Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 178(1): 152-159.e11, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31178121

RESUMEN

Intrinsic and acquired drug resistance and induction of secondary malignancies limit successful chemotherapy. Because mutagenic translesion synthesis (TLS) contributes to chemoresistance as well as treatment-induced mutations, targeting TLS is an attractive avenue for improving chemotherapeutics. However, development of small molecules with high specificity and in vivo efficacy for mutagenic TLS has been challenging. Here, we report the discovery of a small-molecule inhibitor, JH-RE-06, that disrupts mutagenic TLS by preventing recruitment of mutagenic POL ζ. Remarkably, JH-RE-06 targets a nearly featureless surface of REV1 that interacts with the REV7 subunit of POL ζ. Binding of JH-RE-06 induces REV1 dimerization, which blocks the REV1-REV7 interaction and POL ζ recruitment. JH-RE-06 inhibits mutagenic TLS and enhances cisplatin-induced toxicity in cultured human and mouse cell lines. Co-administration of JH-RE-06 with cisplatin suppresses the growth of xenograft human melanomas in mice, establishing a framework for developing TLS inhibitors as a novel class of chemotherapy adjuvants.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Mutagénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Quinolinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/efectos adversos , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Mad2/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/química , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Quinolinas/química , Quinolinas/farmacología , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Biol Chem ; 299(2): 102859, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36592930

RESUMEN

Translesion synthesis (TLS) DNA polymerase Polζ is crucial for the bypass replication over sites of DNA damage. The Rev7 subunit of Polζ is a HORMA (Hop1, Rev7, Mad2) protein that facilitates recruitment of Polζ to the replication fork via interactions with the catalytic subunit Rev3 and the translesion synthesis scaffold protein Rev1. Human Rev7 (hRev7) interacts with two Rev7-binding motifs (RBMs) of hRev3 by a mechanism conserved among HORMA proteins whereby the safety-belt loop of hRev7 closes on the top of the ligand. The two copies of hRev7 tethered by the two hRev3-RBMs form a symmetric head-to-head dimer through the canonical HORMA dimerization interface. Recent cryo-EM structures reveal that Saccharomyces cerevisiae Polζ (scPolζ) also includes two copies of scRev7 bound to distinct regions of scRev3. Surprisingly, the HORMA dimerization interface is not conserved in scRev7, with the two scRev7 protomers forming an asymmetric head-to-tail dimer with a much smaller interface than the hRev7 dimer. Here, we validated the two adjacent RBM motifs in scRev3, which bind scRev7 with affinities that differ by two orders of magnitude and confirmed the 2:1 stoichiometry of the scRev7:Rev3 complex in solution. However, our biophysical studies reveal that scRev7 does not form dimers in solution either on its own accord or when tethered by the two RBMs in scRev3. These findings imply that the scRev7 dimer observed in the cryo-EM structures is induced by scRev7 interactions with other Polζ subunits and that Rev7 homodimerization via the HORMA interface is a mechanism that emerged later in evolution.


Asunto(s)
Replicación del ADN , ADN Polimerasa Dirigida por ADN , Humanos , Daño del ADN , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas Mad2/química , Proteínas Mad2/metabolismo , Nucleotidiltransferasas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
3.
Bioorg Med Chem ; 106: 117755, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38749343

RESUMEN

Translesion synthesis (TLS) is a cellular mechanism through which actively replicating cells recruit specialized, low-fidelity DNA polymerases to damaged DNA to allow for replication past these lesions. REV1 is one of these TLS DNA polymerases that functions primarily as a scaffolding protein to organize the TLS heteroprotein complex and ensure replication occurs in the presence of DNA lesions. The C-Terminal domain of REV1 (REV1-CT) forms many protein-protein interactions (PPIs) with other TLS polymerases, making it essential for TLS function and a promising drug target for anti-cancer drug development. We utilized several lead identification strategies to identify various small molecules capable of disrupting the PPI between REV1-CT and the REV1 Interacting Regions (RIR) present in several other TLS polymerases. These lead compounds were profiled in several in vitro potency and PK assays to identify two scaffolds (1 and 6) as the most promising for further development. Both 1 and 6 synergized with cisplatin in a REV1-dependent fashion and demonstrated promising in vivo PK and toxicity profiles.


Asunto(s)
Nucleotidiltransferasas , Bibliotecas de Moléculas Pequeñas , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/metabolismo , Humanos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/síntesis química , Animales , Relación Estructura-Actividad , Unión Proteica , Estructura Molecular , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , ADN Polimerasa Dirigida por ADN/metabolismo , Ratones , Síntesis Translesional de ADN
4.
Histochem Cell Biol ; 159(2): 119-125, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36260111

RESUMEN

Quantitative analysis of microscopy images from samples stained with fluorescent probes necessitates a very low fluorescence background signal. In tissues prepared by immersion in a chemical fixative, followed by conventional processing for paraffin embedding, red blood cell autofluorescence across several imaging channels can be a nuisance. Although many protocols have been proposed to suppress red blood cell autofluorescence prior to microscopy imaging, in many instances they may not prove totally effective. Moreover, in environments such as core facilities where control over tissue processing and staining may not be feasible, methods to address autofluorescence via post-image acquisition processing may be of some advantage. To this end, we have developed an image analysis algorithm using a commercially based software platform to remove contaminating red blood cell autofluorescence during quantitative evaluation of the fluorescence signal from an immunostaining protocol. The method is based upon the low autofluorescence signal of red blood cells exhibited in the blue channel (used to detect DAPI nuclear signal of all cells), which can be subtracted from the total channel signal by increasing the threshold for DAPI signal in the nuclear detection settings during nuclear segmentation. With the contributing signal from the red blood cells eliminated, the specific immunostained signal for the antigen of interest could be determined. We believe that this simple algorithm performed on post-acquisition microscopy images will be of use for quantitative fluorescence analyses whenever red blood cell autofluorescence is present, especially in amounts where creating regions of interest for evaluation is not possible.


Asunto(s)
Eritrocitos , Colorantes Fluorescentes , Microscopía Fluorescente , Coloración y Etiquetado , Procesamiento de Imagen Asistido por Computador
5.
Proc Natl Acad Sci U S A ; 117(46): 28918-28921, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33168727

RESUMEN

REV1/POLζ-dependent mutagenic translesion synthesis (TLS) promotes cell survival after DNA damage but is responsible for most of the resulting mutations. A novel inhibitor of this pathway, JH-RE-06, promotes cisplatin efficacy in cancer cells and mouse xenograft models, but the mechanism underlying this combinatorial effect is not known. We report that, unexpectedly, in two different mouse xenograft models and four human and mouse cell lines we examined in vitro cisplatin/JH-RE-06 treatment does not increase apoptosis. Rather, it increases hallmarks of senescence such as senescence-associated ß-galactosidase, increased p21 expression, micronuclei formation, reduced Lamin B1, and increased expression of the immune regulators IL6 and IL8 followed by cell death. Moreover, although p-γ-H2AX foci formation was elevated and ATR expression was low in single agent cisplatin-treated cells, the opposite was true in cells treated with cisplatin/JH-RE-06. These observations suggest that targeting REV1 with JH-RE-06 profoundly affects the nature of the persistent genomic damage after cisplatin treatment and also the resulting physiological responses. These data highlight the potential of REV1/POLζ inhibitors to alter the biological response to DNA-damaging chemotherapy and enhance the efficacy of chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Nitroquinolinas/farmacología , Nucleotidiltransferasas/antagonistas & inhibidores , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Línea Celular Tumoral , Cisplatino/administración & dosificación , Cisplatino/farmacología , ADN/biosíntesis , Daño del ADN/fisiología , Reparación del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Humanos , Proteínas Mad2/metabolismo , Ratones , Mutagénesis , Neoplasias/enzimología , Neoplasias/patología , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Biochem Biophys Res Commun ; 579: 141-145, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34600299

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus responsible for the current COVID-19 pandemic and has now infected more than 200 million people with more than 4 million deaths globally. Recent data suggest that symptoms and general malaise may continue long after the infection has ended in recovered patients, suggesting that SARS-CoV-2 infection has profound consequences in the host cells. Here we report that SARS-CoV-2 infection can trigger a DNA damage response (DDR) in African green monkey kidney cells (Vero E6). We observed a transcriptional upregulation of the Ataxia telangiectasia and Rad3 related protein (ATR) in infected cells. In addition, we observed enhanced phosphorylation of CHK1, a downstream effector of the ATR DNA damage response, as well as H2AX. Strikingly, SARS-CoV-2 infection lowered the expression of TRF2 shelterin-protein complex, and reduced telomere lengths in infected Vero E6 cells. Thus, our observations suggest SARS-CoV-2 may have pathological consequences to host cells beyond evoking an immunopathogenic immune response.


Asunto(s)
COVID-19/genética , Daño del ADN , Interacciones Huésped-Patógeno/genética , SARS-CoV-2/patogenicidad , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Chlorocebus aethiops , Histonas/genética , Fosforilación , Telómero , Células Vero
7.
Proc Natl Acad Sci U S A ; 115(35): E8191-E8200, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30111544

RESUMEN

The translesion synthesis (TLS) polymerases Polζ and Rev1 form a complex that enables replication of damaged DNA. The Rev7 subunit of Polζ, which is a multifaceted HORMA (Hop1, Rev7, Mad2) protein with roles in TLS, DNA repair, and cell-cycle control, facilitates assembly of this complex by binding Rev1 and the catalytic subunit of Polζ, Rev3. Rev7 interacts with Rev3 by a mechanism conserved among HORMA proteins, whereby an open-to-closed transition locks the ligand underneath the "safety belt" loop. Dimerization of HORMA proteins promotes binding and release of this ligand, as exemplified by the Rev7 homolog, Mad2. Here, we investigate the dimerization of Rev7 when bound to the two Rev7-binding motifs (RBMs) in Rev3 by combining in vitro analyses of Rev7 structure and interactions with a functional assay in a Rev7-/- cell line. We demonstrate that Rev7 uses the conventional HORMA dimerization interface both to form a homodimer when tethered by the two RBMs in Rev3 and to heterodimerize with other HORMA domains, Mad2 and p31comet Structurally, the Rev7 dimer can bind only one copy of Rev1, revealing an unexpected Rev1/Polζ architecture. In cells, mutation of the Rev7 dimer interface increases sensitivity to DNA damage. These results provide insights into the structure of the Rev1/Polζ TLS assembly and highlight the function of Rev7 homo- and heterodimerization.


Asunto(s)
Proteínas Mad2 , Proteínas Nucleares , Nucleotidiltransferasas , Multimerización de Proteína , Línea Celular , Daño del ADN , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Proteínas Mad2/química , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Dominios Proteicos
8.
Proc Natl Acad Sci U S A ; 112(12): 3764-9, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25775519

RESUMEN

The dynamic mutability of microsatellite repeats is implicated in the modification of gene function and disease phenotype. Studies of the enhanced instability of long trinucleotide repeats (TNRs)-the cause of multiple human diseases-have revealed a remarkable complexity of mutagenic mechanisms. Here, we show that cold, heat, hypoxic, and oxidative stresses induce mutagenesis of a long CAG repeat tract in human cells. We show that stress-response factors mediate the stress-induced mutagenesis (SIM) of CAG repeats. We show further that SIM of CAG repeats does not involve mismatch repair, nucleotide excision repair, or transcription, processes that are known to promote TNR mutagenesis in other pathways of instability. Instead, we find that these stresses stimulate DNA rereplication, increasing the proportion of cells with >4 C-value (C) DNA content. Knockdown of the replication origin-licensing factor CDT1 eliminates both stress-induced rereplication and CAG repeat mutagenesis. In addition, direct induction of rereplication in the absence of stress also increases the proportion of cells with >4C DNA content and promotes repeat mutagenesis. Thus, environmental stress triggers a unique pathway for TNR mutagenesis that likely is mediated by DNA rereplication. This pathway may impact normal cells as they encounter stresses in their environment or during development or abnormal cells as they evolve metastatic potential.


Asunto(s)
Mutagénesis , Estrés Fisiológico , Repeticiones de Trinucleótidos , Frío , ADN/química , ADN/genética , Reparación del ADN , Replicación del ADN , Redes Reguladoras de Genes , Inestabilidad Genómica , Proteínas Fluorescentes Verdes/química , Células HEK293 , Calor , Humanos , Repeticiones de Microsatélite , Estrés Oxidativo , Fenotipo , Expansión de Repetición de Trinucleótido
10.
Biochem Biophys Res Commun ; 431(2): 270-3, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23313845

RESUMEN

DNA polymerase η mostly catalyzes an error-free bypass of the most frequent UV lesions, pyrimidine dimers of the cyclobutane-type. In addition to its nuclear localization, we show here for the first time its mitochondrial localization in budding yeast. In mitochondria, this polymerase improves bypass replication fidelity opposite UV damage as shown in base pair substitution and frameshift assays. For base pair substitutions, polymerase η appears to be related in function and epistatic to DNA polymerase ζ which, however, plays the opposite role in the nucleus.


Asunto(s)
Reparación del ADN , ADN Mitocondrial/genética , ADN Polimerasa Dirigida por ADN/fisiología , Mitocondrias/enzimología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , ADN Mitocondrial/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Mitocondrias/genética , Mutagénesis , Dímeros de Pirimidina/genética , Saccharomyces cerevisiae/efectos de la radiación , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Rayos Ultravioleta
11.
Res Sq ; 2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35441168

RESUMEN

The repertoire of coronavirus disease 2019 (COVID-19)-mediated adverse health outcomes has continued to expand in infected patients, including the susceptibility to developing long-COVID; however, the molecular underpinnings at the cellular level are poorly defined. In this study, we report that SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection triggers host cell genome instability by modulating the expression of molecules of DNA repair and mutagenic translesion synthesis. Further, SARS-CoV-2 infection causes genetic alterations, such as increased mutagenesis, telomere dysregulation, and elevated microsatellite instability (MSI). The MSI phenotype was coupled to reduced MLH1, MSH6, and MSH2 in infected cells. Strikingly, pre-treatment of cells with the REV1-targeting translesion DNA synthesis inhibitor, JH-RE-06, suppresses SARS-CoV-2 proliferation and dramatically represses the SARS-CoV-2-dependent genome instability. Mechanistically, JH-RE-06 treatment induces autophagy, which we hypothesize limits SARS-CoV-2 proliferation and, therefore, the hijacking of host-cell genome instability pathways. These results have implications for understanding the pathobiological consequences of COVID-19.

12.
Cancers (Basel) ; 13(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-34771454

RESUMEN

Cancer therapy resistance is a persistent clinical challenge. Recently, inhibition of the mutagenic translesion synthesis (TLS) protein REV1 was shown to enhance tumor cell response to chemotherapy by triggering senescence hallmarks. These observations suggest REV1's important role in determining cancer cell response to chemotherapy. Whether REV1 inhibition would similarly sensitize cancer cells to radiation treatment is unknown. This study reports a lack of radiosensitization in response to REV1 inhibition by small molecule inhibitors in ionizing radiation-exposed cancer cells. Instead, REV1 inhibition unexpectedly triggers autophagy, which is a known biomarker of radioresistance. We report a possible role of the REV1 TLS protein in determining cancer treatment outcomes depending upon the type of DNA damage inflicted. Furthermore, we discover that REV1 inhibition directly triggers autophagy, an uncharacterized REV1 phenotype, with a significant bearing on cancer treatment regimens.

13.
Environ Mol Mutagen ; 61(8): 830-836, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32573829

RESUMEN

Stapled α-helical RIR (Rev1-interacting region) peptides of DNA POL κ bind more effectively to the RIR-interface of the C-terminal recruitment domain of the translesion synthesis DNA polymerase Rev1 than unstapled peptide. The tightest-binding stapled peptide translocates into cells and enhances the cytotoxicity of DNA damaging agents while reducing mutagenesis. Drugs with these characteristics could potentially serve as adjuvants to improve chemotherapy and reduce acquired resistance by inhibiting Rev1-dependent mutagenic translesion synthesis.


Asunto(s)
Daño del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Mutágenos/toxicidad , Nucleotidiltransferasas/metabolismo
14.
ChemMedChem ; 14(17): 1610-1617, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31361935

RESUMEN

Translesion synthesis (TLS) has emerged as a mechanism through which several forms of cancer develop acquired resistance to first-line genotoxic chemotherapies by allowing replication to continue in the presence of damaged DNA. Small molecules that inhibit TLS hold promise as a novel class of anticancer agents that can serve to enhance the efficacy of these front-line therapies. We previously used a structure-based rational design approach to identify the phenazopyridine scaffold as an inhibitor of TLS that functions by disrupting the protein-protein interaction (PPI) between the C-terminal domain of the TLS DNA polymerase Rev1 (Rev1-CT) and the Rev1 interacting regions (RIR) of other TLS DNA polymerases. To continue the identification of small molecules that disrupt the Rev1-CT/RIR PPI, we generated a pharmacophore model based on the phenazopyridine scaffold and used it in a structure-based virtual screen. In vitro analysis of promising hits identified several new chemotypes with the ability to disrupt this key TLS PPI. In addition, several of these compounds were found to enhance the efficacy of cisplatin in cultured cells, highlighting their anti-TLS potential.


Asunto(s)
Compuestos Azo/farmacología , ADN Polimerasa Dirigida por ADN/metabolismo , Nucleotidiltransferasas/metabolismo , Unión Proteica/efectos de los fármacos , Piridinas/farmacología , Animales , ADN Polimerasa Dirigida por ADN/química , Evaluación Preclínica de Medicamentos , Ratones , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nucleotidiltransferasas/química , Dominios Proteicos
15.
Environ Mol Mutagen ; 58(5): 235-263, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28485537

RESUMEN

Living organisms are continuously exposed to a myriad of DNA damaging agents that can impact health and modulate disease-states. However, robust DNA repair and damage-bypass mechanisms faithfully protect the DNA by either removing or tolerating the damage to ensure an overall survival. Deviations in this fine-tuning are known to destabilize cellular metabolic homeostasis, as exemplified in diverse cancers where disruption or deregulation of DNA repair pathways results in genome instability. Because routinely used biological, physical and chemical agents impact human health, testing their genotoxicity and regulating their use have become important. In this introductory review, we will delineate mechanisms of DNA damage and the counteracting repair/tolerance pathways to provide insights into the molecular basis of genotoxicity in cells that lays the foundation for subsequent articles in this issue. Environ. Mol. Mutagen. 58:235-263, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN/fisiología , Mutagénesis/fisiología , Animales , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Mutagénesis/genética , Telómero/genética
16.
ACS Chem Biol ; 12(7): 1903-1912, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28541665

RESUMEN

Translesion synthesis (TLS) is an important mechanism through which proliferating cells tolerate DNA damage during replication. The mutagenic Rev1/Polζ-dependent branch of TLS helps cancer cells survive first-line genotoxic chemotherapy and introduces mutations that can contribute to the acquired resistance so often observed with standard anticancer regimens. As such, inhibition of Rev1/Polζ-dependent TLS has recently emerged as a strategy to enhance the efficacy of first-line chemotherapy and reduce the acquisition of chemoresistance by decreasing tumor mutation rate. The TLS DNA polymerase Rev1 serves as an integral scaffolding protein that mediates the assembly of the active multiprotein TLS complexes. Protein-protein interactions (PPIs) between the C-terminal domain of Rev1 (Rev1-CT) and the Rev1-interacting region (RIR) of other TLS DNA polymerases play an essential role in regulating TLS activity. To probe whether disrupting the Rev1-CT/RIR PPI is a valid approach for developing a new class of targeted anticancer agents, we designed a fluorescence polarization-based assay that was utilized in a pilot screen for small molecule inhibitors of this PPI. Two small molecule scaffolds that disrupt this interaction were identified, and secondary validation assays confirmed that compound 5 binds to Rev1-CT at the RIR interface. Finally, survival and mutagenesis assays in mouse embryonic fibroblasts and human fibrosarcoma HT1080 cells treated with cisplatin and ultraviolet light indicate that these compounds inhibit mutagenic Rev1/Polζ-dependent TLS in cells, validating the Rev1-CT/RIR PPI for future anticancer drug discovery and identifying the first small molecule inhibitors of TLS that target Rev1-CT.


Asunto(s)
ADN Polimerasa Dirigida por ADN , Sistemas de Liberación de Medicamentos , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Nucleotidiltransferasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Sitios de Unión , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Descubrimiento de Drogas , Activación Enzimática/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas Nucleares/genética , Inhibidores de la Síntesis del Ácido Nucleico/química , Nucleotidiltransferasas/genética , Proteínas/química , Bibliotecas de Moléculas Pequeñas/química
17.
J Mol Biol ; 428(15): 2978-80, 2016 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-27318194

RESUMEN

Multiple pathways modulate the dynamic mutability of trinucleotide repeats (TNRs), which are implicated in neurodegenerative disease and evolution. Recently, we reported that environmental stresses induce TNR mutagenesis via stress responses and rereplication, with more than 50% of mutants carrying deletions or insertions-molecular signatures of DNA double-strand break repair. We now show that knockdown of alt-nonhomologous end joining (alt-NHEJ) components-XRCC1, LIG3, and PARP1-suppresses stress-induced TNR mutagenesis, in contrast to the components of homologous recombination and NHEJ, which have no effect. Thus, alt-NHEJ, which contributes to genetic mutability in cancer cells, also plays a novel role in environmental stress-induced TNR mutagenesis.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Mutagénesis/genética , Estrés Fisiológico/genética , Repeticiones de Trinucleótidos/genética , Roturas del ADN de Doble Cadena , Replicación del ADN/genética , Recombinación Homóloga/genética , Humanos
18.
DNA Repair (Amst) ; 42: 26-32, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27131875

RESUMEN

Trinucleotide repeat (TNR) expansion beyond a certain threshold results in some 20 incurable neurodegenerative disorders where disease anticipation positively correlates with repeat length. Long TNRs typically display a bias toward further expansion during germinal transmission from parents to offspring, and then are highly unstable in somatic tissues of affected individuals. Understanding mechanisms of TNR instability will provide insights into disease pathogenesis. Previously, we showed that enhanced convergent transcription at long CAG repeat tracks induces TNR instability and cell death via ATR activation. Components of TC-NER (transcription-coupled nucleotide excision repair) and RNaseH enzymes that resolve RNA/DNA hybrids oppose cell death, whereas the MSH2 component of MMR (mismatch repair) enhances cell death. The exact role of the MMR pathway during convergent transcription-induced cell death at CAG repeats is not well understood. In this study, we show that siRNA knockdowns of MMR components-MSH2, MSH3, MLHI, PMS2, and PCNA-reduce DNA toxicity. Furthermore, knockdown of MSH2, MLH1, and PMS2 significantly reduces the frequency of ATR foci formation. These observations suggest that MMR proteins activate DNA toxicity by modulating ATR foci formation during convergent transcription.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Transcripción Genética/genética , Repeticiones de Trinucleótidos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Secuencia de Bases , Muerte Celular/genética , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Activación Enzimática/genética , Técnicas de Silenciamiento del Gen , Humanos , Homólogo 1 de la Proteína MutL/deficiencia , Homólogo 1 de la Proteína MutL/genética , Proteína 2 Homóloga a MutS/deficiencia , Proteína 2 Homóloga a MutS/genética , Proteína 3 Homóloga de MutS , ARN Interferente Pequeño/genética
19.
Postdoc J ; 4(5): 46-54, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27595121

RESUMEN

Almost 20 incurable neurodegenerative disorders are caused by trinucleotide repeat (TNR) expansion beyond a certain threshold, with disease time of onset and severity positively correlating with repeat length. Typically, long TNRs display a bias toward further expansion and repeats continue to expand not only during germline transmissions from parents to offspring, but also remain highly unstable in somatic tissues of patients. Hence, understanding TNR instability mechanisms sheds light on underlying disease pathology. Recently, we showed that activated ATR is the major signal for convergent-transcription-induced cell death at CAG repeats and is regulated by the mismatch repair (MMR) pathway. Additionally, components of other DNA repair pathways such as transcription-coupled nucleotide excision repair (TC-NER) and R-loop resolution by RNaseH reduce cell death. Because activated ATR signals the Fanconi anemia (FA) pathway of interstrand crosslink DNA repair, we asked whether the FA pathway also modulates convergent-transcription-induced cell death at expanded CAG repeats. We show here that siRNA knockdown of FA components-FANCI, FANCJ, FANCM, FANCA, and FANCD2-decreases cell death, suggesting that FA proteins, like MMR proteins, are activators of cell death during convergent transcription.

20.
Cold Spring Harb Perspect Biol ; 5(12): a019836, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24296172

RESUMEN

DNA damage is one of many possible perturbations that challenge the mechanisms that preserve genetic stability during the copying of the eukaryotic genome in S phase. This short review provides, in the first part, a general introduction to the topic and an overview of checkpoint responses. In the second part, the mechanisms of error-free tolerance in response to fork-arresting DNA damage will be discussed in some detail.


Asunto(s)
Daño del ADN , Reparación del ADN , Replicación del ADN , Eucariontes/fisiología , Ciclo Celular/fisiología , Eucariontes/citología , Eucariontes/metabolismo , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA