Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167190, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38657912

RESUMEN

Cervical cancer cells possess high levels of reactive oxygen species (ROS); thus, increasing oxidative stress above the toxicity threshold to induce cell death is a promising chemotherapeutic strategy. However, the underlying mechanisms of cell death are elusive, and efficacy and toxicity issues remain. Within DNA, 8-oxo-7,8-dihydroguanine (8-oxoG) is the most frequent base lesion repaired by 8-oxoguanine glycosylase 1 (OGG1)-initiated base excision repair. Cancer cells also express high levels of MutT homolog 1 (MTH1), which prevents DNA replication-induced incorporation of 8-oxoG into the genome by hydrolyzing 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate (8-oxo-dGTP). Here, we revealed that ROS-inducing agents triggered cervical cancer to undergo parthanatos, which was mainly induced by massive DNA strand breaks resulting from overwhelming 8-oxoG excision by OGG1. Furthermore, the MTH1 inhibitor synergized with a relatively low dose of ROS-inducing agents by enhancing 8-oxoG loading in the DNA. In vivo, this drug combination suppressed the growth of tumor xenografts, and this inhibitory effect was significantly decreased in the absence of OGG1. Hence, the present study highlights the roles of base repair enzymes in cell death induction and suggests that the combination of lower doses of ROS-inducing agents with MTH1 inhibitors may be a more selective and safer strategy for cervical cancer chemotherapy.


Asunto(s)
ADN Glicosilasas , Enzimas Reparadoras del ADN , Monoéster Fosfórico Hidrolasas , Especies Reactivas de Oxígeno , Neoplasias del Cuello Uterino , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Humanos , Femenino , Especies Reactivas de Oxígeno/metabolismo , Animales , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/genética , Ratones , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/genética , Guanina/análogos & derivados , Guanina/farmacología , Línea Celular Tumoral , Reparación del ADN/efectos de los fármacos , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Sinergismo Farmacológico , Células HeLa , Estrés Oxidativo/efectos de los fármacos
2.
Expert Opin Ther Targets ; 28(3): 117-130, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38344773

RESUMEN

INTRODUCTION: Pulmonary diseases impose a daunting burden on healthcare systems and societies. Current treatment approaches primarily address symptoms, underscoring the urgency for the development of innovative pharmaceutical solutions. A noteworthy focus lies in targeting enzymes recognizing oxidatively modified DNA bases within gene regulatory elements, given their pivotal role in governing gene expression. AREAS COVERED: This review delves into the intricate interplay between the substrate-specific binding of 8-oxoguanine DNA glycosylase 1 (OGG1) and epigenetic regulation, with a focal point on elucidating the molecular underpinnings and their biological implications. The absence of OGG1 distinctly attenuates the binding of transcription factors to cis elements, thereby modulating pro-inflammatory or pro-fibrotic transcriptional activity. Through a synergy of experimental insights gained from cell culture studies and murine models, utilizing prototype OGG1 inhibitors (O8, TH5487, and SU0268), a promising panorama emerges. These investigations underscore the absence of cytotoxicity and the establishment of a favorable tolerance profile for these OGG1 inhibitors. EXPERT OPINION: Thus, the strategic targeting of the active site pocket of OGG1 through the application of small molecules introduces an innovative trajectory for advancing redox medicine. This approach holds particular significance in the context of pulmonary diseases, offering a refined avenue for their management.


Asunto(s)
ADN Glicosilasas , Enfermedades Pulmonares , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Animales , Humanos , Enfermedades Pulmonares/tratamiento farmacológico , Ratones , Epigénesis Genética , Desarrollo de Medicamentos , Terapia Molecular Dirigida , Inhibidores Enzimáticos/farmacología
3.
Chem Biol Interact ; 362: 109999, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35654123

RESUMEN

Pulmonary fibrosis is a highly aggressive and lethal disease that currently lacks effective targeting therapies. Herein, we established a mouse model of pulmonary fibrosis induced by intratracheal instillation of bleomycin (BLM) in wild-type (WT) and 8-oxoguanine DNA glycosylase-1 (OGG1) knockout (Ogg1-/-) mice. TH5487, a specific small-molecule inhibitor of OGG1, was found to ameliorate BLM-induced pulmonary fibrosis in WT mice. Concomitantly, TH5487 treatment markedly suppressed the BLM-mediated alveolar epithelial-mesenchymal transition (EMT) and increase in OGG1 protein level in the lungs of WT mice. However, administration of TH5487 did not further improve this fibrotic transformation in Ogg1-/- mice. More importantly, adeno-associated virus-mediated lung-specific OGG1 overexpression accelerated alveolar EMT and the resultant fibrosis progression antagonized by TH5487 in the fibrotic lungs of WT mice, suggesting that the down-regulation of OGG1 protein level could be essential for TH5487 to exert its anti-fibrogenic function. Mechanism study in alveolar epithelial cells demonstrated that TH5487 treatment canceled TGF-ß1-mediated suppression of NEDD4-like E3 ubiquitin ligase (NEDD4L), which ubiquitinated OGG1 and targeted it for proteasomal degradation. Furthermore, TH5487-mediated suppression of alveolar EMT and the fibrotic processes was counteracted by silencing NEDD4L in TGF-ß1-induced alveolar epithelial cells. Collectively, these data underline the potential of TH5487 as an effective anti-fibrotic agent for pulmonary fibrosis.


Asunto(s)
Bencimidazoles , ADN Glicosilasas , Piperidinas , Fibrosis Pulmonar , Animales , Bencimidazoles/farmacología , Bleomicina/farmacología , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Ratones , Ubiquitina-Proteína Ligasas Nedd4 , Piperidinas/farmacología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
4.
Nature ; 600(7888): 324-328, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819670

RESUMEN

Activation-induced cytidine deaminase (AID) catalyses the deamination of deoxycytidines to deoxyuracils within immunoglobulin genes to induce somatic hypermutation and class-switch recombination1,2. AID-generated deoxyuracils are recognized and processed by subverted base-excision and mismatch repair pathways that ensure a mutagenic outcome in B cells3-6. However, why these DNA repair pathways do not accurately repair AID-induced lesions remains unknown. Here, using a genome-wide CRISPR screen, we show that FAM72A is a major determinant for the error-prone processing of deoxyuracils. Fam72a-deficient CH12F3-2 B cells and primary B cells from Fam72a-/- mice exhibit reduced class-switch recombination and somatic hypermutation frequencies at immunoglobulin and Bcl6 genes, and reduced genome-wide deoxyuracils. The somatic hypermutation spectrum in B cells from Fam72a-/- mice is opposite to that observed in mice deficient in uracil DNA glycosylase 2 (UNG2)7, which suggests that UNG2 is hyperactive in FAM72A-deficient cells. Indeed, FAM72A binds to UNG2, resulting in reduced levels of UNG2 protein in the G1 phase of the cell cycle, coinciding with peak AID activity. FAM72A therefore causes U·G mispairs to persist into S phase, leading to error-prone processing by mismatch repair. By disabling the DNA repair pathways that normally efficiently remove deoxyuracils from DNA, FAM72A enables AID to exert its full effects on antibody maturation. This work has implications in cancer, as the overexpression of FAM72A that is observed in many cancers8 could promote mutagenesis.


Asunto(s)
Linfocitos B , ADN Glicosilasas , Reparación de la Incompatibilidad de ADN , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana , Mutación , Proteínas de Neoplasias , Hipermutación Somática de Inmunoglobulina , Animales , Femenino , Humanos , Ratones , Linfocitos B/metabolismo , Sistemas CRISPR-Cas , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Epistasis Genética , Células HEK293 , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Hipermutación Somática de Inmunoglobulina/genética
5.
Sci Rep ; 11(1): 3490, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568707

RESUMEN

The most common oxidative DNA lesion is 8-oxoguanine which is mainly recognized and excised by the 8-oxoG DNA glycosylase 1 (OGG1), initiating the base excision repair (BER) pathway. Telomeres are particularly sensitive to oxidative stress (OS) which disrupts telomere homeostasis triggering genome instability. In the present study, we have investigated the effects of inactivating BER in OS conditions, by using a specific inhibitor of OGG1 (TH5487). We have found that in OS conditions, TH5487 blocks BER initiation at telomeres causing an accumulation of oxidized bases, that is correlated with telomere losses, micronuclei formation and mild proliferation defects. Moreover, the antimetabolite methotrexate synergizes with TH5487 through induction of intracellular reactive oxygen species (ROS) formation, which potentiates TH5487-mediated telomere and genome instability. Our findings demonstrate that OGG1 is required to protect telomeres from OS and present OGG1 inhibitors as a tool to induce oxidative DNA damage at telomeres, with the potential for developing new combination therapies for cancer treatment.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Bencimidazoles/farmacología , ADN Glicosilasas/antagonistas & inhibidores , Reparación del ADN/efectos de los fármacos , Metotrexato/farmacología , Estrés Oxidativo , Piperidinas/farmacología , Telómero/metabolismo , Ciclo Celular , Línea Celular Tumoral , ADN Glicosilasas/metabolismo , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Inestabilidad Genómica , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
6.
ACS Chem Biol ; 16(1): 45-51, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33331782

RESUMEN

DNA glycosylases involved in the first step of the DNA base excision repair pathway are promising targets in cancer therapy. There is evidence that reduction of their activities may enhance cell killing in malignant tumors. Recently, two tetrahydroquinoline compounds named SU0268 and SU0383 were reported to inhibit OGG1 for the excision of 8-hydroxyguanine. This DNA repair protein is one of the major cellular enzymes responsible for excision of a number of oxidatively induced lesions from DNA. In this work, we used gas chromatography-tandem mass spectrometry with isotope-dilution to measure the excision of not only 8-hydroxyguanine but also that of the other major substrate of OGG1, i.e., 2,6-diamino-4-hydroxy-5-formamidopyrimidine, using genomic DNA with multiple purine- and pyrimidine-derived lesions. The excision of a minor substrate 4,6-diamino-5-formamidopyrimidine was also measured. Both SU0268 and SU0383 efficiently inhibited OGG1 activity for these three lesions, with the former being more potent than the latter. Dependence of inhibition on concentrations of SU0268 and SU0383 from 0.05 µmol/L to 10 µmol/L was also demonstrated. The approach used in this work may be applied to the investigation of OGG1 inhibition by SU0268 and SU0383 and other small molecule inhibitors in further studies including cellular and animal models of disease.


Asunto(s)
Daño del ADN , ADN Glicosilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Quinolinas/farmacología , Sulfonamidas/química , Cromatografía de Gases/métodos , Células HeLa , Humanos , Oxidación-Reducción , Quinolinas/química , Espectrometría de Masas en Tándem/métodos
7.
Int J Mol Sci ; 21(23)2020 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-33287345

RESUMEN

Cancer is the second leading cause of death with tens of millions of people diagnosed with cancer every year around the world. Most radio- and chemotherapies aim to eliminate cancer cells, notably by causing severe damage to the DNA. However, efficient repair of such damage represents a common mechanism of resistance to initially effective cytotoxic agents. Thus, development of new generation anticancer drugs that target DNA repair pathways, and more particularly the base excision repair (BER) pathway that is responsible for removal of damaged bases, is of growing interest. The BER pathway is initiated by a set of enzymes known as DNA glycosylases. Unlike several downstream BER enzymes, DNA glycosylases have so far received little attention and the development of specific inhibitors of these enzymes has been lagging. Yet, dysregulation of DNA glycosylases is also known to play a central role in numerous cancers and at different stages of the disease, and thus inhibiting DNA glycosylases is now considered a valid strategy to eliminate cancer cells. This review provides a detailed overview of the activities of DNA glycosylases in normal and cancer cells, their modes of regulation, and their potential as anticancer drug targets.


Asunto(s)
ADN Glicosilasas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Daño del ADN , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/química , Reparación del ADN , Susceptibilidad a Enfermedades , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Relación Estructura-Actividad
8.
Nucleic Acids Res ; 48(21): 12234-12251, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33211885

RESUMEN

Altered oncogene expression in cancer cells causes loss of redox homeostasis resulting in oxidative DNA damage, e.g. 8-oxoguanine (8-oxoG), repaired by base excision repair (BER). PARP1 coordinates BER and relies on the upstream 8-oxoguanine-DNA glycosylase (OGG1) to recognise and excise 8-oxoG. Here we hypothesize that OGG1 may represent an attractive target to exploit reactive oxygen species (ROS) elevation in cancer. Although OGG1 depletion is well tolerated in non-transformed cells, we report here that OGG1 depletion obstructs A3 T-cell lymphoblastic acute leukemia growth in vitro and in vivo, validating OGG1 as a potential anti-cancer target. In line with this hypothesis, we show that OGG1 inhibitors (OGG1i) target a wide range of cancer cells, with a favourable therapeutic index compared to non-transformed cells. Mechanistically, OGG1i and shRNA depletion cause S-phase DNA damage, replication stress and proliferation arrest or cell death, representing a novel mechanistic approach to target cancer. This study adds OGG1 to the list of BER factors, e.g. PARP1, as potential targets for cancer treatment.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , ADN Glicosilasas/genética , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Poli(ADP-Ribosa) Polimerasa-1/inmunología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/mortalidad , Daño del ADN , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Guanina/análogos & derivados , Guanina/metabolismo , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Molecules ; 25(21)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114511

RESUMEN

With increasing antimicrobial resistance there is an urgent need for new strategies to control harmful biofilms. In this study, we have investigated the possibility of utilizing ruthenium(IV) complexes (H3O)2(HL1)2[RuCl6]·2Cl·2EtOH (1) and [RuCl4(CH3CN)2](L32)·H2O (2) (where L1-2-hydroxymethylbenzimadazole, L32-1,4-dihydroquinoxaline-2,3-dione) as effective inhibitors for biofilms formation. The biological activities of the compounds were explored using E. coli, S. aureus, P. aeruginosa PAO1, and P. aeruginosa LES B58. The new chloride ruthenium complexes were characterized by single-crystal X-ray diffraction analysis, Hirshfeld surface analysis, FT-IR, UV-Vis, magnetic and electrochemical (CV, DPV) measurements, and solution conductivity. In the obtained complexes, the ruthenium(IV) ions possess an octahedral environment. The intermolecular classical and rare weak hydrogen bonds, and π···π stacking interactions significantly contribute to structure stabilization, leading to the formation of a supramolecular assembly. The microbiological tests have shown complex 1 exhibited a slightly higher anti-biofilm activity than that of compound 2. Interestingly, electrochemical studies have allowed us to determine the relationship between the oxidizing properties of complexes and their biological activity. Probably the mechanism of action of 1 and 2 is associated with generating a cellular response similar to oxidative stress in bacterial cells.


Asunto(s)
Bacterias/efectos de los fármacos , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Rutenio/química , Antibacterianos/química , Antibacterianos/farmacología , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/antagonistas & inhibidores , Electroquímica , Modelos Moleculares , Conformación Molecular
10.
Biomolecules ; 10(11)2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114607

RESUMEN

8-oxoguanine DNA glycosylase (OGG1) is the main DNA glycosylase responsible for the excision of 7,8-dihydro-8-oxoguanine (8-oxoG) from duplex DNA to initiate base excision repair. This glycosylase activity is relevant in many pathological conditions including cancer, inflammation, and neurodegenerative diseases. To have a better understanding of the role of OGG1, we previously reported TH5487, a potent active site inhibitor of OGG1. Here, we further investigate the consequences of inhibiting OGG1 with TH5487. TH5487 treatment induces accumulation of genomic 8-oxoG lesions. Furthermore, it impairs the chromatin binding of OGG1 and results in lower recruitment of OGG1 to regions of DNA damage. Inhibiting OGG1 with TH5487 interferes with OGG1's incision activity, resulting in fewer DNA double-strand breaks in cells exposed to oxidative stress. This study validates TH5487 as a potent OGG1 inhibitor that prevents the repair of 8-oxoG and alters OGG1-chromatin dynamics and OGG1's recruitment kinetics.


Asunto(s)
Bencimidazoles/farmacología , Cromatina/efectos de los fármacos , ADN Glicosilasas/antagonistas & inhibidores , Piperidinas/farmacología , Cromatina/metabolismo , ADN Glicosilasas/metabolismo , Humanos , Células Tumorales Cultivadas
11.
J Immunol ; 205(8): 2231-2242, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32929043

RESUMEN

The DNA repair enzyme 8-oxoguanine DNA glycosylase 1 (OGG1), which excises 8-oxo-7,8-dihydroguanine lesions induced in DNA by reactive oxygen species, has been linked to the pathogenesis of lung diseases associated with bacterial infections. A recently developed small molecule, SU0268, has demonstrated selective inhibition of OGG1 activity; however, its role in attenuating inflammatory responses has not been tested. In this study, we report that SU0268 has a favorable effect on bacterial infection both in mouse alveolar macrophages (MH-S cells) and in C57BL/6 wild-type mice by suppressing inflammatory responses, particularly promoting type I IFN responses. SU0268 inhibited proinflammatory responses during Pseudomonas aeruginosa (PA14) infection, which is mediated by the KRAS-ERK1-NF-κB signaling pathway. Furthermore, SU0268 induces the release of type I IFN by the mitochondrial DNA-cGAS-STING-IRF3-IFN-ß axis, which decreases bacterial loads and halts disease progression. Collectively, our results demonstrate that the small-molecule inhibitor of OGG1 (SU0268) can attenuate excessive inflammation and improve mouse survival rates during PA14 infection. This strong anti-inflammatory feature may render the inhibitor as an alternative treatment for controlling severe inflammatory responses to bacterial infection.


Asunto(s)
ADN Glicosilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , ADN Glicosilasas/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/patología
12.
Eur Rev Med Pharmacol Sci ; 24(11): 6409-6416, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32572938

RESUMEN

OBJECTIVE: The aim of this study was to investigate the effects of salinomycin (Sal) on expressions of baculoviral IAP repeat-containing 5 (BIRC5) and Nei endonuclease VIII-like 2 (NEIL2) and radiotherapy sensitivity of nasopharyngeal carcinoma (NPC). MATERIALS AND METHODS: Human NPC CNE-2 cell lines were used as research objects in this study. Subsequently, the cells received intervention with Sal at different concentrations, radioactive rays at different doses and Sal combined with radioactive rays. The growth inhibition rate of CNE-2 cells was detected via methyl thiazolyl tetrazolium (MTT) assay. The dose-effect relations of Sal, radioactive rays and combination therapy with the inhibitory effect on CNE-2 cells were obtained. CNE-2 cells receiving intervention with Sal at an appropriate concentration or radioactive rays at an appropriate dose alone and Sal combined with radioactive rays were used as intervention groups (Sal group, Radiation group and Combination group). However, those added with an equal amount of DMSO were set as Control group. Next, the cycle, apoptosis and apoptotic morphology of CNE-2 cells were observed via flow cytometry and Hoechst assay, respectively. Moreover, the expressions of apoptosis-related proteins Caspase-3, B-cell lymphoma-2 (Bcl-2) and Bcl-2 associated X protein (Bax), as well as BIRC5 and NEIL2 proteins in CNE-2 cells were determined using Western blotting. RESULTS: Under the intervention with Sal or radioactive rays alone, the growth inhibition rate of CNE-2 cells rose in a concentration/dose-dependent manner. With the increase in Sal concentration in combination therapy, the growth inhibition rate of CNE-2 cells significantly increased (p<0.05). Compared with Control group, Sal group, Radiation group, and Combination group exhibited remarkably lower colony formation rate, higher proportion of CNE-2 cells in the G2/M phase, enhanced apoptosis of CNE-2 cells with nuclear fragmentation, increased expressions of pro-apoptotic proteins Caspase-3 and Bax, decreased expression of anti-apoptotic protein Bcl-2, and lower protein expressions of BIRC5 and NEIL2 in cells (p<0.05). Compared with Radiation group, the Combination group had significantly decreased colony formation rate, increased proportion of CNE-2 cells in the G2/M phase, enhanced apoptosis of CNE-2 cells with more nuclear fragmentation and other apoptosis characteristics, increased expressions of pro-apoptotic proteins Caspase-3 and Bax, decreased expression of anti-apoptotic protein Bcl-2, and decreased protein expressions of BIRC5 and NEIL2 in cells (p<0.05). CONCLUSIONS: Sal enhances the radiotherapy sensitivity of NPC and reduces the protein expressions of BIRC5 and NEIL2 in cells.


Asunto(s)
Antineoplásicos/farmacología , ADN Glicosilasas/antagonistas & inhibidores , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/terapia , Piranos/farmacología , Survivin/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Humanos , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Survivin/genética , Survivin/metabolismo , Células Tumorales Cultivadas
13.
Int J Mol Sci ; 21(9)2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-32354123

RESUMEN

DNA glycosylases are enzymes that initiate the base excision repair pathway, a major biochemical process that protects the genomes of all living organisms from intrinsically and environmentally inflicted damage. Recently, base excision repair inhibition proved to be a viable strategy for the therapy of tumors that have lost alternative repair pathways, such as BRCA-deficient cancers sensitive to poly(ADP-ribose)polymerase inhibition. However, drugs targeting DNA glycosylases are still in development and so far have not advanced to clinical trials. In this review, we cover the attempts to validate DNA glycosylases as suitable targets for inhibition in the pharmacological treatment of cancer, neurodegenerative diseases, chronic inflammation, bacterial and viral infections. We discuss the glycosylase inhibitors described so far and survey the advances in the assays for DNA glycosylase reactions that may be used to screen pharmacological libraries for new active compounds.


Asunto(s)
ADN Glicosilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/enzimología , Reparación del ADN , Descubrimiento de Drogas , Inhibidores Enzimáticos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Virosis/tratamiento farmacológico , Virosis/enzimología
14.
Bioorg Med Chem ; 28(11): 115507, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32327352

RESUMEN

The DNA repair enzyme AAG has been shown in mice to promote tissue necrosis in response to ischaemic reperfusion or treatment with alkylating agents. A chemical probe inhibitor is required for investigations of the biological mechanism causing this phenomenon and as a lead for drugs that are potentially protective against tissue damage from organ failure and transplantation, and alkylative chemotherapy. Herein, we describe the rationale behind the choice of arylmethylpyrrolidines as appropriate aza-nucleoside mimics for an inhibitor followed by their synthesis and the first use of a microplate-based assay for quantification of their inhibition of AAG. We finally report the discovery of an imidazol-4-ylmethylpyrrolidine as a fragment-sized, weak inhibitor of AAG.


Asunto(s)
Alquilantes/farmacología , Compuestos Aza/farmacología , ADN Glicosilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Nucleósidos/farmacología , Alquilantes/síntesis química , Alquilantes/química , Animales , Compuestos Aza/síntesis química , Compuestos Aza/química , Cristalografía por Rayos X , ADN Glicosilasas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Ratones , Modelos Moleculares , Estructura Molecular , Nucleósidos/síntesis química , Nucleósidos/química , Relación Estructura-Actividad
15.
Int J Mol Sci ; 21(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192183

RESUMEN

DNA glycosylases are emerging as relevant pharmacological targets in inflammation, cancer and neurodegenerative diseases. Consequently, the search for inhibitors of these enzymes has become a very active research field. As a continuation of previous work that showed that 2-thioxanthine (2TX) is an irreversible inhibitor of zinc finger (ZnF)-containing Fpg/Nei DNA glycosylases, we designed and synthesized a mini-library of 2TX-derivatives (TXn) and evaluated their ability to inhibit Fpg/Nei enzymes. Among forty compounds, four TXn were better inhibitors than 2TX for Fpg. Unexpectedly, but very interestingly, two dithiolated derivatives more selectively and efficiently inhibit the zincless finger (ZnLF)-containing enzymes (human and mimivirus Neil1 DNA glycosylases hNeil1 and MvNei1, respectively). By combining chemistry, biochemistry, mass spectrometry, blind and flexible docking and X-ray structure analysis, we localized new TXn binding sites on Fpg/Nei enzymes. This endeavor allowed us to decipher at the atomic level the mode of action for the best TXn inhibitors on the ZnF-containing enzymes. We discovered an original inhibition mechanism for the ZnLF-containing Fpg/Nei DNA glycosylases by disulfide cyclic trimeric forms of dithiopurines. This work paves the way for the design and synthesis of a new structural class of inhibitors for selective pharmacological targeting of hNeil1 in cancer and neurodegenerative diseases.


Asunto(s)
ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Purinas/química , Purinas/farmacología , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología , Bacterias/enzimología , Sitios de Unión , Cristalografía por Rayos X , Reparación del ADN , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , Relación Estructura-Actividad , Tioxantenos/química , Tioxantenos/farmacología
16.
Oncogene ; 39(3): 603-616, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511648

RESUMEN

Epstein-Barr virus (EBV) immortalizes human B-lymphocytes and is implicated in the pathogenesis of lymphoid and epithelial cell malignancies. The EBV nuclear antigen (EBNA)-1 induces the accumulation of reactive oxygen species (ROS), which enables B-cell immortalization but causes oxidative DNA damage and triggers antiproliferative DNA damage responses. By comparing pairs of EBV-negative and -positive tumor cell lines we found that, while associated with the accumulation of oxidized nucleotides, EBV carriage promotes the concomitant activation of oxo-dNTP sanitization and purging pathways, including upregulation of the nucleoside triphosphatase mut-T homolog 1 (MTH1) and the DNA glycosylases 8-oxoguanine-glycosylase-1 (OGG1) and mut-Y homolog (MUTYH). Expression of EBNA1 was reversibly associated with transcriptional activation of this cellular response. DNA damage and apoptosis were preferentially induced in EBNA1-positive cell lines by treatment with MTH1 inhibitors, suggesting that virus carriage is linked to enhanced vulnerability to oxidative stress. MTH1, OGG1, and MUTYH were upregulated upon EBV infection in primary B-cells and treatment with MTH1 inhibitors prevented B-cell immortalization. These findings highlight an important role of the cellular antioxidant response in sustaining EBV infection, and suggests that targeting this cellular defense may offer a novel approach to antiviral therapy and could reduce the burden of EBV associated cancer.


Asunto(s)
Linfocitos B/patología , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/patología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/patogenicidad , Neoplasias/patología , Linfocitos B/virología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Crizotinib/farmacología , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/metabolismo , Humanos , Neoplasias/virología , Estrés Oxidativo/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Cultivo Primario de Células , Pirimidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
17.
Biochem Biophys Res Commun ; 521(1): 245-251, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31653343

RESUMEN

Parkinson's disease (PD) is well known as a neurodegenerative disorder with progressive loss of dopaminergic (DA) neurons. Nei-like 1 (NEIL1) is one of four mammalian DNA glycosylases involved in the progression of various diseases, including neuroinflammation. However, it is still unknown if the expression changes of NEIL1 could contribute to PD progression. In the present study, we established mouse model with PD using 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to explore the effects of NEIL1 on PD development. Here, we found that NEIL1 deletion significantly promoted the motor dysfunction in the wild type mice treated with 6-OHDA. Furthermore, DA neuronal loss was further accelerated by NEIL1 deletion in 6-OHDA-injected mice, as evidenced by the significantly reduced expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT). Furthermore, in PD mice induced by MPTP, remarkably reduced expression of NEIL1 was observed in nigra and striatum of mice. A strong positive correlation was detected in the expression of NEIL1 and the survival rate of DA neurons. Also, NEIL1 ablation further elevated the DA neuronal loss in MPTP-treated mice, accompanied with higher glial activation, as evidenced by the obvious up-regulation of glial fibrillary acidic protein (GFAP) and Ionized calcium-Binding Adapter molecule 1 (Iba1). Moreover, MPTP-triggered inflammation was highly aggravated by the loss of NEIL1 through inducing the expression of pro-inflammatory cytokines and chemokines. In contrast, promoting NEIL1 expression effectively reversedPD progression induced by MPTP in mice. Together, these results demonstrated that NEIL1 insufficiency might be a contributing factor for the progression of PD, which therefore could be considered as a novel candidate to develop effective treatments against PD progression.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Apomorfina/farmacología , ADN Glicosilasas/antagonistas & inhibidores , Inflamación/inducido químicamente , Oxidopamina/farmacología , Enfermedad de Parkinson/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Animales , Apomorfina/administración & dosificación , ADN Glicosilasas/deficiencia , ADN Glicosilasas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Oxidopamina/administración & dosificación , Técnicas Estereotáxicas
18.
ACS Chem Biol ; 14(12): 2606-2615, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31622553

RESUMEN

Oxidative damage in DNA is one of the primary sources of mutations in the cell. The activities of repair enzymes 8-oxoguanine DNA glycosylase (OGG1) and human MutT Homologue 1 (NUDT1 or MTH1), which work together to ameliorate this damage, are closely linked to mutagenesis, genotoxicity, cancer, and inflammation. Here we have undertaken the development of small-molecule dual inhibitors of the two enzymes as tools to test the relationships between these pathways and disease. The compounds preserve key structural elements of known inhibitors of the two enzymes, and they were synthesized and assayed with recently developed luminescence assays of the enzymes. Further structural refinement of initial lead molecules yielded compound 5 (SU0383) with IC50(NUDT1) = 0.034 µM and IC50(OGG1) = 0.49 µM. The compound SU0383 displayed low toxicity in two human cell lines at 10 µM. Experiments confirm the ability of SU0383 to increase sensitivity of tumor cells to oxidative stress. Dual inhibitors of these two enzymes are expected to be useful in testing multiple hypotheses regarding the roles of 8-oxo-dG in multiple disease states.


Asunto(s)
ADN Glicosilasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Guanina/análogos & derivados , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Línea Celular Tumoral , ADN Glicosilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Guanina/antagonistas & inhibidores , Humanos
19.
Proc Natl Acad Sci U S A ; 116(44): 22158-22163, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31611371

RESUMEN

Human cells express up to 9 active DNA cytosine deaminases with functions in adaptive and innate immunity. Many cancers manifest an APOBEC mutation signature and APOBEC3B (A3B) is likely the main enzyme responsible. Although significant numbers of APOBEC signature mutations accumulate in tumor genomes, the majority of APOBEC-catalyzed uracil lesions are probably counteracted in an error-free manner by the uracil base excision repair pathway. Here, we show that A3B-expressing cells can be selectively killed by inhibiting uracil DNA glycosylase 2 (UNG) and that this synthetic lethal phenotype requires functional mismatch repair (MMR) proteins and p53. UNG knockout human 293 and MCF10A cells elicit an A3B-dependent death. This synthetic lethal phenotype is dependent on A3B catalytic activity and reversible by UNG complementation. A3B expression in UNG-null cells causes a buildup of genomic uracil, and the ensuing lethality requires processing of uracil lesions (likely U/G mispairs) by MSH2 and MLH1 (likely noncanonical MMR). Cancer cells expressing high levels of endogenous A3B and functional p53 can also be killed by expressing an UNG inhibitor. Taken together, UNG-initiated base excision repair is a major mechanism counteracting genomic mutagenesis by A3B, and blocking UNG is a potential strategy for inducing the selective death of tumors.


Asunto(s)
Muerte Celular , Citidina Desaminasa/genética , ADN Glicosilasas/genética , Desaminasas APOBEC , Línea Celular Tumoral , ADN Glicosilasas/antagonistas & inhibidores , Reparación de la Incompatibilidad de ADN , Reparación del ADN , Técnicas de Inactivación de Genes , Humanos , Modelos Moleculares , Ubiquitinación
20.
Nucleic Acids Res ; 47(17): 9132-9143, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31329989

RESUMEN

Poly(ADP-ribose) polymerases (PARPs) facilitate the repair of DNA single-strand breaks (SSBs). When PARPs are inhibited, unrepaired SSBs colliding with replication forks give rise to cytotoxic double-strand breaks. These are normally rescued by homologous recombination (HR), but, in cells with suboptimal HR, PARP inhibition leads to genomic instability and cell death, a phenomenon currently exploited in the therapy of ovarian cancers in BRCA1/2 mutation carriers. In spite of their promise, resistance to PARP inhibitors (PARPis) has already emerged. In order to identify the possible underlying causes of the resistance, we set out to identify the endogenous source of DNA damage that activates PARPs. We argued that if the toxicity of PARPis is indeed caused by unrepaired SSBs, these breaks must arise spontaneously, because PARPis are used as single agents. We now show that a significant contributor to PARPi toxicity is oxygen metabolism. While BRCA1-depleted or -mutated cells were hypersensitive to the clinically approved PARPi olaparib, its toxicity was significantly attenuated by depletion of OGG1 or MYH DNA glycosylases, as well as by treatment with reactive oxygen species scavengers, growth under hypoxic conditions or chemical OGG1 inhibition. Thus, clinical resistance to PARPi therapy may emerge simply through reduced efficiency of oxidative damage repair.


Asunto(s)
Proteína BRCA1/genética , ADN Glicosilasas/genética , Neoplasias Ováricas/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasas/genética , Línea Celular Tumoral , Roturas del ADN de Cadena Simple/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Femenino , Recombinación Homóloga/efectos de los fármacos , Recombinación Homóloga/genética , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Oxidación-Reducción/efectos de los fármacos , Ftalazinas/efectos adversos , Ftalazinas/farmacología , Piperazinas/efectos adversos , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Mutaciones Letales Sintéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA