Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Cancer Res ; 81(24): 6171-6182, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34548335

RESUMEN

The BRCA1 tumor suppressor gene encodes a multidomain protein for which several functions have been described. These include a key role in homologous recombination repair (HRR) of DNA double-strand breaks, which is shared with two other high-risk hereditary breast cancer suppressors, BRCA2 and PALB2. Although both BRCA1 and BRCA2 interact with PALB2, BRCA1 missense variants affecting its PALB2-interacting coiled-coil domain are considered variants of uncertain clinical significance (VUS). Using genetically engineered mice, we show here that a BRCA1 coiled-coil domain VUS, Brca1 p.L1363P, disrupts the interaction with PALB2 and leads to embryonic lethality. Brca1 p.L1363P led to a similar acceleration in the development of Trp53-deficient mammary tumors as Brca1 loss, but the tumors showed distinct histopathologic features, with more stable DNA copy number profiles in Brca1 p.L1363P tumors. Nevertheless, Brca1 p.L1363P mammary tumors were HRR incompetent and responsive to cisplatin and PARP inhibition. Overall, these results provide the first direct evidence that a BRCA1 missense variant outside of the RING and BRCT domains increases the risk of breast cancer. SIGNIFICANCE: These findings reveal the importance of a patient-derived BRCA1 coiled-coil domain sequence variant in embryonic development, mammary tumor suppression, and therapy response.See related commentary by Mishra et al., p. 6080.


Asunto(s)
Proteína BRCA1/fisiología , Proteína del Grupo de Complementación N de la Anemia de Fanconi/fisiología , Regulación Neoplásica de la Expresión Génica , Recombinación Homóloga , Neoplasias Mamarias Animales/patología , Reparación del ADN por Recombinación , Animales , Apoptosis , Proteína BRCA2/fisiología , Proliferación Celular , Femenino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
2.
Genes (Basel) ; 12(8)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34440403

RESUMEN

The tumor suppressor BRCA2 functions as a central caretaker of genome stability, and individuals who carry BRCA2 mutations are predisposed to breast, ovarian, and other cancers. Recent research advanced our mechanistic understanding of BRCA2 and its various interaction partners in DNA repair, DNA replication support, and DNA double-strand break repair pathway choice. In this review, we discuss the biochemical and structural properties of BRCA2 and examine how these fundamental properties contribute to DNA repair and replication fork stabilization in living cells. We highlight selected BRCA2 binding partners and discuss their role in BRCA2-mediated homologous recombination and fork protection. Improved mechanistic understanding of how BRCA2 functions in genome stability maintenance can enable experimental evidence-based evaluation of pathogenic BRCA2 mutations and BRCA2 pseudo-revertants to support targeted therapy.


Asunto(s)
Proteína BRCA2/fisiología , Proteína BRCA2/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Inestabilidad Genómica , Recombinación Homóloga , Humanos , Mutación
3.
Nucleic Acids Res ; 49(13): e74, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-33877327

RESUMEN

Double strand break (DSB) repair primarily occurs through 3 pathways: non-homologous end-joining (NHEJ), alternative end-joining (Alt-EJ), and homologous recombination (HR). Typical methods to measure pathway usage include integrated cassette reporter assays or visualization of DNA damage induced nuclear foci. It is now well understood that repair of Cas9-induced breaks also involves NHEJ, Alt-EJ, and HR pathways, providing a new format to measure pathway usage. Here, we have developed a simple Cas9-based system with validated repair outcomes that accurately represent each pathway and then converted it to a droplet digital PCR (ddPCR) readout, thus obviating the need for Next Generation Sequencing and bioinformatic analysis with the goal to make Cas9-based system accessible to more laboratories. The assay system has reproduced several important insights. First, absence of the key Alt-EJ factor Pol θ only abrogates ∼50% of total Alt-EJ. Second, single-strand templated repair (SSTR) requires BRCA1 and MRE11 activity, but not BRCA2, establishing that SSTR commonly used in genome editing is not conventional HR. Third, BRCA1 promotes Alt-EJ usage at two-ended DSBs in contrast to BRCA2. This assay can be used in any system, which permits Cas9 delivery and, importantly, allows rapid genotype-to-phenotype correlation in isogenic cell line pairs.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Reacción en Cadena de la Polimerasa , Reparación del ADN por Recombinación , Proteína BRCA1/fisiología , Proteína BRCA2/fisiología , Proteína 9 Asociada a CRISPR , Línea Celular , Roturas del ADN de Doble Cadena , Sitios Genéticos , Humanos , Transfección
4.
Genes (Basel) ; 11(12)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371494

RESUMEN

Fanconi anemia (FA), a chromosomal instability syndrome, is caused by inherited pathogenic variants in any of 22 FANC genes, which cooperate in the FA/BRCA pathway. This pathway regulates the repair of DNA interstrand crosslinks (ICLs) through homologous recombination. In FA proper repair of ICLs is impaired and accumulation of toxic DNA double strand breaks occurs. To repair this type of DNA damage, FA cells activate alternative error-prone DNA repair pathways, which may lead to the formation of gross structural chromosome aberrations of which radial figures are the hallmark of FA, and their segregation during cell division are the origin of subsequent aberrations such as translocations, dicentrics and acentric fragments. The deficiency in DNA repair has pleiotropic consequences in the phenotype of patients with FA, including developmental alterations, bone marrow failure and an extreme risk to develop cancer. The mechanisms leading to the physical abnormalities during embryonic development have not been clearly elucidated, however FA has features of premature aging with chronic inflammation mediated by pro-inflammatory cytokines, which results in tissue attrition, selection of malignant clones and cancer onset. Moreover, chromosomal instability and cell death are not exclusive of the somatic compartment, they also affect germinal cells, as evidenced by the infertility observed in patients with FA.


Asunto(s)
Inestabilidad Cromosómica , Reparación del ADN , Anemia de Fanconi/genética , Envejecimiento/genética , Proteína BRCA1/fisiología , Proteína BRCA2/fisiología , Trastornos de Fallo de la Médula Ósea/etiología , Ciclo Celular , Cromátides/ultraestructura , Aberraciones Cromosómicas , Cromosomas Humanos/ultraestructura , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Anemia de Fanconi/complicaciones , Anemia de Fanconi/diagnóstico , Proteína del Grupo de Complementación C de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/fisiología , Humanos , Infertilidad/genética , Síndromes Neoplásicos Hereditarios/genética , Fenotipo , Procesamiento Proteico-Postraduccional , Ubiquitinación
5.
Nucleic Acids Res ; 48(10): 5467-5484, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32329774

RESUMEN

Transcription-replication (T-R) conflicts are profound threats to genome integrity. However, whilst much is known about the existence of T-R conflicts, our understanding of the genetic and temporal nature of how cells respond to them is poorly established. Here, we address this by characterizing the early cellular response to transient T-R conflicts (TRe). This response specifically requires the DNA recombination repair proteins BLM and BRCA2 as well as a non-canonical monoubiquitylation-independent function of FANCD2. A hallmark of the TRe response is the rapid co-localization of these three DNA repair factors at sites of T-R collisions. We find that the TRe response relies on basal activity of the ATR kinase, yet it does not lead to hyperactivation of this key checkpoint protein. Furthermore, specific abrogation of the TRe response leads to DNA damage in mitosis, and promotes chromosome instability and cell death. Collectively our findings identify a new role for these well-established tumor suppressor proteins at an early stage of the cellular response to conflicts between DNA transcription and replication.


Asunto(s)
Replicación del ADN , Reparación del ADN por Recombinación , Transcripción Genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA2/fisiología , Línea Celular , Supervivencia Celular , Quinasa 9 Dependiente de la Ciclina/metabolismo , ADN/metabolismo , Daño del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Humanos , Mitosis/genética , Regiones Promotoras Genéticas , ARN/metabolismo , ARN Polimerasa II/metabolismo , Empalme del ARN , RecQ Helicasas/fisiología , Ubiquitinación
6.
Mol Genet Genomic Med ; 7(9): e879, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31317679

RESUMEN

BACKGROUND: BRCA1/2 pathogenic variants have become associated with familial breast and ovarian cancers, and hereditary cancer-predisposition syndrome. With advances in molecular biology, BRCA profiling facilitates early diagnosis and the implementation of preventive and therapeutic strategies. The genes exhibit variable prevalence among different individuals and moderate interpretation complexity. BRCA deficiency is instrumental in cancer development, affects therapeutic options and is instrumental in drug resistance. In addition, BRCA1/2 profile is diverse across different groups and has been associated with the "founder effect" in certain populations. METHODS: In this review, we aim to detail the spectrum of BRCA1/2 variants and their associated risk estimates. RESULTS: The relationship between BRCA1/2 and hereditary and familial cancers is indisputable, yet BRCA screening methods are beset with limitations and lack clinical confidence. CONCLUSION: This review emphasizes the importance of screening BRCA genetics, in addition to their clinical utility. Furthermore, founder variants are anticipated in the Saudi population.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA1/fisiología , Proteína BRCA2/genética , Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Neoplasias Ováricas/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Carcinogénesis/genética , Detección Precoz del Cáncer , Femenino , Efecto Fundador , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Epidemiología Molecular , Síndromes Neoplásicos Hereditarios/genética , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/terapia , Arabia Saudita
7.
Nucleic Acids Res ; 47(13): 6796-6810, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31114910

RESUMEN

Stabilization of stalled replication forks prevents excessive fork reversal or degradation, which can undermine genome integrity. The WRN protein is unique among the other human RecQ family members to possess exonuclease activity. However, the biological role of the WRN exonuclease is poorly defined. Recently, the WRN exonuclease has been linked to protection of stalled forks from degradation. Alternative processing of perturbed forks has been associated to chemoresistance of BRCA-deficient cancer cells. Thus, we used WRN exonuclease-deficiency as a model to investigate the fate of perturbed forks undergoing degradation, but in a BRCA wild-type condition. We find that, upon treatment with clinically-relevant nanomolar doses of the Topoisomerase I inhibitor camptothecin, loss of WRN exonuclease stimulates fork inactivation and accumulation of parental gaps, which engages RAD51. Such mechanism affects reinforcement of CHK1 phosphorylation and causes persistence of RAD51 during recovery from treatment. Notably, in WRN exonuclease-deficient cells, persistence of RAD51 correlates with elevated mitotic phosphorylation of MUS81 at Ser87, which is essential to prevent excessive mitotic abnormalities. Altogether, these findings indicate that aberrant fork degradation, in the presence of a wild-type RAD51 axis, stimulates RAD51-mediated post-replicative repair and engagement of the MUS81 complex to limit genome instability and cell death.


Asunto(s)
Camptotecina/farmacología , Replicación del ADN/efectos de los fármacos , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/fisiología , Endonucleasas/fisiología , Conformación de Ácido Nucleico/efectos de los fármacos , Recombinasa Rad51/fisiología , Inhibidores de Topoisomerasa I/farmacología , Helicasa del Síndrome de Werner/deficiencia , Proteína BRCA2/fisiología , Línea Celular Transformada , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Roturas del ADN de Doble Cadena , Activación Enzimática , Fibroblastos , Humanos , Mitocondrias/efectos de los fármacos , Mitosis/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner/fisiología
8.
Int J Nanomedicine ; 13: 8063-8074, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555227

RESUMEN

BACKGROUND: PARP inhibitors, such as Olaparib, have advanced the treatment of ovarian cancer by providing patients with an effective and molecularly-targeted maintenance therapy. However, all orally-administered drugs, including Olaparib, must undergo first-pass metabolism. In contrast, a nanoparticle delivery system has the advantage of administering Olaparib directly into the peritoneal cavity for local treatment. Consequently, we sought to optimize the sustained-release formulation NanoOlaparib, previously deemed effective as an intravenous solid tumor treatment, for the local treatment of disseminated disease via intraperitoneal (i.p.) therapy. METHODS: The tumor cell line 404, which was derived from a Brca2 -/-, Tp53 -/-, Pten -/- genetically engineered mouse model, exhibited high sensitivity to Olaparib in vitro. It was chosen for use in developing an i.p. spread xenograft for testing nanotherapy efficacy in vivo. NanoOlaparib as a monotherapy or in combination with cisplatin was compared to oral Olaparib alone or in combination using two different dose schedules. A pilot biodistribution study was performed to determine drug accumulation in various organs following i.p. administration. RESULTS: Daily administration of NanoOlaparib reduced tumor growth and decreased the variability of the treatment response observed with daily oral Olaparib administration. However, systemic toxicity was observed in both the NanoOlaparib and vehicle (empty nanoparticle) treated groups. Scaling back the administration to twice weekly was well tolerated up to 100 mg/kg but reduced the effect on tumor growth. Biodistribution profiles indicated that NanoOlaparib began accumulating in tissues within an hour of administration and persisted for at least 72 hours after a single dose, exiting the peritoneal cavity faster than expected. CONCLUSION: NanoOlaparib must be modified for use against disseminated disease. Future avenues to develop NanoOlaparib as an i.p. therapy include a modified surface-coating to retain it in the peritoneal cavity and prevent entry into systemic circulation, in addition to targeting moieties for localization in tumor cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Nanopartículas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Peritoneales/tratamiento farmacológico , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proteína BRCA2/fisiología , Femenino , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Terapia Molecular Dirigida , Nanopartículas/química , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfohidrolasa PTEN/fisiología , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario , Ftalazinas/farmacocinética , Ftalazinas/farmacología , Piperazinas/farmacocinética , Piperazinas/farmacología , Distribución Tisular , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
10.
N Engl J Med ; 379(11): 1042-1049, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30207912

RESUMEN

The causes of ovarian dysgenesis remain incompletely understood. Two sisters with XX ovarian dysgenesis carried compound heterozygous truncating mutations in the BRCA2 gene that led to reduced BRCA2 protein levels and an impaired response to DNA damage, which resulted in chromosomal breakage and the failure of RAD51 to be recruited to double-stranded DNA breaks. The sisters also had microcephaly, and one sister was in long-term remission from leukemia, which had been diagnosed when she was 5 years old. Drosophila mutants that were null for an orthologue of BRCA2 were sterile, and gonadal dysgenesis was present in both sexes. These results revealed a new role for BRCA2 and highlight the importance to ovarian development of genes that are critical for recombination during meiosis. (Funded by the Israel Science Foundation and others.).


Asunto(s)
Proteína BRCA2/deficiencia , Rotura Cromosómica , Reparación del ADN , Genes BRCA2 , Disgenesia Gonadal/genética , Ovario/crecimiento & desarrollo , Adolescente , Animales , Proteína BRCA2/fisiología , Rotura Cromosómica/efectos de los fármacos , Análisis Mutacional de ADN , Drosophila melanogaster , Femenino , Humanos , Hipogonadismo/genética , Masculino , Microcefalia/genética , Mitomicina/farmacología , Modelos Animales , Ovario/fisiología , Linaje , Hermanos , Adulto Joven
11.
Proc Natl Acad Sci U S A ; 115(31): E7379-E7388, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30018062

RESUMEN

The precise correction of genetic mutations at the nucleotide level is an attractive permanent therapeutic strategy for human disease. However, despite significant progress, challenges to efficient and accurate genome editing persist. Here, we report a genome editing platform based upon a class of hematopoietic stem cell (HSC)-derived clade F adeno-associated virus (AAV), which does not require prior nuclease-mediated DNA breaks and functions exclusively through BRCA2-dependent homologous recombination. Genome editing is guided by complementary homology arms and is highly accurate and seamless, with no evidence of on-target mutations, including insertion/deletions or inclusion of AAV inverted terminal repeats. Efficient genome editing was demonstrated at different loci within the human genome, including a safe harbor locus, AAVS1, and the therapeutically relevant IL2RG gene, and at the murine Rosa26 locus. HSC-derived AAV vector (AAVHSC)-mediated genome editing was robust in primary human cells, including CD34+ cells, adult liver, hepatic endothelial cells, and myocytes. Importantly, high-efficiency gene editing was achieved in vivo upon a single i.v. injection of AAVHSC editing vectors in mice. Thus, clade F AAV-mediated genome editing represents a promising, highly efficient, precise, single-component approach that enables the development of therapeutic in vivo genome editing for the treatment of a multitude of human gene-based diseases.


Asunto(s)
Dependovirus/genética , Edición Génica , Células Madre Hematopoyéticas/metabolismo , Recombinación Homóloga , Proteína BRCA2/fisiología , Vectores Genéticos , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Células K562
12.
Nat Commun ; 9(1): 537, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416040

RESUMEN

BRCA2 is essential for maintaining genomic integrity. BRCA2-deficient primary cells are either not viable or exhibit severe proliferation defects. Yet, BRCA2 deficiency contributes to tumorigenesis. It is believed that mutations in genes such as TRP53 allow BRCA2 heterozygous cells to overcome growth arrest when they undergo loss of heterozygosity. Here, we report the use of an insertional mutagenesis screen to identify a role for BRE (Brain and Reproductive organ Expressed, also known as BRCC45), known to be a part of the BRCA1-DNA damage sensing complex, in the survival of BRCA2-deficient mouse ES cells. Cell viability by BRE overexpression is mediated by deregulation of CDC25A phosphatase, a key cell cycle regulator and an oncogene. We show that BRE facilitates deubiquitylation of CDC25A by recruiting ubiquitin-specific-processing protease 7 (USP7) in the presence of DNA damage. Additionally, we uncovered the role of CDC25A in BRCA-mediated tumorigenesis, which can have implications in cancer treatment.


Asunto(s)
Daño del ADN , Proteínas del Tejido Nervioso/fisiología , Peptidasa Específica de Ubiquitina 7/metabolismo , Fosfatasas cdc25/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA1/fisiología , Proteína BRCA2/genética , Proteína BRCA2/fisiología , Carcinogénesis , Supervivencia Celular/fisiología , Células Cultivadas , Estabilidad de Enzimas , Heterocigoto , Humanos , Pérdida de Heterocigocidad , Células MCF-7 , Ratones , Ratones Noqueados , Mutagénesis Insercional , Procesamiento Proteico-Postraduccional , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
13.
PLoS Genet ; 14(2): e1007225, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447171

RESUMEN

The cohesin complex topologically encircles chromosomes and mediates sister chromatid cohesion to ensure accurate chromosome segregation upon cell division. Cohesin also participates in DNA repair and gene transcription. The Nipped-B-Mau2 protein complex loads cohesin onto chromosomes and the Pds5-Wapl complex removes cohesin. Pds5 is also essential for sister chromatid cohesion, indicating that it has functions beyond cohesin removal. The Brca2 DNA repair protein interacts with Pds5, but the roles of this complex beyond DNA repair are unknown. Here we show that Brca2 opposes Pds5 function in sister chromatid cohesion by assaying precocious sister chromatid separation in metaphase spreads of cultured cells depleted for these proteins. By genome-wide chromatin immunoprecipitation we find that Pds5 facilitates SA cohesin subunit association with DNA replication origins and that Brca2 inhibits SA binding, mirroring their effects on sister chromatid cohesion. Cohesin binding is maximal at replication origins and extends outward to occupy active genes and regulatory sequences. Pds5 and Wapl, but not Brca2, limit the distance that cohesin extends from origins, thereby determining which active genes, enhancers and silencers bind cohesin. Using RNA-seq we find that Brca2, Pds5 and Wapl influence the expression of most genes sensitive to Nipped-B and cohesin, largely in the same direction. These findings demonstrate that Brca2 regulates sister chromatid cohesion and gene expression in addition to its canonical role in DNA repair and expand the known functions of accessory proteins in cohesin's diverse functions.


Asunto(s)
Proteína BRCA2/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/fisiología , Segregación Cromosómica/genética , Proteínas de Drosophila/fisiología , Animales , Animales Modificados Genéticamente , Proteína BRCA2/genética , Proteínas de Ciclo Celular/fisiología , Células Cultivadas , Cromátides/genética , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/genética , Reparación del ADN/genética , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Embrión no Mamífero , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Unión Proteica , Cohesinas
14.
Nucleic Acids Res ; 46(5): 2398-2416, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29309696

RESUMEN

RAD51 recombinase assembles on single-stranded (ss)DNA substrates exposed by DNA end-resection to initiate homologous recombination (HR), a process fundamental to genome integrity. RAD51 assembly has been characterized using purified proteins, but its ultrastructural topography in the cell nucleus is unexplored. Here, we combine cell genetics with single-molecule localization microscopy and a palette of bespoke analytical tools, to visualize molecular transactions during RAD51 assembly in the cellular milieu at resolutions approaching 30-40 nm. In several human cell types, RAD51 focalizes in clusters that progressively extend into long filaments, which abut-but do not overlap-with globular bundles of replication protein A (RPA). Extended filaments alter topographically over time, suggestive of succeeding steps in HR. In cells depleted of the tumor suppressor protein BRCA2, or overexpressing its RAD51-binding BRC repeats, RAD51 fails to assemble at damage sites, although RPA accumulates unhindered. By contrast, in cells lacking a BRCA2 carboxyl (C)-terminal region targeted by cancer-causing mutations, damage-induced RAD51 assemblies initiate but do not extend into filaments. We suggest a model wherein RAD51 assembly proceeds concurrently with end-resection at adjacent sites, via an initiation step dependent on the BRC repeats, followed by filament extension through the C-terminal region of BRCA2.


Asunto(s)
Daño del ADN , Recombinasa Rad51/metabolismo , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Proteína BRCA2/fisiología , Línea Celular , Reparación del ADN , ADN de Cadena Simple , Células HeLa , Humanos , Cinética , Microscopía , Proteína de Replicación A/metabolismo
15.
Cancer Res ; 76(20): 6054-6065, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27550454

RESUMEN

Unrepaired DNA double-strand breaks (DSB) are the most destructive chromosomal lesions driving genomic instability, a core hallmark of cancer. Here, we identify the antioncogenic breast cancer factor INT6/EIF3E as an essential regulator of DSB repair that promotes homologous recombination (HR)-mediated repair and, to a lesser extent, nonhomologous end-joining repair. INT6 silencing impaired the accrual of the ubiquitin ligase RNF8 at DSBs and the formation of ubiquitin conjugates at DSB sites, especially Lys63-linked polyubiquitin chains, resulting in impaired recruitment of BRCA1, BRCA2, and RAD51, which are all involved in HR repair. In contrast, INT6 deficiency did not affect the accumulation of RNF168, 53BP1, or RPA at DSBs. In INT6-silenced cells, there was also an alteration in DNA damage-induced localization of MDC1, a key target for ATM phosphorylation, which is a prerequisite for RNF8 recruitment. The attenuated DNA damage localization of RNF8 resulting from INT6 depletion could be attributed to the defective retention of ATM previously reported by us. Our findings deepen insights into how INT6 protects against breast cancer by showing how it functions in DSB repair, with potential clinical implications for cancer therapy. Cancer Res; 76(20); 6054-65. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Factor 3 de Iniciación Eucariótica/fisiología , Ubiquitinación , Proteínas Adaptadoras Transductoras de Señales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA2/fisiología , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular , Femenino , Células HeLa , Recombinación Homóloga , Humanos , Proteínas Nucleares/metabolismo , Recombinasa Rad51/metabolismo , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas
16.
Endocr Relat Cancer ; 23(10): T1-T17, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27530658

RESUMEN

Maintaining genomic integrity is essential to preserve normal cellular physiology and to prevent the emergence of several human pathologies including cancer. The breast cancer susceptibility gene 2 (BRCA2, also known as the Fanconi anemia (FA) complementation group D1 (FANCD1)) is a potent tumor suppressor that has been extensively studied in DNA double-stranded break (DSB) repair by homologous recombination (HR). However, BRCA2 participates in numerous other processes central to maintaining genome stability, including DNA replication, telomere homeostasis and cell cycle progression. Consequently, inherited mutations in BRCA2 are associated with an increased risk of breast, ovarian and pancreatic cancers. Furthermore, bi-allelic mutations in BRCA2 are linked to FA, a rare chromosome instability syndrome characterized by aplastic anemia in children as well as susceptibility to leukemia and cancer. Here, we discuss the recent developments underlying the functions of BRCA2 in the maintenance of genomic integrity. The current model places BRCA2 as a central regulator of genome stability by repairing DSBs and limiting replication stress. These findings have direct implications for the development of novel anticancer therapeutic approaches.


Asunto(s)
Proteína BRCA2/fisiología , Reparación del ADN/genética , Replicación del ADN/genética , Inestabilidad Genómica/genética , Animales , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Humanos , Origen de Réplica/genética
17.
Arch Gynecol Obstet ; 294(6): 1311-1316, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27535760

RESUMEN

AIM: Breast cancer 2, early onset (BRCA2) has been reported to be associated with familial breast and ovarian cancer. Several proteins interact with conserved regions of BRCA2, which play significant roles in DNA damage repair and centrosomal localization. This study was aimed to identify a novel protein, Wilms tumor 1 interacting protein (WTIP), which might interact with the conserved regions of BRCA2, as well as the functional role of silencing of WTIP in response to centrosomal localization. MATERIALS AND METHODS: Hela S3 cells were used in our study. A yeast two-hybrid screening was used to identify a novel BRCA2-interacting protein. Coimmunoprecipitation and glutathione S-transferase (GST) pull-down assays were performed to detect protein-protein interaction between BRCA2 and hemaglutinin (HA)-WTIP. The expression of WTIP was silenced by short hairpin RNA (shRNA) and the levels of WTIP were confirmed by Western blot. Immunofluorescence microscopy was performed to study the centrosome localization. The functional role of knocking down WTIP expression in response to centrosomal localization was then investigated. RESULTS: The results showed that there was an interaction between WTIP and BRCA2 (amino acids 2750-2864) in Hela S3 cells. We found that WTIP interacted with BRCA2 in both exogenous and endogenous level. The expression levels of WTIP were significantly decreased by siRNA compared to the control group. Downregulation of WTIP abolished BRCA2 centrosome localization and abnormal cell division. CONCLUSION: This study indicates that WTIP interacts with BRCA2 and might be responsible for BRCA2 centrosome localization in cervical cancer cell.


Asunto(s)
Proteína BRCA2/fisiología , Proteínas Portadoras/fisiología , Centrosoma/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Proteína BRCA2/análisis , Proteínas Co-Represoras , Proteínas del Citoesqueleto , Femenino , Células HeLa , Humanos , ARN Interferente Pequeño/genética
18.
Cancer Res ; 76(20): 6118-6129, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27530326

RESUMEN

There is a need for transplantable murine models of ovarian high-grade serous carcinoma (HGSC) with regard to mutations in the human disease to assist investigations of the relationships between tumor genotype, chemotherapy response, and immune microenvironment. In addressing this need, we performed whole-exome sequencing of ID8, the most widely used transplantable model of ovarian cancer, covering 194,000 exomes at a mean depth of 400× with 90% exons sequenced >50×. We found no functional mutations in genes characteristic of HGSC (Trp53, Brca1, Brca2, Nf1, and Rb1), and p53 remained transcriptionally active. Homologous recombination in ID8 remained intact in functional assays. Further, we found no mutations typical of clear cell carcinoma (Arid1a, Pik3ca), low-grade serous carcinoma (Braf), endometrioid (Ctnnb1), or mucinous (Kras) carcinomas. Using CRISPR/Cas9 gene editing, we modeled HGSC by generating novel ID8 derivatives that harbored single (Trp53-/-) or double (Trp53-/-;Brca2-/-) suppressor gene deletions. In these mutants, loss of p53 alone was sufficient to increase the growth rate of orthotopic tumors with significant effects observed on the immune microenvironment. Specifically, p53 loss increased expression of the myeloid attractant CCL2 and promoted the infiltration of immunosuppressive myeloid cell populations into primary tumors and their ascites. In Trp53-/-;Brca2-/- mutant cells, we documented a relative increase in sensitivity to the PARP inhibitor rucaparib and slower orthotopic tumor growth compared with Trp53-/- cells, with an appearance of intratumoral tertiary lymphoid structures rich in CD3+ T cells. This work validates new CRISPR-generated models of HGSC to investigate its biology and promote mechanism-based therapeutics discovery. Cancer Res; 76(20); 6118-29. ©2016 AACR.


Asunto(s)
Proteína BRCA2/fisiología , Sistemas CRISPR-Cas/fisiología , Cistadenocarcinoma Seroso/etiología , Modelos Animales de Enfermedad , Neoplasias Ováricas/etiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Proteína BRCA2/genética , Línea Celular Tumoral , Cistadenocarcinoma Seroso/tratamiento farmacológico , Exoma , Femenino , Edición Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética
19.
Pathol Biol (Paris) ; 63(4-5): 185-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26320393

RESUMEN

BACKGROUND: Breast cancer is increasing among young women in Tunisia. Germline mutations in the BRCA1/2 genes are associated with a high risk for breast cancer development. However, the true contribution of BRCA1/2 mutation in sporadic breast cancer is not well documented. Our aim is to identify the BRCA2 mutation spectrum in Tunisian young women with breast cancer. METHODS: Screening the BRCA2 gene was performed using DHPLC, DNA sequencing and PCR-RFLP. RESULTS: We identified, in a woman diagnosed with early onset breast cancer, and without family history, a novel in frame deletion 5456delGTAGCA in the exon 11 of the BRCA2 gene which causes a loss of two residues Ser1743-Ser1744. The absence of this deletion in the patients' parents suggests that it is a de novo variant. Furthermore, we screened 108 sporadic cases, 50 familial cases, and 60 controls for the identified del6bp using PCR-RFLP. None of them carried this deletion suggesting that this variant is not a benign polymorphism and probably rare in our population. With regards to the position of the Ser1743-1744 in the BRCT domain, sequence alignment revealed that the Ser1743 is conserved among several species, which may reflect its importance in the BRCA2 function. A modeling of the wild-type and mutated BRC5-BRC6 domain revealed that the deletion of the 2 Serine residues might affect the structure of this BRCA2 domain. CONCLUSIONS: A novel in frame deletion 5456del6bp in BRCA2 gene was identified in an early onset woman with breast cancer and without family history.


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA2 , Eliminación de Secuencia , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Proteína BRCA2/química , Proteína BRCA2/fisiología , Neoplasias de la Mama/epidemiología , Secuencia Conservada , Exones/genética , Femenino , Pruebas Genéticas , Humanos , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Conformación Proteica , Estructura Terciaria de Proteína , Sistemas de Lectura/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serina/química , Especificidad de la Especie , Túnez/epidemiología , Adulto Joven
20.
Biochem J ; 460(3): 331-42, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24870022

RESUMEN

PALB2 [partner and localizer of BRCA2 (breast cancer early-onset 2)] [corrected] has emerged as a key player in the maintenance of genome integrity. Biallelic mutations in PALB2 cause FA (Fanconi's anaemia) subtype FA-N, a devastating inherited disorder marked by developmental abnormalities, bone marrow failure and childhood cancer susceptibility, whereas monoallelic mutations predispose to breast, ovarian and pancreatic cancer. The tumour suppressor role of PALB2 has been intimately linked to its ability to promote HR (homologous recombination)-mediated repair of DNA double-strand breaks. Because PALB2 lies at the crossroads between FA, HR and cancer susceptibility, understanding its function has become the primary focus of several studies. The present review discusses a current synthesis of the contribution of PALB2 to these pathways. We also provide a molecular description of FA- or cancer-associated PALB2 mutations.


Asunto(s)
Reparación del ADN , Recombinación Homóloga , Neoplasias/fisiopatología , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama Masculina/genética , Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Humanos , Masculino , Ratones , Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias Pancreáticas/genética , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...