMG cells of substrates for the alternate salvage pyrimidine biosynthesis pathway. A clinical stage inhibitor of DHODH (rate-limiting enzyme in the de novo pathway) diminishes uridine-5'-phosphate (UMP) pools, generates DNA damage, and induces apoptosis through suppression of replication forks-an "on-target" effect, as shown by uridine rescue. Matrix-assisted laser desorption/ionization (MALDI) mass spectroscopy imaging demonstrates that this DHODH inhibitor (BAY2402234) accumulates in the brain at therapeutically relevant concentrations, suppresses de novo pyrimidine biosynthesis in vivo, and prolongs survival of mice bearing intracranial DMG xenografts, highlighting BAY2402234 as a promising therapy against DMGs.


Subject(s)
Glioma , Pyrimidines , Animals , Glioma/drug therapy , Glioma/genetics , Humans , Mice , Uridine/metabolism , Uridine/pharmacology
5.
Blood Adv ; 6(12): 3803-3811, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35500223

ABSTRACT

Fanconi anemia (FA), a genetic DNA repair disorder characterized by marrow failure and cancer susceptibility. In FA mice, metformin improves blood counts and delays tumor development. We conducted a single institution study of metformin in nondiabetic patients with FA to determine feasibility and tolerability of metformin treatment and to assess for improvement in blood counts. Fourteen of 15 patients with at least 1 cytopenia (hemoglobin < 10 g/dL; platelet count < 100 000 cells/µL; or an absolute neutrophil count < 1000 cells/µL) were eligible to receive metformin for 6 months. Median patient age was 9.4 years (range 6.0-26.5 ). Thirteen of 14 subjects (93%) tolerated maximal dosing for age; 1 subject had dose reduction for grade 2 gastrointestinal symptoms. No subjects developed hypoglycemia or metabolic acidosis. No subjects had dose interruptions caused by toxicity, and no grade 3 or higher adverse events attributed to metformin were observed. Hematologic response based on modified Myelodysplastic Syndrome International Working Group criteria was observed in 4 of 13 evaluable patients (30.8%; 90% confidence interval, 11.3-57.3). Median time to response was 84.5 days (range 71-128 days). Responses were noted in neutrophils (n = 3), platelets (n = 1), and red blood cells (n = 1). No subjects met criteria for disease progression or relapse during treatment. Correlative studies explored potential mechanisms of metformin activity in FA. Plasma proteomics showed reduction in inflammatory pathways with metformin. Metformin is safe and tolerable in nondiabetic patients with FA and may provide therapeutic benefit. This trial was registered at as #NCT03398824.


Subject(s)
Fanconi Anemia , Metformin , Child , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Humans , Metformin/therapeutic use , Young Adult
6.
Cancer Res ; 81(10): 2774-2787, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33514515

ABSTRACT

Homologous recombination (HR)-deficient cancers are sensitive to poly-ADP ribose polymerase inhibitors (PARPi), which have shown clinical efficacy in the treatment of high-grade serous cancers (HGSC). However, the majority of patients will relapse, and acquired PARPi resistance is emerging as a pressing clinical problem. Here we generated seven single-cell clones with acquired PARPi resistance derived from a PARPi-sensitive TP53 -/- and BRCA1 -/- epithelial cell line generated using CRISPR/Cas9. These clones showed diverse resistance mechanisms, and some clones presented with multiple mechanisms of resistance at the same time. Genomic analysis of the clones revealed unique transcriptional and mutational profiles and increased genomic instability in comparison with a PARPi-sensitive cell line. Clonal evolutionary analyses suggested that acquired PARPi resistance arose via clonal selection from an intrinsically unstable and heterogenous cell population in the sensitive cell line, which contained preexisting drug-tolerant cells. Similarly, clonal and spatial heterogeneity in tumor biopsies from a clinical patient with BRCA1-mutant HGSC with acquired PARPi resistance was observed. In an imaging-based drug screening, the clones showed heterogenous responses to targeted therapeutic agents, indicating that not all PARPi-resistant clones can be targeted with just one therapy. Furthermore, PARPi-resistant clones showed mechanism-dependent vulnerabilities to the selected agents, demonstrating that a deeper understanding on the mechanisms of resistance could lead to improved targeting and biomarkers for HGSC with acquired PARPi resistance. SIGNIFICANCE: This study shows that BRCA1-deficient cells can give rise to multiple genomically and functionally heterogenous PARPi-resistant clones, which are associated with various vulnerabilities that can be targeted in a mechanism-specific manner.


Subject(s)
BRCA1 Protein/physiology , Clonal Evolution , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Cell Proliferation , Female , Genomic Instability , Homologous Recombination , Humans , Mice , Mice, Knockout , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Transcriptome , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 117(43): 26795-26803, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33051298

ABSTRACT

The repair of DNA double strand breaks (DSBs) that arise from external mutagenic agents and routine cellular processes is essential for life. DSBs are repaired by two major pathways, homologous recombination (HR) and classical nonhomologous end joining (C-NHEJ). DSB repair pathway choice is largely dictated at the step of 5'-3' DNA end resection, which is promoted during S phase, in part by BRCA1. Opposing end resection is the 53BP1 protein, which recruits the ssDNA-binding REV7-Shieldin complex to favor C-NHEJ repair. We recently identified TRIP13 as a proresection factor that remodels REV7, causing its dissociation from the Shieldin subunit SHLD3. Here, we identify p31comet, a negative regulator of MAD2 and the spindle assembly checkpoint, as an important mediator of the TRIP13-REV7 interaction. p31comet binds to the REV7-Shieldin complex in cells, promotes REV7 inactivation, and causes PARP inhibitor resistance. p31comet also participates in the extraction of REV7 from the chromatin. Furthermore, p31comet can counteract REV7 function in translesion synthesis (TLS) by releasing it from REV3 in the Pol ζ complex. Finally, p31comet, like TRIP13, is overexpressed in many cancers and this correlates with poor prognosis. Thus, we reveal a key player in the regulation of HR and TLS with significant clinical implications.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Mad2 Proteins/metabolism , Nuclear Proteins/metabolism , Recombinational DNA Repair , Cell Line, Tumor , HEK293 Cells , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/mortality
8.
Cell Rep ; 30(7): 2402-2415.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075772

ABSTRACT

Cells deficient in ataxia telangiectasia mutated (ATM) are hypersensitive to ionizing radiation and other anti-cancer agents that induce double-strand DNA breaks. ATM inhibitors may therefore sensitize cancer cells to these agents. Some cancers may also have underlying genetic defects predisposing them to an ATM inhibitor monotherapy response. We have conducted a genome-wide CRISPR screen to identify genetic vulnerabilities that sensitize lung cancer cells to ATM inhibitors. Knockout of genes in the Fanconi anemia (FA)/BRCA pathway results in hypersensitivity to the ATM inhibitor M3541. Knockdown of either an FA gene or of ATM results in reduced double-strand break end resection, enhanced non-homologous end joining (NHEJ) repair, and decreased homologous recombination repair. Knockout of both the FA/BRCA pathway and ATM strongly inhibits end resection and generates toxic levels of NHEJ, thereby elucidating a mechanism of cellular death by synthetic lethality. ATM inhibitors may therefore be useful for the treatment of tumors with a defective FA/BRCA pathway.


Subject(s)
Ataxia Telangiectasia/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Humans
9.
Nat Cell Biol ; 22(1): 87-96, 2020 01.
Article in English | MEDLINE | ID: mdl-31915374

ABSTRACT

DNA double-strand breaks (DSBs) are repaired through homology-directed repair (HDR) or non-homologous end joining (NHEJ). BRCA1/2-deficient cancer cells cannot perform HDR, conferring sensitivity to poly(ADP-ribose) polymerase inhibitors (PARPi). However, concomitant loss of the pro-NHEJ factors 53BP1, RIF1, REV7-Shieldin (SHLD1-3) or CST-DNA polymerase alpha (Pol-α) in BRCA1-deficient cells restores HDR and PARPi resistance. Here, we identify the TRIP13 ATPase as a negative regulator of REV7. We show that REV7 exists in active 'closed' and inactive 'open' conformations, and TRIP13 catalyses the inactivating conformational change, thereby dissociating REV7-Shieldin to promote HDR. TRIP13 similarly disassembles the REV7-REV3 translesion synthesis (TLS) complex, a component of the Fanconi anaemia pathway, inhibiting error-prone replicative lesion bypass and interstrand crosslink repair. Importantly, TRIP13 overexpression is common in BRCA1-deficient cancers, confers PARPi resistance and correlates with poor prognosis. Thus, TRIP13 emerges as an important regulator of DNA repair pathway choice-promoting HDR, while suppressing NHEJ and TLS.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , BRCA1 Protein/deficiency , Cell Cycle Proteins/genetics , DNA Repair/genetics , Recombinational DNA Repair/genetics , ATPases Associated with Diverse Cellular Activities/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , DNA Damage/drug effects , DNA End-Joining Repair/genetics , DNA Repair/drug effects , DNA Replication/drug effects , DNA Replication/genetics , Humans , Mad2 Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Telomere-Binding Proteins/drug effects , Telomere-Binding Proteins/genetics
10.
PLoS One ; 14(11): e0221288, 2019.
Article in English | MEDLINE | ID: mdl-31721781

ABSTRACT

BRCA2 (also known as FANCD1) is a core component of the Fanconi pathway and suppresses transformation of immature T-cells in mice. However, the contribution of Fanconi-BRCA pathway deficiency to human T-cell acute lymphoblastic leukemia (T-ALL) remains undefined. We identified point mutations in 9 (23%) of 40 human T-ALL cases analyzed, with variant allele fractions consistent with heterozygous mutations early in tumor evolution. Two of these mutations were present in remission bone marrow specimens, suggesting germline alterations. BRCA2 was the most commonly mutated gene. The identified Fanconi-BRCA mutations encode hypomorphic or null alleles, as evidenced by their inability to fully rescue Fanconi-deficient cells from chromosome breakage, cytotoxicity and/or G2/M arrest upon treatment with DNA cross-linking agents. Disabling the tumor suppressor activity of the Fanconi-BRCA pathway is generally thought to require biallelic gene mutations. However, all mutations identified were monoallelic, and most cases appeared to retain expression of the wild-type allele. Using isogenic T-ALL cells, we found that BRCA2 haploinsufficiency induces selective hypersensitivity to ATR inhibition, in vitro and in vivo. These findings implicate Fanconi-BRCA pathway haploinsufficiency in the molecular pathogenesis of T-ALL, and provide a therapeutic rationale for inhibition of ATR or other druggable effectors of homologous recombination.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Line, Tumor , Child , Genes, BRCA1 , Genes, BRCA2 , Haploinsufficiency , Heterografts , Humans , Jurkat Cells , Male , Mice , Mice, Inbred NOD , Mutagenesis, Site-Directed , Mutation , Radiation Tolerance/genetics , Sequence Analysis, DNA , Sequence Analysis, RNA , Ultraviolet Rays
11.
Nature ; 572(7771): 676-680, 2019 08.
Article in English | MEDLINE | ID: mdl-31391581

ABSTRACT

The CCCTC-binding factor (CTCF), which anchors DNA loops that organize the genome into structural domains, has a central role in gene control by facilitating or constraining interactions between genes and their regulatory elements1,2. In cancer cells, the disruption of CTCF binding at specific loci by somatic mutation3,4 or DNA hypermethylation5 results in the loss of loop anchors and consequent activation of oncogenes. By contrast, the germ-cell-specific paralogue of CTCF, BORIS (brother of the regulator of imprinted sites, also known as CTCFL)6, is overexpressed in several cancers7-9, but its contributions to the malignant phenotype remain unclear. Here we show that aberrant upregulation of BORIS promotes chromatin interactions in ALK-mutated, MYCN-amplified neuroblastoma10 cells that develop resistance to ALK inhibition. These cells are reprogrammed to a distinct phenotypic state during the acquisition of resistance, a process defined by the initial loss of MYCN expression followed by subsequent overexpression of BORIS and a concomitant switch in cellular dependence from MYCN to BORIS. The resultant BORIS-regulated alterations in chromatin looping lead to the formation of super-enhancers that drive the ectopic expression of a subset of proneural transcription factors that ultimately define the resistance phenotype. These results identify a previously unrecognized role of BORIS-to promote regulatory chromatin interactions that support specific cancer phenotypes.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Mice , Molecular Targeted Therapy , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/enzymology , Neuroblastoma/genetics , Phenotype , Protein Binding
12.
Cell Rep ; 17(9): 2367-2381, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880910

ABSTRACT

Although poly(ADP-ribose) polymerase (PARP) inhibitors are active in homologous recombination (HR)-deficient cancers, their utility is limited by acquired resistance after restoration of HR. Here, we report that dinaciclib, an inhibitor of cyclin-dependent kinases (CDKs) 1, 2, 5, and 9, additionally has potent activity against CDK12, a transcriptional regulator of HR. In BRCA-mutated triple-negative breast cancer (TNBC) cells and patient-derived xenografts (PDXs), dinaciclib ablates restored HR and reverses PARP inhibitor resistance. Additionally, we show that de novo resistance to PARP inhibition in BRCA1-mutated cell lines and a PDX derived from a PARP-inhibitor-naive BRCA1 carrier is mediated by residual HR and is reversed by CDK12 inhibition. Finally, dinaciclib augments the degree of response in a PARP-inhibitor-sensitive model, converting tumor growth inhibition to durable regression. These results highlight the significance of HR disruption as a therapeutic strategy and support the broad use of combined CDK12 and PARP inhibition in TNBC.


Subject(s)
BRCA1 Protein/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Mutation/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Amino Acid Sequence , Animals , BRCA1 Protein/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cyclic N-Oxides , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , DNA Damage/genetics , DNA Repair/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Homologous Recombination/drug effects , Humans , Indolizines , Mice , Protein Kinase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
13.
Cell Stem Cell ; 18(5): 668-81, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27053300

ABSTRACT

Fanconi anemia (FA) is an inherited DNA repair disorder characterized by progressive bone marrow failure (BMF) from hematopoietic stem and progenitor cell (HSPC) attrition. A greater understanding of the pathogenesis of BMF could improve the therapeutic options for FA patients. Using a genome-wide shRNA screen in human FA fibroblasts, we identify transforming growth factor-ß (TGF-ß) pathway-mediated growth suppression as a cause of BMF in FA. Blocking the TGF-ß pathway improves the survival of FA cells and rescues the proliferative and functional defects of HSPCs derived from FA mice and FA patients. Inhibition of TGF-ß signaling in FA HSPCs results in elevated homologous recombination (HR) repair with a concomitant decrease in non-homologous end-joining (NHEJ), accounting for the improvement in cellular growth. Together, our results suggest that elevated TGF-ß signaling contributes to BMF in FA by impairing HSPC function and may be a potential therapeutic target for the treatment of FA.


Subject(s)
Bone Marrow/pathology , Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Acetaldehyde/toxicity , Animals , Cell Survival/drug effects , DNA End-Joining Repair/drug effects , Down-Regulation/drug effects , Hematopoietic Stem Cells/drug effects , Homologous Recombination/genetics , Humans , Mice , Mice, Inbred C57BL , Mutagens/toxicity , Signal Transduction/drug effects , Stress, Physiological/drug effects , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects
14.
J Clin Invest ; 125(4): 1523-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751062

ABSTRACT

The Fanconi anemia/BRCA (FA/BRCA) pathway is a DNA repair pathway that is required for excision of DNA interstrand cross-links. The 17 known FA proteins, along with several FA-associated proteins (FAAPs), cooperate in this pathway to detect, unhook, and excise DNA cross-links and to subsequently repair the double-strand breaks generated in the process. In the current study, we identified a patient with FA with a point mutation in FANCA, which encodes a mutant FANCA protein (FANCAI939S). FANCAI939S failed to bind to the FAAP20 subunit of the FA core complex, leading to decreased stability. Loss of FAAP20 binding exposed a SUMOylation site on FANCA at amino acid residue K921, resulting in E2 SUMO-conjugating enzyme UBC9-mediated SUMOylation, RING finger protein 4-mediated (RNF4-mediated) polyubiquitination, and proteasome-mediated degradation of FANCA. Mutation of the SUMOylation site of FANCA rescued the expression of the mutant protein. Wild-type FANCA was also subject to SUMOylation, RNF4-mediated polyubiquitination, and degradation, suggesting that regulated release of FAAP20 from FANCA is a critical step in the normal FA pathway. Consistent with this model, cells lacking RNF4 exhibited interstrand cross-linker hypersensitivity, and the gene encoding RNF4 was epistatic with the other genes encoding members of the FA/BRCA pathway. Together, the results from our study underscore the importance of analyzing unique patient-derived mutations for dissecting complex DNA repair processes.


Subject(s)
BRCA1 Protein/physiology , DNA Repair/physiology , Fanconi Anemia Complementation Group A Protein/physiology , Fanconi Anemia/genetics , Nuclear Proteins/physiology , Transcription Factors/physiology , Adult , Cell Line, Tumor , DNA Repair/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group Proteins/antagonists & inhibitors , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/physiology , Female , Genes, BRCA1 , Humans , Mutation, Missense , Pedigree , Phenotype , Point Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Processing, Post-Translational/physiology , Proteolysis , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Sumoylation , Triple Negative Breast Neoplasms/genetics , Ubiquitination/physiology
15.
Cancer Discov ; 5(2): 135-42, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25472942

ABSTRACT

UNLABELLED: Deficiency in BRCA-dependent DNA interstrand crosslink (ICL) repair is intimately connected to breast cancer susceptibility and to the rare developmental syndrome Fanconi anemia. Bona fide Fanconi anemia proteins, BRCA2 (FANCD1), PALB2 (FANCN), and BRIP1 (FANCJ), interact with BRCA1 during ICL repair. However, the lack of detailed phenotypic and cellular characterization of a patient with biallelic BRCA1 mutations has precluded assignment of BRCA1 as a definitive Fanconi anemia susceptibility gene. Here, we report the presence of biallelic BRCA1 mutations in a woman with multiple congenital anomalies consistent with a Fanconi anemia-like disorder and breast cancer at age 23. Patient cells exhibited deficiency in BRCA1 and RAD51 localization to DNA-damage sites, combined with radial chromosome formation and hypersensitivity to ICL-inducing agents. Restoration of these functions was achieved by ectopic introduction of a BRCA1 transgene. These observations provide evidence in support of BRCA1 as a new Fanconi anemia gene (FANCS). SIGNIFICANCE: We establish that biallelic BRCA1 mutations cause a distinct FA-S, which has implications for risk counselling in families where both parents harbor BRCA1 mutations. The genetic basis of hereditary cancer susceptibility syndromes provides diagnostic information, insights into treatment strategies, and more accurate recurrence risk counseling to families.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia/genetics , Genes, BRCA1 , Mutation , Adult , Alleles , BRCA1 Protein/genetics , Base Sequence , Fanconi Anemia Complementation Group Proteins/genetics , Female , Genetic Predisposition to Disease , Humans , Young Adult
16.
J Biol Chem ; 289(10): 7003-7010, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24451376

ABSTRACT

Fanconi anemia (FA) is a genome instability syndrome characterized by bone marrow failure and cellular hypersensitivity to DNA cross-linking agents. In response to DNA damage, the FA pathway is activated through the cooperation of 16 FA proteins. A central player in the pathway is a multisubunit E3 ubiquitin ligase complex or the FA core complex, which monoubiquitinates its substrates FANCD2 and FANCI. FANCE, a subunit of the FA core complex, plays an essential role by promoting the integrity of the complex and by directly recognizing FANCD2. To delineate its role in substrate ubiquitination from the core complex assembly, we analyzed a series of mutations within FANCE. We report that a phenylalanine located at the highly conserved extreme C terminus, referred to as Phe-522, is a critical residue for mediating the monoubiquitination of the FANCD2-FANCI complex. Using the FANCE mutant that specifically disrupts the FANCE-FANCD2 interaction as a tool, we found that the interaction-deficient mutant conferred cellular sensitivity in reconstituted FANCE-deficient cells to a similar degree as FANCE null cells, suggesting the significance of the FANCE-FANCD2 interaction in promoting cisplatin resistance. Intriguingly, ectopic expression of the FANCE C terminus fragment alone in FA normal cells disrupts DNA repair, consolidating the importance of the FANCE-FANCD2 interaction in the DNA cross-link repair.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group E Protein/metabolism , Fanconi Anemia/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group E Protein/chemistry , Fanconi Anemia Complementation Group E Protein/genetics , Fanconi Anemia Complementation Group L Protein/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine/metabolism , Protein Structure, Tertiary , Ubiquitination
17.
Proc Natl Acad Sci U S A ; 110(42): 17041-6, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24085845

ABSTRACT

Breast Cancer Type 1 Susceptibility Protein (BRCA1)-deficient cells have compromised DNA repair and are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. Despite initial responses, the development of resistance limits clinical efficacy. Mutations in the BRCA C-terminal (BRCT) domain of BRCA1 frequently create protein products unable to fold that are subject to protease-mediated degradation. Here, we show HSP90-mediated stabilization of a BRCT domain mutant BRCA1 protein under PARP inhibitor selection pressure. The stabilized mutant BRCA1 protein interacted with PALB2-BRCA2-RAD51, was essential for RAD51 focus formation, and conferred PARP inhibitor as well as cisplatin resistance. Treatment of resistant cells with the HSP90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin reduced mutant BRCA1 protein levels and restored their sensitivity to PARP inhibition. Resistant cells also acquired a TP53BP1 mutation that facilitated DNA end resection in the absence of a BRCA1 protein capable of binding CtIP. Finally, concomitant increased mutant BRCA1 and decreased 53BP1 protein expression occur in clinical samples of BRCA1-mutated recurrent ovarian carcinomas that have developed resistance to platinum. These results provide evidence for a two-event mechanism by which BRCA1-mutant tumors acquire anticancer therapy resistance.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Mutation , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , BRCA1 Protein/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Benzoquinones/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group N Protein , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Structure, Tertiary , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
Mol Cell Biol ; 33(22): 4360-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24001775

ABSTRACT

The deubiquitinating enzyme heterodimeric complex USP1-UAF1 regulates the Fanconi anemia (FA) DNA repair pathway. Absence of this complex leads to increased cellular levels of ubiquitinated FANCD2 (FANCD2-Ub) and ubiquitinated PCNA (PCNA-Ub). Mice deficient in the catalytic subunit of the complex, USP1, exhibit an FA-like phenotype and have a cellular deficiency in homologous-recombination (HR) repair. Here, we have characterized mice deficient in the UAF1 subunit. Uaf1(+/-) mice were small at birth and exhibited reduced fertility, thus resembling Usp1(-/-) mice. Unexpectedly, homozygous Uaf1(-/-) embryos died at embryonic day 7.5 (E7.5). These mutant embryos were small and developmentally retarded. As expected, Uaf1 deficiency in mice led to increased levels of cellular Fancd2-Ub and Pcna-Ub. Uaf1(+/-) murine embryonic fibroblasts (MEFs) exhibited profound chromosome instability, genotoxin hypersensitivity, and a significant defect in homologous-recombination repair. Moreover, Uaf1(-/-) mouse embryonic stem cells (mESCs) showed chromosome instability, genotoxin hypersensitivity, and impaired Fancd2 focus assembly. Similar to USP1 knockdown, UAF1 knockdown in tumor cells caused suppression of tumor growth in vivo. Taken together, our data demonstrate the important regulatory role of the USP1-UAF1 complex in HR repair through its regulation of the FANCD2-Ub and PCNA-Ub cellular pools.


Subject(s)
Embryo Loss/genetics , Gene Deletion , Homologous Recombination , Mice/embryology , Mice/genetics , Nuclear Proteins/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cells, Cultured , Chromosomal Instability , DNA Repair , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Homozygote , Humans , Infertility/genetics , Male , Mice, Inbred C57BL , Mutagens/pharmacology , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
19.
Cancer Cell ; 22(1): 117-30, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22789543

ABSTRACT

The ALK(F1174L) mutation is associated with intrinsic and acquired resistance to crizotinib and cosegregates with MYCN in neuroblastoma. In this study, we generated a mouse model overexpressing ALK(F1174L) in the neural crest. Compared to ALK(F1174L) and MYCN alone, co-expression of these two oncogenes led to the development of neuroblastomas with earlier onset, higher penetrance, and enhanced lethality. ALK(F1174L)/MYCN tumors exhibited increased MYCN dosage due to ALK(F1174L)-induced activation of the PI3K/AKT/mTOR and MAPK pathways, coupled with suppression of MYCN pro-apoptotic effects. Combined treatment with the ATP-competitive mTOR inhibitor Torin2 overcame the resistance of ALK(F1174L)/MYCN tumors to crizotinib. Our findings demonstrate a pathogenic role for ALK(F1174L) in neuroblastomas overexpressing MYCN and suggest a strategy for improving targeted therapy for ALK-positive neuroblastoma.


Subject(s)
Mutation , Neuroblastoma/genetics , Oncogenes , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Animals , Disease Models, Animal , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Signal Transduction
20.
Nat Med ; 18(7): 1118-22, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22683780

ABSTRACT

Although the treatment of acute myeloid leukemia (AML) has improved substantially in the past three decades, more than half of all patients develop disease that is refractory to intensive chemotherapy. Functional genomics approaches offer a means to discover specific molecules mediating the aberrant growth and survival of cancer cells. Thus, using a loss-of-function RNA interference genomic screen, we identified the aberrant expression of hepatocyte growth factor (HGF) as a crucial element in AML pathogenesis. We found HGF expression leading to autocrine activation of its receptor tyrosine kinase, MET, in nearly half of the AML cell lines and clinical samples we studied. Genetic depletion of HGF or MET potently inhibited the growth and survival of HGF-expressing AML cells. However, leukemic cells treated with the specific MET kinase inhibitor crizotinib developed resistance resulting from compensatory upregulation of HGF expression, leading to the restoration of MET signaling. In cases of AML where MET is coactivated with other tyrosine kinases, such as fibroblast growth factor receptor 1 (FGFR1), concomitant inhibition of FGFR1 and MET blocked this compensatory HGF upregulation, resulting in sustained logarithmic cell killing both in vitro and in xenograft models in vivo. Our results show a widespread dependence of AML cells on autocrine activation of MET, as well as the key role of compensatory upregulation of HGF expression in maintaining leukemogenic signaling by this receptor. We anticipate that these findings will lead to the design of additional strategies to block adaptive cellular responses that drive compensatory ligand expression as an essential component of the targeted inhibition of oncogenic receptors in human cancers.


Subject(s)
Autocrine Communication , Leukemia, Myeloid, Acute/enzymology , Proto-Oncogene Proteins c-met/metabolism , Animals , Autocrine Communication/drug effects , Autocrine Communication/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Crizotinib , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Immunohistochemistry , Kinetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Luminescent Measurements , Male , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Remission Induction
SELECTION OF CITATIONS
SEARCH DETAIL
...