Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 596(7872): 433-437, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34321663

RESUMEN

Protein ubiquitination at sites of DNA double-strand breaks (DSBs) by RNF168 recruits BRCA1 and 53BP11,2, which are mediators of the homologous recombination and non-homologous end joining DSB repair pathways, respectively3. Non-homologous end joining relies on 53BP1 binding directly to ubiquitinated lysine 15 on H2A-type histones (H2AK15ub)4,5 (which is an RNF168-dependent modification6), but how RNF168 promotes BRCA1 recruitment and function remains unclear. Here we identify a tandem BRCT-domain-associated ubiquitin-dependent recruitment motif (BUDR) in BRCA1-associated RING domain protein 1 (BARD1) (the obligate partner protein of BRCA1) that, by engaging H2AK15ub, recruits BRCA1 to DSBs. Disruption of the BUDR of BARD1 compromises homologous recombination and renders cells hypersensitive to PARP inhibition and cisplatin. We further show that BARD1 binds nucleosomes through multivalent interactions: coordinated binding of H2AK15ub and unmethylated H4 lysine 20 by its adjacent BUDR and ankyrin repeat domains, respectively, provides high-affinity recognition of DNA lesions in replicated chromatin and promotes the homologous recombination activities of the BRCA1-BARD1 complex. Finally, our genetic epistasis experiments confirm that the need for BARD1 chromatin-binding activities can be entirely relieved upon deletion of RNF168 or 53BP1. Thus, our results demonstrate that by sensing DNA-damage-dependent and post-replication histone post-translation modification states, BRCA1-BARD1 complexes coordinate the antagonization of the 53BP1 pathway with promotion of homologous recombination, establishing a simple paradigm for the governance of the choice of DSB repair pathway.


Asunto(s)
Recombinación Homóloga , Lisina/química , Lisina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adulto , Secuencias de Aminoácidos , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Cromatina/metabolismo , Cisplatino/farmacología , Roturas del ADN de Doble Cadena , Daño del ADN/efectos de los fármacos , Femenino , Células HCT116 , Células HEK293 , Histonas/química , Histonas/metabolismo , Humanos , Masculino , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Dominios Proteicos , Reparación del ADN por Recombinación , Proteínas Supresoras de Tumor/química , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/deficiencia
2.
Nat Commun ; 12(1): 3636, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140467

RESUMEN

To identify approaches to target DNA repair vulnerabilities in cancer, we discovered nanomolar potent, selective, low molecular weight (MW), allosteric inhibitors of the polymerase function of DNA polymerase Polθ, including ART558. ART558 inhibits the major Polθ-mediated DNA repair process, Theta-Mediated End Joining, without targeting Non-Homologous End Joining. In addition, ART558 elicits DNA damage and synthetic lethality in BRCA1- or BRCA2-mutant tumour cells and enhances the effects of a PARP inhibitor. Genetic perturbation screening revealed that defects in the 53BP1/Shieldin complex, which cause PARP inhibitor resistance, result in in vitro and in vivo sensitivity to small molecule Polθ polymerase inhibitors. Mechanistically, ART558 increases biomarkers of single-stranded DNA and synthetic lethality in 53BP1-defective cells whilst the inhibition of DNA nucleases that promote end-resection reversed these effects, implicating these in the synthetic lethal mechanism-of-action. Taken together, these observations describe a drug class that elicits BRCA-gene synthetic lethality and PARP inhibitor synergy, as well as targeting a biomarker-defined mechanism of PARPi-resistance.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Reparación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/genética , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Mutaciones Letales Sintéticas/efectos de los fármacos , Regulación Alostérica , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Desoxirribonucleasas/antagonistas & inhibidores , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Recombinación Homóloga/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Ratones , Organoides/efectos de los fármacos , Neoplasias Ováricas/genética , Ratas , Mutaciones Letales Sintéticas/genética , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , ADN Polimerasa theta
3.
Cell Death Differ ; 27(9): 2552-2567, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32139898

RESUMEN

BRCA1 is critical for DNA double-strand break (DSB) repair by homologous recombination (HR). BRCA1 deficient mice are embryonic lethal. Previous studies have shown that 53BP1 knockout (KO) rescues embryonic lethality of BRCA1 hypomorphic mutant mice by restoring HR. Here, we show that 53BP1 KO can partially rescue embryonic lethality of BRCA1 total KO mice, but HR is not restored in BRCA1-53BP1 double knockout (DKO) mice. As a result, BRCA1-53BP1 DKO cells are extremely sensitive to PARP inhibitors (PARPi). In addition to HR deficiency, BRCA1-53BP1 DKO cells have elevated microhomology-mediated end joining (MMEJ) activity and G2/M cell cycle checkpoint defects, causing severe genomic instability in these cells. Interestingly, BRCA1-53BP1 DKO mice rapidly develop thymic lymphoma that is 100% penetrant, which is not observed in any BRCA1 mutant mice rescued by 53BP1 KO. Taken together, our study reveals that 53BP1 KO can partially rescue embryonic lethality caused by complete BRCA1 loss without rescuing HR-related defects. This finding suggests that loss of 53BP1 can support the development of cancers with silenced BRCA1 expression without causing PARPi resistance.


Asunto(s)
Proteína BRCA1/deficiencia , Pérdida del Embrión/genética , Inestabilidad Genómica , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Animales , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Supervivencia sin Enfermedad , Pérdida del Embrión/patología , Silenciador del Gen , Recombinación Homóloga , Humanos , Linfoma/patología , Metafase , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Dominios Proteicos , Timo/patología , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
4.
Nat Commun ; 11(1): 819, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041954

RESUMEN

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


Asunto(s)
Proteína BRCA1/deficiencia , Cromatina/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Recombinación Homóloga , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Secuencias de Aminoácidos , Proteína BRCA2/deficiencia , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/química , Proteína del Grupo de Complementación N de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Humanos , Nucleosomas/metabolismo
5.
Genes Dev ; 34(1-2): 7-23, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31896689

RESUMEN

53BP1 is an enigmatic DNA damage response factor that gained prominence because it determines the efficacy of PARP1 inhibitory drugs (PARPi) in BRCA1-deficient cancers. Recent studies have elevated 53BP1 from its modest status of (yet another) DNA damage factor to master regulator of double-strand break (DSB) repair pathway choice. Our review of the literature suggests an alternative view. We propose that 53BP1 has evolved to avoid mutagenic repair outcomes and does so by controlling the processing of DNA ends and the dynamics of DSBs. The consequences of 53BP1 deficiency, such as diminished PARPi efficacy in BRCA1-deficient cells and altered repair of damaged telomeres, can be explained from this viewpoint. We further propose that some of the fidelity functions of 53BP1 coevolved with class switch recombination (CSR) in the immune system. We speculate that, rather than being deterministic in DSB repair pathway choice, 53BP1 functions as a DSB escort that guards against illegitimate and potentially tumorigenic recombination.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Evolución Molecular , Humanos , Cambio de Clase de Inmunoglobulina/genética , Telómero/genética , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/genética
6.
Nature ; 574(7779): 571-574, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645724

RESUMEN

To safeguard genome integrity in response to DNA double-strand breaks (DSBs), mammalian cells mobilize the neighbouring chromatin to shield DNA ends against excessive resection that could undermine repair fidelity and cause damage to healthy chromosomes1. This form of genome surveillance is orchestrated by 53BP1, whose accumulation at DSBs triggers sequential recruitment of RIF1 and the shieldin-CST-POLα complex2. How this pathway reflects and influences the three-dimensional nuclear architecture is not known. Here we use super-resolution microscopy to show that 53BP1 and RIF1 form an autonomous functional module that stabilizes three-dimensional chromatin topology at sites of DNA breakage. This process is initiated by accumulation of 53BP1 at regions of compact chromatin that colocalize with topologically associating domain (TAD) sequences, followed by recruitment of RIF1 to the boundaries between such domains. The alternating distribution of 53BP1 and RIF1 stabilizes several neighbouring TAD-sized structures at a single DBS site into an ordered, circular arrangement. Depletion of 53BP1 or RIF1 (but not shieldin) disrupts this arrangement and leads to decompaction of DSB-flanking chromatin, reduction in interchromatin space, aberrant spreading of DNA repair proteins, and hyper-resection of DNA ends. Similar topological distortions are triggered by depletion of cohesin, which suggests that the maintenance of chromatin structure after DNA breakage involves basic mechanisms that shape three-dimensional nuclear organization. As topological stabilization of DSB-flanking chromatin is independent of DNA repair, we propose that, besides providing a structural scaffold to protect DNA ends against aberrant processing, 53BP1 and RIF1 safeguard epigenetic integrity at loci that are disrupted by DNA breakage.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Inestabilidad Genómica , Conformación de Ácido Nucleico , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/química , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas de Unión a Telómeros/deficiencia , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
7.
Gynecol Oncol ; 153(1): 127-134, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30686551

RESUMEN

OBJECTIVE: Poly(ADP-ribose) polymerase (PARP) inhibitors have shown substantial activity in homologous recombination- (HR-) deficient ovarian cancer and are undergoing testing in other HR-deficient tumors. For reasons that are incompletely understood, not all patients with HR-deficient cancers respond to these agents. Preclinical studies have demonstrated that changes in alternative DNA repair pathways affect PARP inhibitor (PARPi) sensitivity in ovarian cancer models. This has not previously been assessed in the clinical setting. METHODS: Clonogenic and plasmid-based HR repair assays were performed to compare BRCA1-mutant COV362 ovarian cancer cells with or without 53BP1 gene deletion. Archival biopsies from ovarian cancer patients in the phase I, open-label clinical trial of PARPi ABT-767 were stained for PARP1, RAD51, 53BP1 and multiple components of the nonhomologous end-joining (NHEJ) DNA repair pathway. Modified histochemistry- (H-) scores were determined for each repair protein in each sample. HRD score was determined from tumor DNA. RESULTS: 53BP1 deletion increased HR in BRCA1-mutant COV362 cells and decreased PARPi sensitivity in vitro. In 36 women with relapsed ovarian cancer, responses to the PARPi ABT-767 were observed exclusively in cancers with HR deficiency. In this subset, 7 of 18 patients (39%) had objective responses. The actual HRD score did not further correlate with change from baseline tumor volume (r = 0.050; p = 0.87). However, in the HR-deficient subset, decreased 53BP1 H-score was associated with decreased antitumor efficacy of ABT-767 (r = -0.69, p = 0.004). CONCLUSION: Differences in complementary repair pathways, particularly 53BP1, correlate with PARPi response of HR-deficient ovarian cancers.


Asunto(s)
Benzamidas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Sulfonamidas/administración & dosificación , Proteína 1 de Unión al Supresor Tumoral P53/genética , Línea Celular Tumoral , Reparación del ADN , Resistencia a Antineoplásicos , Femenino , Genes BRCA1 , Genes BRCA2 , Recombinación Homóloga , Humanos , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/biosíntesis , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína 1 de Unión al Supresor Tumoral P53/biosíntesis , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia
8.
Thromb Haemost ; 119(3): 439-448, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30620991

RESUMEN

The replication stress inflicted on retinal endothelial cells (ECs) in the context of hypoxia-induced pathological neovascularization during proliferative retinopathy is linked with activation of the deoxyribonucleic acid (DNA) repair response. Here, we studied the effect of deficiency of the DNA damage response adaptor 53BP1, which is an antagonist of homologous recombination (HR), in the context of proliferative retinopathy. In the model of retinopathy of prematurity (ROP), 53BP1-deficient mice displayed increased hypoxia-driven pathological neovascularization and tuft formation, accompanied by increased EC proliferation and reduced EC apoptosis, as compared with 53BP1-sufficient mice. In contrast, physiological retina angiogenesis was not affected by 53BP1 deficiency. Knockdown of 53BP1 in ECs in vitro also resulted in enhanced proliferation and reduced apoptosis of the cells under hypoxic conditions. Additionally, upon 53BP1 knockdown, ECs displayed increased HR rate in hypoxia. Consistently, treatment with an HR inhibitor reversed the hyper-proliferative angiogenic phenotype associated with 53BP1 deficiency in ROP. Thus, by unleashing HR, 53BP1 deletion increases pathological EC proliferation and neovascularization in the context of ROP. Our data shed light to a previously unknown interaction between the DNA repair response and pathological neovascularization in the retina.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Recombinación Homóloga , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Retinopatía de la Prematuridad/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Animales , Apoptosis , Hipoxia de la Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Predisposición Genética a la Enfermedad , Recombinación Homóloga/efectos de los fármacos , Humanos , Ratones Noqueados , Morfolinas/farmacología , Fenotipo , Pirroles/farmacología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , Neovascularización Retiniana/prevención & control , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/patología , Retinopatía de la Prematuridad/prevención & control , Transducción de Señal , Proteína 1 de Unión al Supresor Tumoral P53/genética
9.
Nature ; 560(7716): 122-127, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30046110

RESUMEN

53BP1 governs a specialized, context-specific branch of the classical non-homologous end joining DNA double-strand break repair pathway. Mice lacking 53bp1 (also known as Trp53bp1) are immunodeficient owing to a complete loss of immunoglobulin class-switch recombination1,2, and reduced fidelity of long-range V(D)J recombination3. The 53BP1-dependent pathway is also responsible for pathological joining events at dysfunctional telomeres4, and its unrestricted activity in Brca1-deficient cellular and tumour models causes genomic instability and oncogenesis5-7. Cells that lack core non-homologous end joining proteins are profoundly radiosensitive8, unlike 53BP1-deficient cells9,10, which suggests that 53BP1 and its co-factors act on specific DNA substrates. Here we show that 53BP1 cooperates with its downstream effector protein REV7 to promote non-homologous end joining during class-switch recombination, but REV7 is not required for 53BP1-dependent V(D)J recombination. We identify shieldin-a four-subunit putative single-stranded DNA-binding complex comprising REV7, c20orf196 (SHLD1), FAM35A (SHLD2) and FLJ26957 (SHLD3)-as the factor that explains this specificity. Shieldin is essential for REV7-dependent DNA end-protection and non-homologous end joining during class-switch recombination, and supports toxic non-homologous end joining in Brca1-deficient cells, yet is dispensable for REV7-dependent interstrand cross-link repair. The 53BP1 pathway therefore comprises distinct double-strand break repair activities within chromatin and single-stranded DNA compartments, which explains both the immunological differences between 53bp1- and Rev7- deficient mice and the context specificity of the pathway.


Asunto(s)
Reparación del ADN por Unión de Extremidades , ADN/química , ADN/metabolismo , Proteínas Mad2/metabolismo , Complejos Multiproteicos/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Cambio de Clase de Inmunoglobulina/genética , Proteínas Mad2/deficiencia , Proteínas Mad2/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/química , Mutación , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Recombinación V(D)J/genética
10.
Cell Rep ; 23(11): 3127-3136, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29898385

RESUMEN

PARP inhibitors (PARPis) have been used to induce synthetic lethality in BRCA-deficient tumors in clinical trials with limited success. We hypothesized that RAD52-mediated DNA repair remains active in PARPi-treated BRCA-deficient tumor cells and that targeting RAD52 should enhance the synthetic lethal effect of PARPi. We show that RAD52 inhibitors (RAD52is) attenuated single-strand annealing (SSA) and residual homologous recombination (HR) in BRCA-deficient cells. Simultaneous targeting of PARP1 and RAD52 with inhibitors or dominant-negative mutants caused synergistic accumulation of DSBs and eradication of BRCA-deficient but not BRCA-proficient tumor cells. Remarkably, Parp1-/-;Rad52-/- mice are normal and display prolonged latency of BRCA1-deficient leukemia compared with Parp1-/- and Rad52-/- counterparts. Finally, PARPi+RAD52i exerted synergistic activity against BRCA1-deficient tumors in immunodeficient mice with minimal toxicity to normal cells and tissues. In conclusion, our data indicate that addition of RAD52i will improve therapeutic outcome of BRCA-deficient malignancies treated with PARPi.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Animales , Proteína BRCA1/deficiencia , Proteína BRCA2/deficiencia , Reparación del ADN/efectos de los fármacos , Femenino , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Recombinación Homóloga/efectos de los fármacos , Humanos , Mesilato de Imatinib/farmacología , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/deficiencia , Proteína Recombinante y Reparadora de ADN Rad52/antagonistas & inhibidores , Proteína Recombinante y Reparadora de ADN Rad52/deficiencia , Mutaciones Letales Sintéticas , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/genética
11.
Cell Cycle ; 17(7): 881-891, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29620483

RESUMEN

'BRCAness' is a term used to describe cancer cells that behave similarly to tumors with BRCA1 or BRCA2 mutations. The BRCAness phenotype is associated with hypersensitivity to chemotherapy agents including PARP inhibitors, which are a promising class of recently-licensed anti-cancer treatments. This hypersensitivity arises because of a deficiency in the homologous recombination (HR) pathway for DNA double-strand break repair. To gain further insight into how genetic modifiers of HR contribute to the BRCAness phenotype, we created a new mouse model of BRCAness by generating mice that are deficient in BLM helicase and the Exo1 exonuclease, which are involved in the early stages of HR. We find that cells lacking BLM and Exo1 exhibit a BRCAness phenotype, with diminished HR, and hypersensitivity to PARP inhibitors. We further tested how 53BP1, an important regulator of HR, affects repair efficiency in our BRCAness model. We find that deletion of 53BP1 can relieve several of the repair deficiencies observed in cells lacking BLM and Exo1, just as it does in cells lacking BRCA1. These results substantiate the importance of BRCAness as a concept for classification of cancer cases, and further clarify the role of 53BP1 in regulation of DNA repair pathway choice in mammalian cells.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Reparación del ADN/efectos de los fármacos , Exodesoxirribonucleasas/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , RecQ Helicasas/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos B/efectos de la radiación , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Enzimas Reparadoras del ADN/deficiencia , Exodesoxirribonucleasas/deficiencia , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Rayos gamma , Eliminación de Gen , Expresión Génica , Inestabilidad Genómica , Humanos , Ratones , Ratones Noqueados , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Cultivo Primario de Células , RecQ Helicasas/deficiencia , Intercambio de Cromátides Hermanas , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia
12.
J Cell Biol ; 216(11): 3521-3534, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28912125

RESUMEN

The BLM gene product, BLM, is a RECQ helicase that is involved in DNA replication and repair of DNA double-strand breaks by the homologous recombination (HR) pathway. During HR, BLM has both pro- and anti-recombinogenic activities, either of which may contribute to maintenance of genomic integrity. We find that in cells expressing a mutant version of BRCA1, an essential HR factor, ablation of BLM rescues genomic integrity and cell survival in the presence of DNA double-strand breaks. Improved genomic integrity in these cells is linked to a substantial increase in the stability of RAD51 at DNA double-strand break sites and in the overall efficiency of HR. Ablation of BLM also rescues RAD51 foci and HR in cells lacking BRCA2 or XRCC2. These results indicate that the anti-recombinase activity of BLM is of general importance for normal retention of RAD51 at DNA break sites and regulation of HR.


Asunto(s)
Roturas del ADN de Doble Cadena , Linfocitos/enzimología , Neoplasias/enzimología , Recombinasa Rad51/metabolismo , RecQ Helicasas/metabolismo , Reparación del ADN por Recombinación , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiencia , Proteína BRCA2/genética , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Genotipo , Humanos , Linfocitos/patología , Ratones Noqueados , Mutación , Neoplasias/genética , Neoplasias/patología , Fenotipo , Estabilidad Proteica , Interferencia de ARN , Recombinasa Rad51/genética , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/genética
13.
Radiat Res ; 188(4.2): 495-504, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28741984

RESUMEN

Computed tomography (CT) scans are a routine diagnostic imaging technique that utilize low-energy X rays with an average absorbed dose of approximately 10 mGy per clinical whole-body CT scan. The growing use of CT scans in the clinic has raised concern of increased carcinogenic risk in patients exposed to ionizing radiation from diagnostic procedures. The goal of this study was to better understand cancer risk associated with low-dose exposures from CT scans. Historically, low-dose exposure preceding a larger challenge dose increases tumor latency, but does little to impact tumor frequency in Trp53+/- mice. To assess the effects of CT scans specifically on tumor progression, whole-body CT scans (10 mGy/scan, 75 kVp) were started at four weeks after 4 Gy irradiation, to allow for completion of tumor initiation. The mice were exposed to weekly CT scans for ten consecutive weeks. In this study, we show that CT scans modify cellular end points commonly associated with carcinogenesis in cancer-prone Trp53+/- heterozygous mice. At five days after completion of CT scan treatment, the multiple CT scans did not cause detectable differences in bone marrow genomic instability, as measured by the formation of micronucleated reticulocytes and H2AX phosphorylation in lymphoid-type cells, and significantly lowered constitutive and radiation induced levels of apoptosis. The overall lifespan of 4 Gy exposed cancer-initiated mice treated with multiple CT scans was increased by approximately 8% compared to mice exposed to 4 Gy alone (P < 0.017). Increased latency periods for lymphoma and sarcoma (P < 0.040) progression contributed to the overall increase in lifespan. However, repeated CT scans did not affect carcinoma latency. To our knowledge, this is the first reported study to show that repeated CT scans, when administered after tumor initiation, can improve cancer morbidity by delaying the progression of specific types of radiation-induced cancers in Trp53+/- mice.


Asunto(s)
Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/patología , Tomografía Computarizada por Rayos X/efectos adversos , Animales , Apoptosis/efectos de la radiación , Carcinogénesis/efectos de la radiación , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta en la Radiación , Femenino , Histonas/metabolismo , Masculino , Ratones , Estadificación de Neoplasias , Neoplasias Inducidas por Radiación/metabolismo , Reticulocitos/patología , Reticulocitos/efectos de la radiación , Medición de Riesgo , Análisis de Supervivencia , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
14.
Acta Pharmacol Sin ; 38(7): 1038-1047, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28414200

RESUMEN

Over half of patients with BRCA1-deficient cancers do not respond to treatment with poly(ADP-ribose) polymerase (PARP) inhibitors. In this study, we report that a combination of 53BP1 and BRCA1 may serve as a biomarker of PARP inhibitor sensitivity. Based on the mRNA levels of four homologous recombination repair (HR) genes and PARP inhibitor sensitivity, we selected BRCA1-deficient MDA-MB-436 cells to conduct RNA interference. Reducing expression of 53BP1, but not the other three HR genes, was found to lower simmiparib sensitivity. Additionally, we generated 53BP1-/-/BRCA1-/- clonal variants by the transcription activator-like effector nuclease (TALEN) technique and found that depleting 53BP1 impaired PARP inhibitor sensitivity with a 36.7-fold increase in their IC50 values. Consistent with its effect on PARP inhibitor sensitivity, 53BP1 loss alleviated cell cycle arrest and apoptosis and partially restored HR function. Importantly, 53BP1 depletion dramatically reduced the ability of PARP inhibitors to suppress tumor growth in vivo. The inhibition rate of simmiparib was 74.16% for BRCA1-deficient MDA-MB-436 xenografts, but only 7.79% for 53BP1/BRCA1-deficient xenografts. Re-expressing 53BP1 in the dual-deficient cells restored PARP inhibitor sensitivity and the levels of HR regulators. Considering that at least 10% of BRCA1-deficient breast and ovarian cancers have reduced expression of 53BP1, using a combination of 53BP1 with BRCA1 as a biomarker for patient selection should reduce the number of patients undergoing futile treatment with PARP inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/antagonistas & inhibidores , Biomarcadores de Tumor/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/antagonistas & inhibidores , Animales , Antineoplásicos/química , Proteína BRCA1/deficiencia , Proteína BRCA1/metabolismo , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Relación Estructura-Actividad , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
15.
Proc Natl Acad Sci U S A ; 114(7): E1196-E1204, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28137874

RESUMEN

Class-switch recombination (CSR) alters the Ig isotype to diversify antibody effector functions. IgD CSR is a rare event, and its regulation is poorly understood. We report that deficiency of 53BP1, a DNA damage-response protein, caused age-dependent overproduction of secreted IgD resulting from increased IgD CSR exclusively within B cells of mucosa-associated lymphoid tissues. IgD overproduction was dependent on activation-induced cytidine deaminase, hematopoietic MyD88 expression, and an intact microbiome, against which circulating IgD, but not IgM, was reactive. IgD CSR occurred via both alternative nonhomologous end-joining and homologous recombination pathways. Microbiota-dependent IgD CSR also was detected in nasal-associated lymphoid tissue of WT mice. These results identify a pathway, present in WT mice and hyperactivated in 53BP1-deficient mice, by which microbiota signal via Toll-like receptors to elicit IgD CSR.


Asunto(s)
Cambio de Clase de Inmunoglobulina , Inmunoglobulina D/inmunología , Tejido Linfoide/inmunología , Microbiota/inmunología , Membrana Mucosa/inmunología , Animales , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Reparación del ADN por Unión de Extremidades , Femenino , Inmunoglobulina D/genética , Inmunoglobulina D/metabolismo , Tejido Linfoide/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/genética , Membrana Mucosa/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Recombinación Genética , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/inmunología
16.
EMBO Rep ; 17(11): 1532-1541, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27670884

RESUMEN

BRCA1 mutations strongly predispose affected individuals to breast and ovarian cancer, but the mechanism by which BRCA1 acts as a tumor suppressor is not fully understood. Homozygous deletion of exon 2 of the mouse Brca1 gene normally causes embryonic lethality, but we show that exon 2-deleted alleles of Brca1 are expressed as a mutant isoform that lacks the N-terminal RING domain. This "RING-less" BRCA1 protein is stable and efficiently recruited to the sites of DNA damage. Surprisingly, robust RAD51 foci form in cells expressing RING-less BRCA1 in response to DNA damage, but the cells nonetheless display the substantial genomic instability. Genomic instability can be rescued by the deletion of Trp53bp1, which encodes the DNA damage response factor 53BP1, and mice expressing RING-less BRCA1 do not show an increased susceptibility to tumors in the absence of 53BP1. Genomic instability in cells expressing RING-less BRCA1 correlates with the loss of BARD1 and a defect in restart of replication forks after hydroxyurea treatment, suggesting a role of BRCA1-BARD1 in genomic integrity that is independent of RAD51 loading.


Asunto(s)
Inestabilidad Genómica , Proteínas Supresoras de Tumor/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Animales , Proteína BRCA1 , Secuencia de Bases , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN , Exones/genética , Femenino , Péptidos y Proteínas de Señalización Intracelular , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN , Eliminación de Secuencia , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
17.
Int J Oncol ; 49(4): 1600-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27499037

RESUMEN

The present study aimed to observe the influence of p53 binding protein 1 (53BP1) silencing on the radiosensitivity of colorectal cancer (CRC) cells and investigated the potential underlying mechanisms. The differences in radiosensitivity among four CRC cells were detected by the clone formation assay, while the expression of their 53BP1 was detected by the western blot analysis. HCT116 cells with relatively high expression of 53BP1 were selected to silence the expression of 53BP1 by shRNA intervention. The influence on proliferation, apoptosis, and cell cycle distribution was detected by immunofluorescent staining of Ki-67 and flow cytometry. The expression of relevant proteins in the apoptotic pathway ATM-CHK2-p53 was further analyzed by western blot analysis. The expression of 53BP1 was found to be closely related to the radiosensitivity of the CRC cells. Decreased expression of 53BP1 led to the tolerance of HCT116 cells to radiation. The detection of tumor proliferation, apoptosis, and cell cycle showed that decreased expression of 53BP1 resulted in an increased S-phase percentage of HCT116 cells, an increased proliferating rate, and a decreased apoptotic rate after radiation. The analysis of the molecular pathway showed that the reduced expression of 53BP1 decreased the protein expression of ATM, CHK2, and the phosphorylated products associated with the p53 apoptotic pathway. In conclusion, decreased expression of 53BP1 leads to radiotolerance of CRC cells, and the underlying mechanism is probably related to the decreased expression of relevant proteins in the ATM-CHK2-p53 pathway, which affects cell cycle, apoptosis and proliferation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/radioterapia , Regulación Neoplásica de la Expresión Génica , Tolerancia a Radiación , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Animales , Biomarcadores de Tumor/genética , Western Blotting , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Directa , Humanos , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...