Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Nucleic Acids Res ; 49(19): 11367-11378, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34614154

RESUMEN

Bacterial chromosome replication is mainly catalyzed by DNA polymerase III, whose beta subunits enable rapid processive DNA replication. Enabled by the clamp-loading complex, the two beta subunits form a ring-like clamp around DNA and keep the polymerase sliding along. Given the essential role of ß-clamp, its inhibitors have been explored for antibacterial purposes. Similarly, ß-clamp is an ideal target for bacteriophages to shut off host DNA synthesis during host takeover. The Gp168 protein of phage Twort is such an example, which binds to the ß-clamp of Staphylococcus aureus and prevents it from loading onto DNA causing replication arrest. Here, we report a cryo-EM structure of the clamp-Gp168 complex at 3.2-Å resolution. In the structure of the complex, the Gp168 dimer occupies the DNA sliding channel of ß-clamp and blocks its loading onto DNA, which represents a new inhibitory mechanism against ß-clamp function. Interestingly, the key residues responsible for this interaction on the ß-clamp are well conserved among bacteria. We therefore demonstrate that Gp168 is potentially a cross-species ß-clamp inhibitor, as it forms complex with the Bacillus subtilis ß-clamp. Our findings reveal an alternative mechanism for bacteriophages to inhibit ß-clamp and provide a new strategy to combat bacterial drug resistance.


Asunto(s)
Bacillus subtilis/efectos de los fármacos , Bacteriófagos/química , ADN Bacteriano/química , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Proteínas Virales/química , Secuencia de Aminoácidos , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Bacteriófagos/genética , Bacteriófagos/metabolismo , Sitios de Unión , Clonación Molecular , Microscopía por Crioelectrón , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Replicación del ADN/efectos de los fármacos , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
2.
Biomolecules ; 10(9)2020 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842666

RESUMEN

Resveratrol, the most widely studied natural phytochemical, has been shown to interact with different target proteins. Previous studies show that resveratrol binds and inhibits DNA polymerases and some other enzymes; however, the binding and functioning mechanisms remain unknown. The elucidated knowledge of inhibitory mechanisms of resveratrol will assist us in new drug discovery. We utilized molecular docking and molecular dynamics (MD) simulation to reveal how resveratrol and structurally similar compounds bind to various nucleotide-dependent enzymes, specifically, DNA polymerases, HIV-1 reverse transcriptase, and ribonucleotide reductase. The results show that resveratrol and its analogs exert their inhibitory effects by competing with the substrate dNTPs in these enzymes and blocking elongation of chain polymerization. In addition, the results imply that resveratrol binds to a variety of other ATP-/NTP-binding proteins.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Resveratrol/análogos & derivados , Unión Competitiva , Dominio Catalítico , ADN Polimerasa I/antagonistas & inhibidores , ADN Polimerasa I/química , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/química , Descubrimiento de Drogas , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/química , Humanos , Enlace de Hidrógeno , Técnicas In Vitro , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Conformación Proteica , Resveratrol/química , Resveratrol/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/química , Relación Estructura-Actividad
3.
Nucleic Acids Res ; 48(10): 5540-5554, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32347931

RESUMEN

In the fight against antimicrobial resistance, the bacterial DNA sliding clamp, ß-clamp, is a promising drug target for inhibition of DNA replication and translesion synthesis. The ß-clamp and its eukaryotic homolog, PCNA, share a C-terminal hydrophobic pocket where all the DNA polymerases bind. Here we report that cell penetrating peptides containing the PCNA-interacting motif APIM (APIM-peptides) inhibit bacterial growth at low concentrations in vitro, and in vivo in a bacterial skin infection model in mice. Surface plasmon resonance analysis and computer modeling suggest that APIM bind to the hydrophobic pocket on the ß-clamp, and accordingly, we find that APIM-peptides inhibit bacterial DNA replication. Interestingly, at sub-lethal concentrations, APIM-peptides have anti-mutagenic activities, and this activity is increased after SOS induction. Our results show that although the sequence homology between the ß-clamp and PCNA are modest, the presence of similar polymerase binding pockets in the DNA clamps allows for binding of the eukaryotic binding motif APIM to the bacterial ß-clamp. Importantly, because APIM-peptides display both anti-mutagenic and growth inhibitory properties, they may have clinical potential both in combination with other antibiotics and as single agents.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , ADN Polimerasa III/antagonistas & inhibidores , Péptidos/química , Péptidos/farmacología , Animales , Antibacterianos/metabolismo , Antibacterianos/uso terapéutico , ADN Polimerasa III/química , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Ratones Endogámicos BALB C , Mutagénesis/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/química , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Péptidos/metabolismo , Péptidos/uso terapéutico , Antígeno Nuclear de Célula en Proliferación/metabolismo , Dominios y Motivos de Interacción de Proteínas , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/crecimiento & desarrollo
4.
Cancer Lett ; 482: 126-135, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31954770

RESUMEN

Glioblastoma (GBM) has limited therapeutic options. DNA repair mechanisms contribute GBM cells to escape therapies and re-establish tumor growth. Multiple studies have shown that POLD2 plays a critical role in DNA replication, DNA repair and genomic stability. We demonstrate for the first time that POLD2 is highly expressed in human glioma specimens and that expression correlates with poor patient survival. siRNA or shRNA POLD2 inhibited GBM cell proliferation, cell cycle progression, invasiveness, sensitized GBM cells to chemo/radiation-induced cell death and reversed the cytoprotective effects of EGFR signaling. Conversely, forced POLD2 expression was found to induce GBM cell proliferation, colony formation, invasiveness and chemo/radiation resistance. POLD2 expression associated with stem-like cell subsets (CD133+ and SSEA-1+ cells) and positively correlated with Sox2 expression in clinical specimens. Its expression was induced by Sox2 and inhibited by the forced differentiation of GBM neurospheres. shRNA-POLD2 modestly inhibited GBM neurosphere-derived orthotopic xenografts growth, when combined with radiation, dramatically inhibited xenograft growth in a cooperative fashion. These novel findings identify POLD2 as a new potential therapeutic target for enhancing GBM response to current standard of care therapeutics.


Asunto(s)
Neoplasias Encefálicas/terapia , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Glioblastoma/terapia , ARN Interferente Pequeño/administración & dosificación , Temozolomida/administración & dosificación , Regulación hacia Arriba , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , ADN Polimerasa III/antagonistas & inhibidores , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , ARN Interferente Pequeño/farmacología , Radioterapia , Análisis de Supervivencia , Temozolomida/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
5.
DNA Cell Biol ; 39(1): 50-56, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31750734

RESUMEN

POLD1 encodes the catalytic subunit of DNA polymerase delta (Polδ), the major lagging strand polymerase, which also participates in DNA repair. Mutations affecting the exonuclease domain increase the risk of various cancers, while mutations that change the polymerase active site cause a progeroid syndrome called mandibular hypoplasia, deafness, progeroid features, and lipodystrophy (MDPL) syndrome. We generated a set of catalytic subunit of human telomerase (hTERT)-immortalized human fibroblasts expressing wild-type or mutant POLD1 using the retroviral LXSN vector system. In the resulting cell lines, expression of endogenous POLD1 was suppressed in favor of the recombinant POLD1. The siRNA screening of DNA damage-related genes revealed that fibroblasts expressing D316H and S605del POLD1 were more sensitive to knockdowns of ribonuclease reductase (RNR) components, RRM1 and RRM2 in the presence of hydroxyurea (HU), an RNR inhibitor. On the contrary, SAMHD1 siRNA, which increases the concentration of dNTPs, increased growth of wild type, D316H, and S605del POLD1 fibroblasts. Hypersensitivity to dNTP synthesis inhibition in POLD1 mutant lines was confirmed using gemcitabine. Our finding is consistent with the notion that reduced dNTP concentration negatively affects the cell growth of hTERT fibroblasts expressing exonuclease and polymerase mutant POLD1.


Asunto(s)
ADN Polimerasa III/genética , Desoxirribonucleótidos/metabolismo , Fibroblastos/metabolismo , Mutación , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/metabolismo , Sordera/genética , Sordera/metabolismo , Exonucleasas/genética , Exonucleasas/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Hidroxiurea/farmacología , Lipodistrofia/genética , Lipodistrofia/metabolismo , Lipodistrofia/patología , Interferencia de ARN , Proteína 1 que Contiene Dominios SAM y HD/genética , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Síndrome
6.
Int J Mol Sci ; 20(18)2019 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540106

RESUMEN

Staphylococcus aureus resistance to current antibiotics has become the greatest global challenge facing public health. The development of new antimicrobial agents is urgent and important and is needed to provide additional therapeutic options. In our previous study, we found out that pterostilbene exhibited potent antibacterial activity, especially against methicillin-resistant Staphylococcus aureus (MRSA). According to previous studies, 1,2,3-triazole, with the characteristic of increasing the interaction with the target readily and enhancing water solubility, were widely used in the approved anti-bacterial drugs. Therefore, these results attract our interest to use the structure of pterostilbene as a scaffold for the hybrid 1,2,3-triazole moiety to develop a novel anti-MRSA infection agent. In this study, we demonstrated the design and synthesis of a series of triazolylpterostilbene derivatives. Among these compounds, compound 4d exhibited the most potent anti-MRSA activity with a minimum inhibitory concentration (MIC) value of 1.2-2.4 µg/mL and a minimum bactericidal concentration (MBC) value of 19.5-39 µg/mL. The structure-activity relationship and antibacterial mechanism were investigated in this study. Molecular docking studies were carried out to verify and rationalize the biological results. In this study, the results confirmed that our design could successfully increase the inhibitory activity and specificity against MRSA. Compound 4d could be used as a candidate for anti-bacterial agents and in depth vivo studies should be further investigated.


Asunto(s)
Antibacterianos/síntesis química , Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Estilbenos/química , Triazoles/química , Antibacterianos/química , ADN Polimerasa III/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
7.
Bioorg Med Chem ; 27(15): 3209-3217, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31221610

RESUMEN

Despite the growing global crisis caused by antimicrobial drug resistance among pathogenic bacteria, the number of new antibiotics, especially new chemical class of antibiotics under development is insufficient to tackle the problem. Our review focuses on an emerging class of antibacterial therapeutic agents that holds a completely novel mechanism of action, namely, inhibition of bacterial DNA polymerase IIIC. The recent entry of this new class into human trials may herald the introduction of novel drugs whose novel molecular target precludes cross-resistance with existing antibiotic classes. This review therefore examines the evolution of DNA pol IIIC inhibitors from the discovery of 6-(p-hydroxyphenylazo)uracil (HPUra) in the 1960s to the development of current first-in-class N7-substituted guanine drug candidate ACX-362E, now under clinical development for the treatment of Clostridioides difficile infection.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , ADN Polimerasa III/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Uracilo/farmacología , ADN Polimerasa III/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Inhibidores de la Síntesis del Ácido Nucleico/síntesis química , Inhibidores de la Síntesis del Ácido Nucleico/química , Uracilo/análogos & derivados , Uracilo/química
8.
DNA Repair (Amst) ; 74: 17-25, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30641156

RESUMEN

DNA replication fidelity maintains low mutation rates in bacteria. The ε-subunit of a replisome generally acts as the main proofreader during replication, using its 3'-5' exonuclease activity to excise misincorporated bases thereby maintaining faithful replication. In Mycobacterium tuberculosis (Mtb), however, the polymerase and histidinol phosphatase (PHP) domain of the DNA polymerase DnaE1 is the primary proofreader. This domain thus maintains low mutation rates during replication and is an attractive target for drug development. Even though the structures of DnaE polymerases are available from various organisms, including Mtb, the mechanism of exonuclease activity remains elusive. In this study, we sought to unravel the mechanism and also to identify scaffolds that can specifically inhibit the exonuclease activity. To gain insight into the mode of action, we also characterized the PHP domain of the Mtb error-prone polymerase DnaE2 which shares a nearly identical active site with DnaE1-PHP. Kinetic and mutational studies allowed us to identify the critical residue involved in catalysis. Combined inhibition and computational studies also revealed a specific mode of inhibition of DnaE1-PHP by nucleoside diphosphates. Thus, this study lays the foundation for the rational design of novel inhibitors which target the Mtb replicative proofreader.


Asunto(s)
ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/metabolismo , Replicación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Exonucleasas/metabolismo , Mycobacterium tuberculosis/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , ADN Polimerasa III/química , Diseño de Fármacos , Cinética , Modelos Moleculares , Mycobacterium tuberculosis/genética
9.
J Biomol Struct Dyn ; 37(16): 4222-4237, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30526389

RESUMEN

Emerging widespread bacterial resistance to current antibiotics with traditional targets is one of the major global concerns. Therefore, so many investigations are exploring the potential of other druggable macromolecules of bacteria such as replication machinery components that are not addressed by previous antibiotics. DNA polymerase is the major part of this machine. However, a few studies have been done on it so far. In this respect, we report the discovery of four new plant-based leads against DNA polymerase (pol) IIIC (three leads) and pol IIIE (one lead) of Gram-positive and negative bacteria by combining a sequentially constrained high-throughput virtual screenings on Traditional Chinese Medicine Database with in vitro assays. The compounds displayed relatively good levels of inhibitory effect. They were active against their designated targets at micromolar concentrations. The IC50 values for them are ranged from 25 to 111 µM. In addition, they showed minimum inhibitory concentrations in the range of 8-128 µg/mL against five representatives of pathogenic bacteria species. However, they were inactive against Pseudomonas aeruginosa. Given these results, these leads hold promise for future modification and optimization to be more effective in lower concentrations and also against most of the important bacterial species. Communicated by Ramaswamy H. Sarma.


Asunto(s)
ADN Polimerasa III/química , Replicación del ADN/efectos de los fármacos , Plomo/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Antibacterianos/efectos adversos , Simulación por Computador , ADN Polimerasa III/antagonistas & inhibidores , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/patogenicidad , Humanos , Plomo/química , Pruebas de Sensibilidad Microbiana , Inhibidores de la Síntesis del Ácido Nucleico/química , Células Procariotas/efectos de los fármacos , Células Procariotas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad
10.
DNA Repair (Amst) ; 73: 64-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30470508

RESUMEN

Human DNA polymerase δ is normally present in unstressed, non-dividing cells as a heterotetramer (Pol δ4). Its smallest subunit, p12, is transiently degraded in response to UV damage, as well as during the entry into S-phase, resulting in the conversion of Pol δ4 to a trimer (Pol δ3). In order to further understand the specific cellular roles of these two forms of Pol δ, the gene (POLD4) encoding p12 was disrupted by CRISPR/Cas9 to produce p12 knockout (p12KO) cells. Thus, Pol δ4 is absent in p12KO cells, leaving Pol δ3 as the sole source of Pol δ activity. GFP reporter assays revealed that the p12KO cells exhibited a defect in homologous recombination (HR) repair, indicating that Pol δ4, but not Pol δ3, is required for HR. Expression of Flag-tagged p12 in p12KO cells to restore Pol δ4 alleviated the HR defect. These results establish a specific requirement for Pol δ4 in HR repair. This leads to the prediction that p12KO cells should be more sensitive to chemotherapeutic agents, and should exhibit synthetic lethal killing by PARP inhibitors. These predictions were confirmed by clonogenic cell survival assays of p12KO cells treated with cisplatin and mitomycin C, and with the PARP inhibitors Olaparib, Talazoparib, Rucaparib, and Niraparib. The sensitivity to PARP inhibitors in H1299-p12KO cells was alleviated by expression of Flag-p12. These findings have clinical significance, as the expression levels of p12 could be a predictive biomarker of tumor response to PARP inhibitors. In addition, small cell lung cancers (SCLC) are known to exhibit a defect in p12 expression. Analysis of several SCLC cell lines showed that they exhibit hypersensitivity to PARP inhibitors, providing evidence that loss of p12 expression could represent a novel molecular basis for HR deficiency.


Asunto(s)
ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/genética , Técnicas de Inactivación de Genes , Recombinación Homóloga/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Línea Celular Tumoral , Cisplatino/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Polimerasa III/deficiencia , Células HeLa , Recombinación Homóloga/efectos de los fármacos , Humanos , Mitomicina/farmacología
11.
Cancer Biol Ther ; 20(4): 474-486, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30427259

RESUMEN

Chromosomal duplication is targeted by various chemotherapeutic agents for the treatment of cancer. However, there is no specific inhibitor of DNA polymerases that is viable for cancer management. Through structure-based in silico screening of the ZINC database, we identified a specific inhibitor of DNA polymerase δ. The discovered inhibitor, Zelpolib, is projected to bind to the active site of Pol δ when it is actively engaged in DNA replication through interactions with DNA template and primer. Zelpolib shows robust inhibition of Pol δ activity in reconstituted DNA replication assays. Under cellular conditions, Zelpolib is taken up readily by cancer cells and inhibits DNA replication in assays to assess global DNA synthesis or single-molecule bases by DNA fiber fluorography. In addition, we show that Zelpolib displays superior antiproliferative properties to methotrexate, 5-flourouracil, and cisplatin in triple-negative breast cancer cell line, pancreatic cancer cell line and platinum-resistant pancreatic cancer cell line. Pol δ is not only involved in DNA replication, it is also a key component in many DNA repair pathways. Pol δ is the key enzyme responsible for D-loop extension during homologous recombination. Indeed, Zelpolib shows robust inhibition of homologous recombination repair of DNA double-strand breaks and induces "BRCAness" in HR-proficient cancer cells and enhances their sensitivity to PARP inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , ADN Polimerasa III/antagonistas & inhibidores , Replicación del ADN/efectos de los fármacos , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Neoplasias/patología , Antineoplásicos/aislamiento & purificación , Simulación por Computador , Daño del ADN , Bases de Datos Farmacéuticas , Inhibidores Enzimáticos/aislamiento & purificación , Recombinación Homóloga , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Reparación del ADN por Recombinación , Células Tumorales Cultivadas
12.
J Am Chem Soc ; 140(50): 17465-17473, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30461272

RESUMEN

Abiespiroside A (1), beshanzuenone C (2), and beshanzuenone D (3) belong to the Abies sesquiterpenoid family. Beshanzuenones C (2) and D (3) are isolated from the critically endangered Chinese fir tree species Abies beshanzuensis and demonstrated weak inhibiting activity against protein tyrosine phosphatase 1B (PTP1B). We describe herein the first total syntheses of these Abies sesquiterpenoids relying on the sustainable and inexpensive chiral pool molecule (+)-carvone. The syntheses feature a palladium-catalyzed hydrocarbonylative lactonization to install the 6,6-fused bicyclic ring system and a Dreiding-Schmidt reaction to build the oxaspirolactone moiety of these target molecules. Our chemical total syntheses of these Abies sesquiterpenoids have enabled (i) the validation of beshanzuenone C's weak PTP1B inhibiting potency, (ii) identification of new synthetic analogs with promising and selective protein tyrosine phosphatase SHP2 inhibiting potency, and (iii) preparation of azide-tagged probe molecules for target identification via a chemoproteomic approach. The latter has resulted in the identification and evaluation of DNA polymerase epsilon subunit 3 (POLE3) as one of the novel cellular targets of these Abies sesquiterpenoids and their analogs. More importantly, via POLE3 inactivation by probe molecule 29 and knockdown experiment, we further demonstrated that targeting POLE3 with small molecules may be a novel strategy for chemosensitization to DNA damaging drugs such as etoposide in cancer.


Asunto(s)
Abies/química , Inhibidores Enzimáticos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Sesquiterpenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Ciclización , ADN Polimerasa III/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Sinergismo Farmacológico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Etopósido/farmacología , Humanos , Nucleoproteínas/antagonistas & inhibidores , Sesquiterpenos/síntesis química , Sesquiterpenos/química
13.
FEBS Lett ; 591(15): 2311-2322, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28656718

RESUMEN

The ß-clamp is the processivity-promoting factor for most of the enzymes in prokaryotic DNA replication; hence, it is a crucial drug target. In the present study, we investigated the ß-clamp from Helicobacter pylori, aiming to seek potential drug molecules against this gastric-cancer-causing bacterium. An in silico screening of Food and Drug Administration (FDA) approved drugs against the H. pylori ß-clamp, followed by its in vitro inhibition using a surface competition approach, yielded the drug diflunisal as a positive initial hit. Diflunisal inhibits the growth of H. pylori in the micromolar range. We determined the structure of diflunisal in complex with the ß-clamp to show that the drug binds at subsite I, which is a protein-protein interaction site. Successful identification of FDA-approved molecules against H. pylori may lead to better and faster drug development.


Asunto(s)
Antibacterianos/farmacología , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/química , Diflunisal/farmacología , Helicobacter pylori/efectos de los fármacos , Antibacterianos/química , Sitios de Unión , Cristalografía por Rayos X , ADN Ligasas/metabolismo , ADN Polimerasa III/metabolismo , Diflunisal/química , Aprobación de Drogas , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Helicobacter pylori/enzimología , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Conformación Proteica , Estados Unidos , United States Food and Drug Administration
14.
Cell Biochem Biophys ; 74(4): 473-481, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27651172

RESUMEN

Tuberculosis, caused by Mycobacterium tuberculosis, is one of the most common causes of death in the world. Mycobacterium tuberculosis -sliding clamp is a protein essential for many important DNA transactions including replication and DNA repair proteins, thus, a potential drug target for tuberculosis. Further investigation is needed in understanding DNA polymerase sliding clamp structure, especially from a computational perspective. In this study, we employ a wide-range of comparative molecular dynamic analyses on two systems: Mycobacterium tuberculosis - sliding clamp enzyme in its apo and bound form. The results reported in this study shows apo conformation to be less stable, as compared to bound conformation with an average radius of gyration of 25.812 and 25.459 Å, respectively. This was further supported by root mean square fluctuation, where an apo enzyme showed a higher degree of flexibility. However, the presence of the ligand lowers radius of gyration and root mean square fluctuation and also leads to an existence of negative correlated motions. Principal component analysis further justifies the same findings, whereby the apo enzyme exhibits a higher fluctuation compared to the bound complex. In addition, a stable 310 helix located at the binding site appears to be unstable in the presence of the ligand. Hence, it is possible that the binding of the ligand may have caused a rearrangement of the structure, leading to a change in the unwinding of 310 helix. Findings reported in this study further enhance the understanding of Mycobacterium tuberculosis -DnaN and also give a lead to the development of potent tuberculosis drugs.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Polimerasa III/metabolismo , Mycobacterium tuberculosis/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Sitios de Unión , ADN Polimerasa III/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Simulación de Dinámica Molecular , Análisis de Componente Principal , Estructura Terciaria de Proteína , Termodinámica , Tuberculosis/tratamiento farmacológico
15.
J Mol Biol ; 428(14): 2805-13, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27320386

RESUMEN

Archaeal family-D DNA polymerases (Pol-D) comprise a small (DP1) proofreading subunit and a large (DP2) polymerase subunit. Pol-D is one of the least studied polymerase families, and this publication investigates the enzyme from Archaeoglobus fulgidus (Afu Pol-D). The C-terminal region of DP2 contains two conserved cysteine clusters, and their roles are investigated using site-directed mutagenesis. The cluster nearest the C terminus is essential for polymerase activity, and the cysteines are shown to serve as ligands for a single, critical Zn(2+) ion. The cysteines farthest from the C terminal were not required for activity, and a role for these amino acids has yet to be defined. Additionally, it is shown that Afu Pol-D activity is slowed by the template strand hypoxanthine, extending previous results that demonstrated inhibition by uracil. Hypoxanthine was a weaker inhibitor than uracil. Investigations with isolated DP2, which has a measurable polymerase activity, localised the deaminated base binding site to this subunit. Uracil and hypoxanthine slowed Afu Pol-D "in trans", that is, a copied DNA strand could be inhibited by a deaminated base in the alternate strand of a replication fork. The error rate of Afu Pol-D, measured in vitro, was 0.24×10(-5), typical for a polymerase that has been proposed to carry out genome replication in the Archaea. Deleting the 3'-5' proofreading exonuclease activity reduced fidelity twofold. The results presented in this publication considerably increase our knowledge of Pol-D.


Asunto(s)
Proteínas Arqueales/antagonistas & inhibidores , Archaeoglobus fulgidus/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , ADN Polimerasa III/antagonistas & inhibidores , Hipoxantina/farmacología , Uracilo/farmacología , Zinc/metabolismo , Sitios de Unión/genética , Cisteína/genética , Replicación del ADN/genética , ADN de Archaea/genética , Mutagénesis Sitio-Dirigida/métodos
16.
Exp Cell Res ; 342(2): 145-58, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26968636

RESUMEN

Although chemical inhibition of the DNA damage response (DDR) in cancer cells triggers cell death, it is not clear if the fork blockade achieved with inhibitors that neutralise proteins of the replisome is sufficient on its own to overcome the DDR. Monoclonal antibodies to PCNA, which block the DNA elongation process in vitro, have been developed. When these antibodies were transduced into cancer cells, they are able to inhibit the incorporation of nucleoside analogues. When co-delivered with anti-PCNA siRNA, the cells were flattened and the size of their nuclei increased by up to 3-fold, prior to cell death. Analysis of these nuclei by super-resolution microscopy revealed the presence of large numbers of phosphorylated histone H2AX foci. A senescence-like phenotype of the transduced cells was also observed upon delivery of the corresponding Fab molecules or following PCNA gene disruption or when the Fab fragment of an antibody that neutralises DNA polymerase alpha was used. Primary melanoma cells and leukaemia cells that are resistant to chemical inhibitors were similarly affected by these antibody treatments. These results demonstrate that transduced antibodies can trigger a lethal DNA replication stress, which kills cancer cells by abolishing the biological activity of several constituents of the replisome.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/genética , Animales , Roturas del ADN de Doble Cadena , ADN Polimerasa III/antagonistas & inhibidores , ADN de Neoplasias/metabolismo , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Técnicas de Silenciamiento del Gen , Células HeLa , Histonas/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/farmacología , Ratones Endogámicos BALB C , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/inmunología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estrés Fisiológico
17.
Nucleic Acids Res ; 44(7): 3176-89, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26792895

RESUMEN

Successful and accurate completion of the replication of damage-containing DNA requires mainly recombination and RAD18-dependent DNA damage tolerance pathways. RAD18 governs at least two distinct mechanisms: translesion synthesis (TLS) and template switching (TS)-dependent pathways. Whereas TS is mainly error-free, TLS can work in an error-prone manner and, as such, the regulation of these pathways requires tight control to prevent DNA errors and potentially oncogenic transformation and tumorigenesis. In humans, the PCNA-associated recombination inhibitor (PARI) protein has recently been shown to inhibit homologous recombination (HR) events. Here, we describe a biochemical mechanism in which PARI functions as an HR regulator after replication fork stalling and during double-strand break repair. In our reconstituted biochemical system, we show that PARI inhibits DNA repair synthesis during recombination events in a PCNA interaction-dependent way but independently of its UvrD-like helicase domain. In accordance, we demonstrate that PARI inhibits HR in vivo, and its knockdown suppresses the UV sensitivity of RAD18-depleted cells. Our data reveal a novel human regulatory mechanism that limits the extent of HR and represents a new potential target for anticancer therapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Reparación del ADN por Recombinación , Secuencias de Aminoácidos , ADN/biosíntesis , ADN Polimerasa III/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Células HEK293 , Humanos , Ubiquitina-Proteína Ligasas/fisiología , Rayos Ultravioleta
18.
DNA Repair (Amst) ; 35: 90-105, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26519823

RESUMEN

Cadmium (Cd) is a carcinogenic and neurotoxic environmental pollutant. Among the proposed mechanisms for Cd toxic effects, its ability to promote oxidative stress and to inhibit, in vitro, the activities of some Base Excision DNA Repair (BER) enzymes, such as hOGG1, XRCC1 and APE1, have been already established. However, the molecular mechanisms at the basis of these processes are largely unknown especially at sub-lethal doses of Cd and no information is available on the effect of Cd on the expression levels of BER enzymes. Here, we show that non-toxic treatment of neuronal cell lines, with pro-mitogenic doses of Cd, promotes a significant time- and dose-dependent down-regulation of DNA polymerase δ (POLD1) expression through a transcriptional mechanism with a modest effect on Polß, XRCC1 and APE1. We further elucidated that the observed transcriptional repression on Polδ is acted by through competition by activated p53 on Sp1 at POLD1 promoter and by a squelching effect. We further proved the positive effect of Sp1 not only on POLD1 expression but also on Polß, XRCC1 and APE1 expression, suggesting that Sp1 has pleiotropic effects on the whole BER pathway. Our results indicated that Cd-mediated impairment of BER pathway, besides acting on the enzymatic functions of some key proteins, is also exerted at the gene expression level of Polδ by acting on the p53-Sp1 regulatory axis. These data may explain not only the Cd-induced neurotoxic effects but also the potential carcinogenicity of this heavy metal.


Asunto(s)
Cadmio/toxicidad , Carcinógenos/toxicidad , ADN Polimerasa III/antagonistas & inhibidores , Contaminantes Ambientales/toxicidad , Neurotoxinas/toxicidad , Factor de Transcripción Sp1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Cadmio/farmacología , Carcinogénesis/inducido químicamente , Dominio Catalítico/genética , Línea Celular Tumoral , ADN Polimerasa III/genética , ADN Polimerasa beta/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Humanos , Neuronas/efectos de los fármacos , Neuronas/enzimología , Regiones Promotoras Genéticas , Activación Transcripcional/efectos de los fármacos , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
19.
Sci Rep ; 5: 13637, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26323712

RESUMEN

As the persistent resistance of Staphylococcus aureus to available antibiotics is associated with high infection incidence, mortality rate and treatment cost, novel antibacterial agents with innovative therapeutic targets must be developed. 3,3'-(4-Nitrobenzylidene)-bis-(4-hydroxycoumarin) (NBH), a dicoumarin derivative, was reported to exert antibacterial activity. This study investigated the underlying mechanisms of in vivo and in vitro activities of NBH against S. aureus. NBH exerted bactericidal effects against the tested S. aureus and Staphylococcus epidermidis strains in vitro, with low cytotoxicity and resistance and high plasma stability. NBH also exhibited therapeutic effects in vivo on septicaemic mice. Results of molecular docking and analysis on morphological change, DNA production and polymerase inhibition suggested that DNA polymerase could be the target of NBH. These findings indicated that dicoumarin derivatives, which interfere with DNA replication, could be developed as a potential agent against S. aureus, particularly methicillin-resistant strains.


Asunto(s)
4-Hidroxicumarinas/química , Antibacterianos/química , ADN Polimerasa III/antagonistas & inhibidores , 4-Hidroxicumarinas/uso terapéutico , 4-Hidroxicumarinas/toxicidad , Animales , Antibacterianos/uso terapéutico , Antibacterianos/toxicidad , Sitios de Unión , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN Polimerasa III/metabolismo , Replicación del ADN/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Sepsis/tratamiento farmacológico , Sepsis/veterinaria , Staphylococcus aureus/efectos de los fármacos , Staphylococcus epidermidis/efectos de los fármacos
20.
BMC Cancer ; 15: 325, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25924900

RESUMEN

BACKGROUND: Hepatocyte carcinoma (HCC) is one of the most common malignancies worldwide. Despite many achievements in diagnosis and treatment, HCC mortality remains high due to the malignant nature of the disease. Novel approaches, especially for targeted therapy, are being extensively explored. Gene therapy is ideal for such purpose for its specific expression of exogenous genes in HCC cells driven by tissue-specific promoter. However strategies based on correction of mutations or altered expressions of genes responsible for the development/progression of HCC have limitations because these aberrant molecules are not presented in all cancerous cells. In the current work, we adopted a novel strategy by targeting the DNA replication step which is essential for proliferation of every cancer cell. METHODS: A recombinant adenovirus with alpha fetoprotein (AFP) promoter-controlled expressions of artificial microRNAs targeting DNA polymerases α, δ, ε and recombinant active Caspase 3, namely Ad/AFP-Casp-AFP-amiR, was constructed. RESULTS: The artificial microRNAs could efficiently inhibit the expression of the target polymerases in AFP-positive HCC cells at both RNA and protein levels, and HCC cells treated with the recombinant virus Ad/AFP-Casp-AFP-amiR exhibited significant G0/1 phase arrest. The proliferation of HCC cells were significantly inhibited by Ad/AFP-Casp-AFP-amiR with increased apoptosis. On the contrary, the recombinant adenovirus Ad/AFP-Casp-AFP-amiR did not inhibit the expression of DNA polymerases α, δ or ε in AFP-negative human normal liver cell HL7702, and showed no effect on the cell cycle progression, proliferation or apoptosis. CONCLUSIONS: Inhibition of DNA polymerases α, δ and ε by AFP promoter-driven artificial microRNAs may lead to effective growth arrest of AFP-positive HCC cells, which may represent a novel strategy for gene therapy by targeting the genes that are essential for the growth/proliferation of cancer cells, avoiding the limitations set by any of the individually altered gene.


Asunto(s)
Carcinoma Hepatocelular/genética , ADN Polimerasa III/genética , ADN Polimerasa II/genética , ADN Polimerasa I/genética , Neoplasias Hepáticas/genética , Adenoviridae/genética , Animales , Apoptosis/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Línea Celular Tumoral , ADN Polimerasa I/antagonistas & inhibidores , ADN Polimerasa II/antagonistas & inhibidores , ADN Polimerasa III/antagonistas & inhibidores , Terapia Genética , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Ratones , MicroARNs/genética , Terapia Molecular Dirigida , Regiones Promotoras Genéticas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA