Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Bioorg Med Chem ; 26(9): 2345-2353, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29598900

RESUMEN

REV1 protein is a mutagenic DNA damage tolerance (DDT) mediator and encodes two ubiquitin-binding motifs (i.e., UBM1 and UBM2) that are essential for the DDT function. REV1 interacts with K164-monoubiquitinated PCNA (UbPCNA) in cells upon DNA-damaging stress. By using AlphaScreen assays to detect inhibition of REV1 and UbPCNA protein interactions along with an NMR-based strategy, we identified small-molecule compounds that inhibit the REV1/UbPCNA interaction and that directly bind to REV1 UBM2. In cells, one of the compound prevented recruitment of REV1 to PCNA foci on chromatin upon cisplatin treatment, delayed removal of UV-induced cyclopyrimidine dimers from nuclei, prevented UV-induced mutation of HPRT gene, and diminished clonogenic survival of cells that were challenged by cyclophosphamide or cisplatin. This study demonstrates the potential utility of a small-molecule REV1 UBM2 inhibitor for preventing DDT.


Asunto(s)
Daño del ADN/efectos de los fármacos , ADN/química , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/metabolismo , Piperazinas/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cisplatino/farmacología , ADN/efectos de la radiación , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Lisina/química , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Proteínas Nucleares/química , Nucleotidiltransferasas/química , Piperazinas/síntesis química , Antígeno Nuclear de Célula en Proliferación/química , Unión Proteica/efectos de los fármacos , Ubiquitinación , Rayos Ultravioleta
2.
Bioorg Med Chem ; 24(5): 1071-8, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26833244

RESUMEN

DNA interstrand crosslinks (ICLs) represent physical obstacles to advancing replication forks and transcription complexes. A range of ICL-inducing agents have successfully been incorporated into cancer therapeutics. While studies have adopted UVA-activated psoralens as model ICL-inducing agents for investigating ICL repair, direct detection of the lesion has often been tempered by tagging the psoralen scaffold with a relatively large reporter group that may perturb the biological activity of the parent psoralen. Here a minimally-modified psoralen probe was prepared featuring a small alkyne handle suitable for click chemistry. The psoralen probe, designated 8-propargyloxypsoralen (8-POP), can be activated by UVA in vitro to generate ICLs that are susceptible to post-labeling with an azide-tagged fluorescent reporter via a copper-catalyzed reaction. A modified alkaline comet assay demonstrated that UVA-activated 8-POP proficiently generated ICLs in cells. Cellular 8-POP-DNA lesions were amenable to click-mediated ligation to fluorescent reporters in situ, which permitted their detection and quantitation by fluorescence microscopy and flow cytometry. Small molecule DNA repair inhibitors to 8-POP-treated cells attenuated the removal of 8-POP-DNA lesions, validating 8-POP as an appropriate probe for investigating cellular ICL repair. The post-labeling strategy applied in this study is inexpensive, rapid and highly modular in nature with the potential for multiple applications in DNA repair studies.


Asunto(s)
Química Clic , Aductos de ADN/análisis , Reparación del ADN , Ficusina/química , Colorantes Fluorescentes/química , Alquinos/química , Azidas/química , Cobre/química , Células HeLa , Humanos , Rayos Ultravioleta
3.
Bioorg Med Chem ; 24(18): 4339-4346, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27448776

RESUMEN

DNA interstrand crosslink (ICL) repair (ICLR) has been implicated in the resistance of cancer cells to ICL-inducing chemotherapeutic agents. Despite the clinical significance of ICL-inducing chemotherapy, few studies have focused on developing small-molecule inhibitors for ICLR. The mammalian DNA polymerase ζ, which comprises the catalytic subunit REV3L and the non-catalytic subunit REV7, is essential for ICLR. To identify small-molecule compounds that are mechanistically capable of inhibiting ICLR by targeting REV7, high-throughput screening and structure-activity relationship (SAR) analysis were performed. Compound 1 was identified as an inhibitor of the interaction of REV7 with the REV7-binding sequence of REV3L. Compound 7 (an optimized analog of compound 1) bound directly to REV7 in nuclear magnetic resonance analyses, and inhibited the reactivation of a reporter plasmid containing an ICL in between the promoter and reporter regions. The normalized clonogenic survival of HeLa cells treated with cisplatin and compound 7 was lower than that for cells treated with cisplatin only. These findings indicate that a small-molecule inhibitor of the REV7/REV3L interaction can chemosensitize cells by inhibiting ICLR.


Asunto(s)
Antineoplásicos/farmacología , Reparación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Mad2/antagonistas & inhibidores , Pirimidinonas/farmacología , Tiofenos/farmacología , Antineoplásicos/síntesis química , Cisplatino/farmacología , ADN Polimerasa Dirigida por ADN , Resistencia a Antineoplásicos , Células HeLa , Humanos , Unión Proteica , Pirimidinonas/síntesis química , Relación Estructura-Actividad , Tiofenos/síntesis química
4.
J Biol Chem ; 289(10): 7109-7120, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24474685

RESUMEN

Small molecule inhibitors of proliferating cell nuclear antigen (PCNA)/PCNA interacting protein box (PIP-Box) interactions, including T2 amino alcohol (T2AA), inhibit translesion DNA synthesis. The crystal structure of PCNA in complex with T2AA revealed that T2AA bound to the surface adjacent to the subunit interface of the homotrimer of PCNA in addition to the PIP-box binding cavity. Because this site is close to Lys-164, which is monoubiquitinated by RAD18, we postulated that T2AA would affect monoubiquitinated PCNA interactions. Binding of monoubiquitinated PCNA and a purified pol η fragment containing the UBZ and PIP-box was inhibited by T2AA in vitro. T2AA decreased PCNA/pol η and PCNA/REV1 chromatin colocalization but did not inhibit PCNA monoubiquitination, suggesting that T2AA hinders interactions of pol η and REV1 with monoubiquitinated PCNA. Interstrand DNA cross-links (ICLs) are repaired by mechanisms using translesion DNA synthesis that is regulated by monoubiquitinated PCNA. T2AA significantly delayed reactivation of a reporter plasmid containing an ICL. Neutral comet analysis of cells receiving T2AA in addition to cisplatin revealed that T2AA significantly enhanced formation of DNA double strand breaks (DSBs) by cisplatin. T2AA promoted colocalized foci formation of phospho-ATM and 53BP1 and up-regulated phospho-BRCA1 in cisplatin-treated cells, suggesting that T2AA increases DSBs. When cells were treated by cisplatin and T2AA, their clonogenic survival was significantly less than that of those treated by cisplatin only. These findings show that the inhibitors of monoubiquitinated PCNA chemosensitize cells by inhibiting repair of ICLs and DSBs.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/metabolismo , Éteres Fenílicos/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Propanolaminas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Células COS , Chlorocebus aethiops , Cristalografía por Rayos X , Células HeLa , Humanos , Neoplasias/genética , Éteres Fenílicos/química , Antígeno Nuclear de Célula en Proliferación/química , Propanolaminas/química
5.
Bioorg Med Chem ; 23(21): 6912-21, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26462050

RESUMEN

Cells have evolved complex biochemical pathways for DNA interstrand crosslink (ICL) removal. Despite the chemotherapeutic importance of ICL repair, there have been few attempts to identify which mechanistic DNA repair inhibitor actually inhibits ICL repair. To identify such compounds, a new and robust ICL repair assay was developed using a novel plasmid that contains synthetic ICLs between a CMV promoter region that drives transcription and a luciferase reporter gene, and an SV40 origin of replication and the large T antigen (LgT) gene that enables self-replication in mammalian cells. In a screen against compounds that are classified as inhibitors of DNA repair or synthesis, the reporter generation was exquisitely sensitive to ribonucleotide reductase (RNR) inhibitors such as gemcitabine and clofarabine, but not to inhibitors of PARP, ATR, ATM, Chk1, and others. The effect was observed also by siRNA downregulation of RNR. Moreover, the reporter generation was also particularly sensitive to 3-AP, a non-nucleoside RNR inhibitor, but not significantly sensitive to DNA replication stressors, suggesting that the involvement of RNR in ICL repair is independent of incorporation of a nucleotide RNR inhibitor into DNA to induce replication stress. The reporter generation from a modified version of the plasmid that lacks the LgT-SV40ori motif was also adversely affected by RNR inhibitors, further indicating a role for RNR in ICL repair that is independent of DNA replication. Intriguingly, unhooking of cisplatin-ICL from nuclear DNA was significantly inhibited by low doses of gemcitabine, suggesting an unidentified functional role for RNR in the process of ICL unhooking. The assay approach could identify other molecules essential for ICLR in quantitative and flexible manner.


Asunto(s)
Inhibidores Enzimáticos/química , Ribonucleótido Reductasas/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Línea Celular Tumoral , Cisplatino/química , Ensayo Cometa , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/metabolismo , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Genes Reporteros , Células HEK293 , Humanos , Plásmidos/genética , Plásmidos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/genética , Ribonucleósido Difosfato Reductasa/metabolismo , Ribonucleótido Reductasas/antagonistas & inhibidores , Ubiquitinación , Gemcitabina
6.
Bioorg Med Chem ; 22(22): 6333-43, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25438756

RESUMEN

Proliferating cell nuclear antigen (PCNA) assumes an indispensable role in supporting cellular DNA replication and repair by organizing numerous protein components of these pathways via a common PCNA-interacting sequence motif called a PIP-box. Given the multifunctional nature of PCNA, the selective inhibition of PIP-box-mediated interactions may represent a new strategy for the chemosensitization of cancer cells to existing DNA-directed therapies; however, promiscuous blockage of these interactions may also be universally deleterious. To address these possibilities, we utilized a chemical strategy to irreversibly block PIP-box-mediated interactions. Initially, we identified and validated PCNA methionine 40 (M40) and histidine 44 (H44) as essential residues for PCNA/PIP-box interactions in general and, more specifically, for efficient PCNA loading onto chromatin within cells. Next, we created a novel small molecule incorporating an electrophilic di-chloro platinum moiety that preferentially alkylated M40 and H44 residues. The compound, designated T2Pt, covalently cross-linked wild-type but not M40A/H44A PCNA, irreversibly inhibited PCNA/PIP-box interactions, and mildly alkylated plasmid DNA in vitro. In cells, T2Pt persistently induced cell cycle arrest, activated ATR-Chk1 signaling and modestly induced DNA strand breaks, features typical of cellular replication stress. Despite sustained activation of the replication stress response by the compound and its modestly genotoxic nature, T2Pt demonstrated little activity in clonogenic survival assays as a single agent, yet sensitized cells to cisplatin. The discovery of T2Pt represents an original effort directed at the development of irreversible PCNA inhibitors and sets the stage for the discovery of analogues more selective for PCNA over other cellular nucleophiles.


Asunto(s)
Compuestos Organoplatinos/química , Antígeno Nuclear de Célula en Proliferación/química , Sitios de Unión , Carbamatos/síntesis química , Carbamatos/química , Carbamatos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Polarización de Fluorescencia , Células HeLa , Humanos , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/farmacología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
7.
J Mol Biol ; 430(17): 2857-2872, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29864443

RESUMEN

REV1 is a DNA damage tolerance protein and encodes two ubiquitin-binding motifs (UBM1 and UBM2) that are essential for REV1 functions in cell survival under DNA-damaging stress. Here we report the first solution and X-ray crystal structures of REV1 UBM2 and its complex with ubiquitin, respectively. Furthermore, we have identified the first small-molecule compound, MLAF50, that directly binds to REV1 UBM2. In the heteronuclear single quantum coherence NMR experiments, peaks of UBM2 but not of UBM1 are significantly shifted by the addition of ubiquitin, which agrees to the observation that REV1 UBM2 but not UBM1 is required for DNA damage tolerance. REV1 UBM2 interacts with hydrophobic residues of ubiquitin such as L8 and L73. NMR data suggest that MLAF50 binds to the same residues of REV1 UBM2 that interact with ubiquitin, indicating that MLAF50 can compete with the REV1 UBM2-ubiquitin interaction orthosterically. Indeed, MLAF50 inhibited the interaction of REV1 UBM2 with ubiquitin and prevented chromatin localization of REV1 induced by cisplatin in U2OS cells. Our results structurally validate REV1 UBM2 as a target of a small-molecule inhibitor and demonstrate a new avenue to targeting ubiquitination-mediated protein interactions with a chemical tool.


Asunto(s)
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Éteres Fenílicos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina/química , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Cromatina/química , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Daño del ADN , Humanos , Modelos Moleculares , Proteínas Nucleares/efectos de los fármacos , Nucleotidiltransferasas/efectos de los fármacos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Unión Proteica , Conformación Proteica , Dominios Proteicos , Células Tumorales Cultivadas , Ubiquitina/efectos de los fármacos , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA