Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Blood ; 141(19): 2372-2389, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-36580665

RESUMEN

Leukemia cells accumulate DNA damage, but altered DNA repair mechanisms protect them from apoptosis. We showed here that formaldehyde generated by serine/1-carbon cycle metabolism contributed to the accumulation of toxic DNA-protein crosslinks (DPCs) in leukemia cells, especially in driver clones harboring oncogenic tyrosine kinases (OTKs: FLT3(internal tandem duplication [ITD]), JAK2(V617F), BCR-ABL1). To counteract this effect, OTKs enhanced the expression of DNA polymerase theta (POLθ) via ERK1/2 serine/threonine kinase-dependent inhibition of c-CBL E3 ligase-mediated ubiquitination of POLθ and its proteasomal degradation. Overexpression of POLθ in OTK-positive cells resulted in the efficient repair of DPC-containing DNA double-strand breaks by POLθ-mediated end-joining. The transforming activities of OTKs and other leukemia-inducing oncogenes, especially of those causing the inhibition of BRCA1/2-mediated homologous recombination with and without concomitant inhibition of DNA-PK-dependent nonhomologous end-joining, was abrogated in Polq-/- murine bone marrow cells. Genetic and pharmacological targeting of POLθ polymerase and helicase activities revealed that both activities are promising targets in leukemia cells. Moreover, OTK inhibitors or DPC-inducing drug etoposide enhanced the antileukemia effect of POLθ inhibitor in vitro and in vivo. In conclusion, we demonstrated that POLθ plays an essential role in protecting leukemia cells from metabolically induced toxic DNA lesions triggered by formaldehyde, and it can be targeted to achieve a therapeutic effect.


Asunto(s)
Proteína BRCA1 , Daño del ADN , Leucemia , Animales , Ratones , Proteína BRCA2 , ADN/metabolismo , Leucemia/enzimología , Leucemia/genética , ADN Polimerasa theta
2.
Adv Exp Med Biol ; 1360: 55-67, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35505162

RESUMEN

Growth arrest and DNA damage 45 (Gadd45) family genes, Gadd45A, Gadd45B, and GADD45 G are implicated as stress sensors that are rapidly induced upon genotoxic/physiological stress. They are involved in regulation of various cellular functions such as DNA repair, senescence, and cell cycle control. Gadd45 family of genes serve as tumor suppressors in response to different stimuli and defects in Gadd45 pathway can give rise to oncogenesis. More recently, Gadd45 has been shown to promote gene activation by demethylation and this function is important for transcriptional regulation and differentiation during development. Gadd45 serves as an adaptor for DNA repair factors to promote removal of 5-methylcytosine from DNA at gene specific loci. Therefore, Gadd45 serves as a powerful link between DNA repair and epigenetic gene regulation.


Asunto(s)
Proteínas de Ciclo Celular , Desmetilación del ADN , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Daño del ADN/genética , Reparación del ADN/genética
3.
Nature ; 510(7506): 556-9, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24776801

RESUMEN

Replication fork stalling can promote genomic instability, predisposing to cancer and other diseases. Stalled replication forks may be processed by sister chromatid recombination (SCR), generating error-free or error-prone homologous recombination (HR) outcomes. In mammalian cells, a long-standing hypothesis proposes that the major hereditary breast/ovarian cancer predisposition gene products, BRCA1 and BRCA2, control HR/SCR at stalled replication forks. Although BRCA1 and BRCA2 affect replication fork processing, direct evidence that BRCA gene products regulate homologous recombination at stalled chromosomal replication forks is lacking, due to a dearth of tools for studying this process. Here we report that the Escherichia coli Tus/Ter complex can be engineered to induce site-specific replication fork stalling and chromosomal HR/SCR in mouse cells. Tus/Ter-induced homologous recombination entails processing of bidirectionally arrested forks. We find that the Brca1 carboxy (C)-terminal tandem BRCT repeat and regions of Brca1 encoded by exon 11-two Brca1 elements implicated in tumour suppression-control Tus/Ter-induced homologous recombination. Inactivation of either Brca1 or Brca2 increases the absolute frequency of 'long-tract' gene conversions at Tus/Ter-stalled forks, an outcome not observed in response to a site-specific endonuclease-mediated chromosomal double-strand break. Therefore, homologous recombination at stalled forks is regulated differently from homologous recombination at double-strand breaks arising independently of a replication fork. We propose that aberrant long-tract homologous recombination at stalled replication forks contributes to genomic instability and breast/ovarian cancer predisposition in BRCA mutant cells.


Asunto(s)
Proteína BRCA1/metabolismo , Replicación del ADN , Proteínas de Escherichia coli/metabolismo , Recombinación Homóloga , Animales , Proteína BRCA1/química , Proteína BRCA1/genética , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Roturas del ADN de Doble Cadena , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Exones/genética , Conversión Génica/genética , Inestabilidad Genómica/genética , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Ratones
4.
J Biol Chem ; 288(10): 7086-95, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23355489

RESUMEN

Ataxia telangiectasia mutated (ATM) is activated upon DNA double strand breaks (DSBs) and phosphorylates numerous DSB response proteins, including histone H2AX on serine 139 (Ser-139) to form γ-H2AX. Through interaction with MDC1, γ-H2AX promotes DSB repair by homologous recombination (HR). H2AX Ser-139 can also be phosphorylated by DNA-dependent protein kinase catalytic subunit and ataxia telangiectasia- and Rad3-related kinase. Thus, we tested whether ATM functions in HR, particularly that controlled by γ-H2AX, by comparing HR occurring at the euchromatic ROSA26 locus between mouse embryonic stem cells lacking either ATM, H2AX, or both. We show here that loss of ATM does not impair HR, including H2AX-dependent HR, but confers sensitivity to inhibition of poly(ADP-ribose) polymerases. Loss of ATM or H2AX has independent contributions to cellular sensitivity to ionizing radiation. The ATM-independent HR function of H2AX requires both Ser-139 phosphorylation and γ-H2AX/MDC1 interaction. Our data suggest that ATM is dispensable for HR, including that controlled by H2AX, in the context of euchromatin, excluding the implication of such an HR function in genomic instability, hypersensitivity to DNA damage, and poly(ADP-ribose) polymerase inhibition associated with ATM deficiency.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Recombinación Homóloga , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Western Blotting , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cromonas/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/efectos de la radiación , Endodesoxirribonucleasas/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas/genética , Proteínas/metabolismo , ARN no Traducido , Radiación Ionizante , Serina/metabolismo , Proteínas Supresoras de Tumor/genética
5.
J Cell Sci ; 125(Pt 2): 350-61, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22331358

RESUMEN

Epithelial tissue morphogenesis is accompanied by the formation of a polarity axis--a feature of tissue architecture that is initiated by the binding of integrins to the basement membrane. Polarity plays a crucial role in tissue homeostasis, preserving differentiation, cell survival and resistance to chemotherapeutic drugs among others. An important aspect in the maintenance of tissue homeostasis is genome integrity. As normal tissues frequently experience DNA double-strand breaks (DSBs), we asked how tissue architecture might participate in the DNA damage response. Using 3D culture models that mimic mammary glandular morphogenesis and tumor formation, we show that DSB repair activity is higher in basally polarized tissues, regardless of the malignant status of cells, and is controlled by hemidesmosomal integrin signaling. In the absence of glandular morphogenesis, in 2D flat monolayer cultures, basal polarity does not affect DNA repair activity but enhances H2AX phosphorylation, an early chromatin response to DNA damage. The nuclear mitotic apparatus protein 1 (NuMA), which controls breast glandular morphogenesis by acting on the organization of chromatin, displays a polarity-dependent pattern and redistributes in the cell nucleus of basally polarized cells upon the induction of DSBs. This is shown using high-content analysis of nuclear morphometric descriptors. Furthermore, silencing NuMA impairs H2AX phosphorylation--thus, tissue polarity and NuMA cooperate to maintain genome integrity.


Asunto(s)
Antígenos Nucleares/fisiología , Reparación del ADN , Morfogénesis , Proteínas Asociadas a Matriz Nuclear/fisiología , Células Acinares/metabolismo , Antígenos Nucleares/metabolismo , Membrana Basal/metabolismo , Mama/citología , Técnicas de Cultivo de Célula , Proteínas de Ciclo Celular , Línea Celular , Línea Celular Tumoral , Polaridad Celular , Roturas del ADN de Doble Cadena , Células Epiteliales , Epitelio/crecimiento & desarrollo , Femenino , Histonas/metabolismo , Humanos , Proteínas Asociadas a Matriz Nuclear/metabolismo
6.
Nat Commun ; 15(1): 2862, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580648

RESUMEN

The DNA damage response (DDR) protein DNA Polymerase θ (Polθ) is synthetic lethal with homologous recombination (HR) factors and is therefore a promising drug target in BRCA1/2 mutant cancers. We discover an allosteric Polθ inhibitor (Polθi) class with 4-6 nM IC50 that selectively kills HR-deficient cells and acts synergistically with PARP inhibitors (PARPi) in multiple genetic backgrounds. X-ray crystallography and biochemistry reveal that Polθi selectively inhibits Polθ polymerase (Polθ-pol) in the closed conformation on B-form DNA/DNA via an induced fit mechanism. In contrast, Polθi fails to inhibit Polθ-pol catalytic activity on A-form DNA/RNA in which the enzyme binds in the open configuration. Remarkably, Polθi binding to the Polθ-pol:DNA/DNA closed complex traps the polymerase on DNA for more than forty minutes which elucidates the inhibitory mechanism of action. These data reveal a unique small-molecule DNA polymerase:DNA trapping mechanism that induces synthetic lethality in HR-deficient cells and potentiates the activity of PARPi.


Asunto(s)
Proteína BRCA1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteína BRCA1/genética , Proteína BRCA2/genética , ADN/metabolismo , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Recombinación Homóloga , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Humanos
7.
Nat Struct Mol Biol ; 30(1): 107-114, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36536104

RESUMEN

The double-strand break (DSB) repair pathway called microhomology-mediated end-joining (MMEJ) is thought to be dependent on DNA polymerase theta (Polθ) and occur independently of nonhomologous end-joining (NHEJ) factors. An unresolved question is whether MMEJ is facilitated by a single Polθ-mediated end-joining pathway or consists of additional undiscovered pathways. We find that human X-family Polλ, which functions in NHEJ, additionally exhibits robust MMEJ activity like Polθ. Polλ promotes MMEJ in mammalian cells independently of essential NHEJ factors LIG4/XRCC4 and Polθ, which reveals a distinct Polλ-dependent MMEJ mechanism. X-ray crystallography employing in situ photo-induced DSB formation captured Polλ in the act of stabilizing a microhomology-mediated DNA synapse with incoming nucleotide at 2.0 Å resolution and reveals how Polλ performs replication across a DNA synapse joined by minimal base-pairing. Last, we find that Polλ is semisynthetic lethal with BRCA1 and BRCA2. Together, these studies indicate Polλ MMEJ as a distinct DSB repair mechanism.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Animales , Humanos , Reparación del ADN por Unión de Extremidades , ADN , Mamíferos
8.
Mol Cancer Ther ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38064712

RESUMEN

Anticancer nucleosides are effective against solid tumors and hematological malignancies, but typically are prone to nucleoside metabolism resistance mechanisms. Using a nucleoside-specific multiplexed high-throughput screening approach, we discovered 4'-ethynyl-2'-deoxycytidine (EdC) as a third-generation anticancer nucleoside prodrug with preferential activity against diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukemia (ALL). EdC requires deoxycytidine kinase (DCK) phosphorylation for its activity and induced replication fork arrest and accumulation of cells in S-phase, indicating it acts as a chain terminator. A 2.1Å co-crystal structure of DCK bound to EdC and UDP reveals how the rigid 4'-alkyne of EdC fits within the active site of DCK. Remarkably, EdC was resistant to cytidine deamination and SAMHD1 metabolism mechanisms and exhibited higher potency against ALL compared to FDA approved nelarabine. Finally, EdC was highly effective against DLBCL tumors and B-ALL in vivo. These data characterize EdC as a pre-clinical nucleoside prodrug candidate for DLBCL and ALL.

9.
Breast Cancer Res ; 13(5): 314, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21996371

RESUMEN

The hereditary breast and ovarian cancer predisposition genes BRCA1 and BRCA2 account for the lion's share of heritable breast cancer risk in the human population. Loss of function of either gene results in defective homologous recombination (HR) and triggers genomic instability, accelerating breast tumorigenesis. A long-standing hypothesis proposes that BRCA1 and BRCA2 mediate HR following attempted replication across damaged DNA, ensuring error-free processing of the stalled replication fork. A recent paper describes a new replication fork protective function of BRCA2, which appears to collaborate with its HR function to suppress genomic instability.


Asunto(s)
Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Replicación del ADN , Recombinación Homóloga , Proteína BRCA1/metabolismo , Replicación del ADN/efectos de los fármacos , Femenino , Inestabilidad Genómica , Humanos , Hidroxiurea/farmacología , Mutación
10.
Cell Rep ; 34(10): 108820, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691100

RESUMEN

DNA polymerase θ (Polθ) confers resistance to chemotherapy agents that cause DNA-protein crosslinks (DPCs) at double-strand breaks (DSBs), such as topoisomerase inhibitors. This suggests Polθ might facilitate DPC repair by microhomology-mediated end-joining (MMEJ). Here, we investigate Polθ repair of DSBs carrying DPCs by monitoring MMEJ in Xenopus egg extracts. MMEJ in extracts is dependent on Polθ, exhibits the MMEJ repair signature, and efficiently repairs 5' terminal DPCs independently of non-homologous end-joining and the replisome. We demonstrate that Polθ promotes the repair of 5' terminal DPCs in mammalian cells by using an MMEJ reporter and find that Polθ confers resistance to formaldehyde in addition to topoisomerase inhibitors. Dual deficiency in Polθ and tyrosyl-DNA phosphodiesterase 2 (TDP2) causes severe cellular sensitivity to etoposide, which demonstrates MMEJ as an independent DPC repair pathway. These studies recapitulate MMEJ in vitro and elucidate how Polθ confers resistance to etoposide.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , Animales , Línea Celular , ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/deficiencia , ADN Polimerasa Dirigida por ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Formaldehído/farmacología , Humanos , Ratones , Óvulo/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Xenopus/crecimiento & desarrollo , Xenopus/metabolismo , ADN Polimerasa theta
11.
Sci Adv ; 7(24)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117057

RESUMEN

Genome-embedded ribonucleotides arrest replicative DNA polymerases (Pols) and cause DNA breaks. Whether mammalian DNA repair Pols efficiently use template ribonucleotides and promote RNA-templated DNA repair synthesis remains unknown. We find that human Polθ reverse transcribes RNA, similar to retroviral reverse transcriptases (RTs). Polθ exhibits a significantly higher velocity and fidelity of deoxyribonucleotide incorporation on RNA versus DNA. The 3.2-Šcrystal structure of Polθ on a DNA/RNA primer-template with bound deoxyribonucleotide reveals that the enzyme undergoes a major structural transformation within the thumb subdomain to accommodate A-form DNA/RNA and forms multiple hydrogen bonds with template ribose 2'-hydroxyl groups like retroviral RTs. Last, we find that Polθ promotes RNA-templated DNA repair in mammalian cells. These findings suggest that Polθ was selected to accommodate template ribonucleotides during DNA repair.


Asunto(s)
ADN Polimerasa Dirigida por ADN , ARN , Animales , ADN/química , Reparación del ADN , ADN Polimerasa Dirigida por ADN/química , Desoxirribonucleótidos , Humanos , Mamíferos/genética , Ribonucleótidos
12.
Cancer Res ; 80(14): 3033-3045, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32193285

RESUMEN

PARP inhibitor monotherapies are effective to treat patients with breast, ovary, prostate, and pancreatic cancer with BRCA1 mutations, but not to the much more frequent BRCA wild-type cancers. Searching for strategies that would extend the use of PARP inhibitors to BRCA1-proficient tumors, we found that the stability of BRCA1 protein following ionizing radiation (IR) is maintained by postphosphorylational prolyl-isomerization adjacent to Ser1191 of BRCA1, catalyzed by prolyl-isomerase Pin1. Extinction of Pin1 decreased homologous recombination (HR) to the level of BRCA1-deficient cells. Pin1 stabilizes BRCA1 by preventing ubiquitination of Lys1037 of BRCA1. Loss of Pin1, or introduction of a BRCA1-mutant refractory to Pin1 binding, decreased the ability of BRCA1 to localize to repair foci and augmented IR-induced DNA damage. In vitro growth of HR-proficient breast, prostate, and pancreatic cancer cells were modestly repressed by olaparib or Pin1 inhibition using all-trans retinoic acid (ATRA), while combination treatment resulted in near-complete block of cell proliferation. In MDA-MB-231 xenografts and triple-negative breast cancer patient-derived xenografts, either loss of Pin1 or ATRA treatment reduced BRCA1 expression and sensitized breast tumors to olaparib. Together, our study reveals that Pin1 inhibition, with clinical widely used ATRA, acts as an effective HR disrupter that sensitizes BRCA1-proficient tumors to PARP inhibition. SIGNIFICANCE: PARP inhibitors have been limited to treat homologous recombination-deficient tumors. All-trans retinoic acid, by inhibiting Pin1 and destabilizing BRCA1, extends benefit of PARP inhibitors to patients with homologous recombination-proficient tumors.See related commentary by Cai, p. 2977.


Asunto(s)
Inhibidores de Poli(ADP-Ribosa) Polimerasas , Neoplasias de la Mama Triple Negativas , Proteína BRCA1/genética , Línea Celular Tumoral , Femenino , Humanos , Masculino , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Isomerasa de Peptidilprolil , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
14.
Nat Commun ; 10(1): 4423, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562312

RESUMEN

DNA polymerase θ (Polθ) is a unique polymerase-helicase fusion protein that promotes microhomology-mediated end-joining (MMEJ) of DNA double-strand breaks (DSBs). How full-length human Polθ performs MMEJ at the molecular level remains unknown. Using a biochemical approach, we find that the helicase is essential for Polθ MMEJ of long ssDNA overhangs which model resected DSBs. Remarkably, Polθ MMEJ of ssDNA overhangs requires polymerase-helicase attachment, but not the disordered central domain, and occurs independently of helicase ATPase activity. Using single-particle microscopy and biophysical methods, we find that polymerase-helicase attachment promotes multimeric gel-like Polθ complexes that facilitate DNA accumulation, DNA synapsis, and MMEJ. We further find that the central domain regulates Polθ multimerization and governs its DNA substrate requirements for MMEJ. These studies identify unexpected functions for the helicase and central domain and demonstrate the importance of polymerase-helicase tethering in MMEJ and the structural organization of Polθ.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/fisiología , ADN Helicasas/metabolismo , ADN de Cadena Simple/metabolismo , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/metabolismo , Dominio Catalítico , Roturas del ADN , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , ADN Polimerasa theta
15.
Nat Commun ; 9(1): 1091, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29545568

RESUMEN

Genetic studies in yeast indicate that RNA transcripts facilitate homology-directed DNA repair in a manner that is dependent on RAD52. The molecular basis for so-called RNA-DNA repair, however, remains unknown. Using reconstitution assays, we demonstrate that RAD52 directly cooperates with RNA as a sequence-directed ribonucleoprotein complex to promote two related modes of RNA-DNA repair. In a RNA-bridging mechanism, RAD52 assembles recombinant RNA-DNA hybrids that coordinate synapsis and ligation of homologous DNA breaks. In an RNA-templated mechanism, RAD52-mediated RNA-DNA hybrids enable reverse transcription-dependent RNA-to-DNA sequence transfer at DNA breaks that licenses subsequent DNA recombination. Notably, we show that both mechanisms of RNA-DNA repair are promoted by transcription of a homologous DNA template in trans. In summary, these data elucidate how RNA transcripts cooperate with RAD52 to coordinate homology-directed DNA recombination and repair in the absence of a DNA donor, and demonstrate a direct role for transcription in RNA-DNA repair.


Asunto(s)
Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Reparación del ADN/fisiología , ARN/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Reparación del ADN por Recombinación/genética , Reparación del ADN por Recombinación/fisiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Nat Struct Mol Biol ; 22(3): 230-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25643323

RESUMEN

Microhomology-mediated end-joining (MMEJ) is an error-prone alternative double-strand break-repair pathway that uses sequence microhomology to recombine broken DNA. Although MMEJ has been implicated in cancer development, the mechanism of this pathway is unknown. We demonstrate that purified human DNA polymerase θ (Polθ) performs MMEJ of DNA containing 3' single-strand DNA overhangs with ≥2 bp of homology, including DNA modeled after telomeres, and show that MMEJ is dependent on Polθ in human cells. Our data support a mechanism whereby Polθ facilitates end-joining and microhomology annealing, then uses the opposing overhang as a template in trans to stabilize the DNA synapse. Polθ exhibits a preference for DNA containing a 5'-terminal phosphate, similarly to polymerases involved in nonhomologous end-joining. Finally, we identify a conserved loop domain that is essential for MMEJ and higher-order structures of Polθ that probably promote DNA synapse formation.


Asunto(s)
Reparación del ADN por Unión de Extremidades/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Modelos Genéticos , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , ADN Polimerasa Dirigida por ADN/química , Humanos , Modelos Moleculares , ADN Polimerasa theta
18.
Chem Biol ; 22(11): 1491-1504, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26548611

RESUMEN

Suppression of RAD52 causes synthetic lethality in BRCA-deficient cells. Yet pharmacological inhibition of RAD52, which binds single-strand DNA (ssDNA) and lacks enzymatic activity, has not been demonstrated. Here, we identify the small molecule 6-hydroxy-DL-dopa (6-OH-dopa) as a major allosteric inhibitor of the RAD52 ssDNA binding domain. For example, we find that multiple small molecules bind to and completely transform RAD52 undecamer rings into dimers, which abolishes the ssDNA binding channel observed in crystal structures. 6-OH-Dopa also disrupts RAD52 heptamer and undecamer ring superstructures, and suppresses RAD52 recruitment and recombination activity in cells with negligible effects on other double-strand break repair pathways. Importantly, we show that 6-OH-dopa selectively inhibits the proliferation of BRCA-deficient cancer cells, including those obtained from leukemia patients. Taken together, these data demonstrate small-molecule disruption of RAD52 rings as a promising mechanism for precision medicine in BRCA-deficient cancers.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Proteína Recombinante y Reparadora de ADN Rad52/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Regulación Alostérica , Apoptosis/efectos de los fármacos , Proteína BRCA1/deficiencia , Proteína BRCA2/deficiencia , Línea Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Dihidroxifenilalanina/análogos & derivados , Dihidroxifenilalanina/química , Dihidroxifenilalanina/metabolismo , Dihidroxifenilalanina/toxicidad , Ensayo de Cambio de Movilidad Electroforética , Humanos , Concentración 50 Inhibidora , Microscopía Fluorescente , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/toxicidad
19.
Nat Commun ; 6: 8325, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26446488

RESUMEN

Leukemia/lymphoma-related factor (LRF) is a POZ/BTB and Krüppel (POK) transcriptional repressor characterized by context-dependent key roles in cell fate decision and tumorigenesis. Here we demonstrate an unexpected transcription-independent function for LRF in the classical non-homologous end joining (cNHEJ) pathway of double-strand break (DSB) repair. We find that LRF loss in cell lines and mouse tissues results in defective cNHEJ, genomic instability and hypersensitivity to ionizing radiation. Mechanistically, we show that LRF binds and stabilizes DNA-PKcs on DSBs, in turn favouring DNA-PK activity. Importantly, LRF loss restores ionizing radiation sensitivity to p53 null cells, making LRF an attractive biomarker to direct p53-null LRF-deficient tumours towards therapeutic treatments based on genotoxic agents or PARP inhibitors following a synthetic lethal strategy.


Asunto(s)
Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Genoma , Ratones , Análisis por Matrices de Proteínas , Factores de Transcripción/genética
20.
Nat Commun ; 4: 2404, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23994874

RESUMEN

BRCA1 controls early steps of the synthesis-dependent strand annealing (SDSA) pathway of homologous recombination, but has no known role following Rad51-mediated synapsis. Here we show that BRCA1 influences post-synaptic homologous recombination events, controlling the balance between short- (STGC) and long-tract gene conversion (LTGC) between sister chromatids. Brca1 mutant cells reveal a bias towards LTGC that is corrected by expression of wild-type but not cancer-predisposing BRCA1 alleles. The LTGC bias is enhanced by depletion of CtIP but reversed by inhibition of 53BP1, implicating DNA end resection as a contributor to the STGC/LTGC balance. The impact of BRCA1/CtIP loss on the STGC/LTGC balance is abolished when the second (non-invading) end of the break is unable to support termination of STGC by homologous pairing (annealing). This suggests that BRCA1/CtIP-mediated processing of the second end of the break controls the annealing step that normally terminates SDSA, thereby suppressing the error-prone LTGC outcome.


Asunto(s)
Proteína BRCA1/metabolismo , Proteínas Portadoras/metabolismo , Cromátides/genética , Conversión Génica , Proteínas Nucleares/metabolismo , Alelos , Intercambio Genético , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Endodesoxirribonucleasas , Citometría de Flujo , Genes Reporteros , Prueba de Complementación Genética , Humanos , Modelos Biológicos , Mutación/genética , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA