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











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 5966, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34645815

RESUMO

The BRCA2 tumor suppressor protects genome integrity by promoting homologous recombination-based repair of DNA breaks, stability of stalled DNA replication forks and DNA damage-induced cell cycle checkpoints. BRCA2 deficient cells display the radio-resistant DNA synthesis (RDS) phenotype, however the mechanism has remained elusive. Here we show that cells without BRCA2 are unable to sufficiently restrain DNA replication fork progression after DNA damage, and the underrestrained fork progression is due primarily to Primase-Polymerase (PRIMPOL)-mediated repriming of DNA synthesis downstream of lesions, leaving behind single-stranded DNA gaps. Moreover, we find that BRCA2 associates with the essential DNA replication factor MCM10 and this association suppresses PRIMPOL-mediated repriming and ssDNA gap formation, while having no impact on the stability of stalled replication forks. Our findings establish an important function for BRCA2, provide insights into replication fork control during the DNA damage response, and may have implications in tumor suppression and therapy response.


Assuntos
Proteína BRCA2/genética , DNA Primase/genética , DNA de Neoplasias/genética , DNA de Cadeia Simples/genética , DNA Polimerase Dirigida por DNA/genética , Proteínas de Manutenção de Minicromossomo/genética , Enzimas Multifuncionais/genética , Reparo de DNA por Recombinação , Proteína BRCA2/antagonistas & inibidores , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Dano ao DNA , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Primase/antagonistas & inibidores , DNA Primase/metabolismo , Replicação do DNA , DNA de Neoplasias/metabolismo , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Proteínas de Manutenção de Minicromossomo/antagonistas & inibidores , Proteínas de Manutenção de Minicromossomo/metabolismo , Enzimas Multifuncionais/antagonistas & inibidores , Enzimas Multifuncionais/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
ACS Chem Biol ; 14(9): 1904-1912, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31479243

RESUMO

Nucleoside analogues are widely used in clinical practice as chemotherapy drugs. Arabinose nucleoside derivatives such as fludarabine are effective in the treatment of patients with acute and chronic leukemias and non-Hodgkin's lymphomas. Although nucleoside analogues are generally known to function by inhibiting DNA synthesis in rapidly proliferating cells, the identity of their in vivo targets and mechanism of action are often not known in molecular detail. Here we provide a structural basis for arabinose nucleotide-mediated inhibition of human primase, the DNA-dependent RNA polymerase responsible for initiation of DNA synthesis in DNA replication. Our data suggest ways in which the chemical structure of fludarabine could be modified to improve its specificity and affinity toward primase, possibly leading to less toxic and more effective therapeutic agents.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Antineoplásicos/química , Antivirais/química , DNA Primase/antagonistas & inibidores , Vidarabina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Antineoplásicos/metabolismo , Antivirais/metabolismo , Domínio Catalítico , Cristalografia por Raios X , DNA Primase/química , DNA Primase/metabolismo , Ensaios Enzimáticos , Humanos , Ligação Proteica , Vidarabina/química , Vidarabina/metabolismo
3.
Antiviral Res ; 168: 114-120, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31152759

RESUMO

Genotypic diagnosis of HSV drug resistance can be performed routinely in a clinically relevant time. Nevertheless, data about HSV mutations (polymorphism or resistance) is not exhaustive which hinders the interpretation of such tests. The UL23, UL30, and UL5 genes are of greatest interest as these encode, respectively, thymidine kinase, DNA polymerase, and helicase, which, if mutated may affect the effectiveness of acyclovir, foscarnet, cidofovir, and helicase-primase inhibitors. The present study aimed to extensively characterize UL23, UL30, and UL5 genes. A total of 239 clinical HSV1 recovered from patients admitted to the hematology departments of the Lyon teaching hospitals were included in this single-center retrospective study. Drug resistance was evaluated using the neutral red dye-uptake assay, and sequencing using the Sanger method. Additional information on HSV1 natural polymorphism and resistance is now available. Twenty-two amino acid substitutions related to polymorphism were described on UL23 (E43A, L50M, L68R, Q109K, A133V, A136N, S150L, D258N, S263L, P280S, N301S, A316S, M322L, I326V, D330A, D338H, Q342H, T344I, Q349R, V352L, R370W, E371D), and 6 amino acid substitutions on UL30 (G641R, G645D, E649G, G679D, R681L, I966M). Moreover, the UL23 substitution L242P was added to ACV resistance-related mutations. There were 12 substitutions on UL23 (A37S, V70M, S74L, H151N, P154S, P155Q, L159R, E225L, Y248H, Q270R, N303Y, M372I), and 8 on UL5 (L49I, L138V, S173L, A280T, A575V, V600A, A602T, D862N) that remain of unclear significance with regards to drug resistance. The mean (±standard deviation, SD) number of natural polymorphisms in UL23 was 2.53 (±2.55), in UL30 it was 0.83 (±1.02), and in UL5 it was 5.00 (±1.59) There was no association between HSV1 phenotype and the frequency of substitutions. The results reported herein provide valuable new information concerning HSV1 mutations that will assist the interpretation of genotypic assays.


Assuntos
Herpes Simples/microbiologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/isolamento & purificação , Proteínas Virais/genética , Antivirais/farmacologia , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , DNA Primase/antagonistas & inibidores , DNA Primase/genética , DNA Polimerase Dirigida por DNA/genética , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Inibidores Enzimáticos/farmacologia , Exodesoxirribonucleases/antagonistas & inibidores , Exodesoxirribonucleases/genética , Feminino , Genótipo , Hematologia , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/classificação , Humanos , Masculino , Mutação , Filogenia , Polimorfismo Genético , Estudos Retrospectivos , Timidina Quinase/antagonistas & inibidores , Timidina Quinase/genética , Proteínas Virais/antagonistas & inibidores
4.
Neoplasia ; 20(11): 1135-1143, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30257222

RESUMO

The phosphoinositide 3-kinase-related kinase ATR is a central regulator of the DNA damage response. Its chemical inhibition eliminates subsets of cancer cells in various tumor types. This effect is caused at least partly by the synthetically lethal relationship between ATR and certain DNA repair genes. In a previous screen using an siRNA library against DNA repair genes, we identified PRIM1, a part of the polymerase α-primase complex, as acting synthetically lethal with ATR. Applying a genetic ATR knock-in model of colorectal cancer cells, we confirmed that PRIM1 depletion inhibited proliferation of ATR-deficient cells and excluded artifacts due to clonal variation using an ATR reexpressing cell clone. We expanded these data by demonstrating in different cell lines that also chemical inhibition of ATR or its main effector kinase CHK1 reduces proliferation upon depletion of PRIM1. Mechanistically, PRIM1 depletion in ATR-deficient cells caused S-phase stasis in the absence of increased DNA damage followed by Wee1-mediated activation of caspase 8 and apoptosis. As PRIM1 inactivation sensitizes cancer cells to ATR and CHK1 inhibitors, mutations in PRIM1 or other components of the polymerase α-primase complex could represent novel targets for individualized tumor therapeutic approaches using ATR/CHK1 inhibitors, as has been previously demonstrated for POLD1, the catalytic subunit of polymerase δ.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Inibidores de Proteínas Quinases/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Histonas/metabolismo , Humanos , Interferência de RNA , RNA Interferente Pequeno/genética , Mutações Sintéticas Letais
5.
J Antibiot (Tokyo) ; 68(3): 153-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25248725

RESUMO

Owing to the rise in drug resistance in tuberculosis combined with the global spread of its causative pathogen, Mycobacterium tuberculosis (Mtb), innovative anti mycobacterial agents are urgently needed. Recently, we developed a novel primase-pyrophosphatase assay and used it to discover inhibitors of an essential Mtb enzyme, primase DnaG (Mtb DnaG), a promising and unexplored potential target for novel antituberculosis chemotherapeutics. Doxorubicin, an anthracycline antibiotic used as an anticancer drug, was found to be a potent inhibitor of Mtb DnaG. In this study, we investigated both inhibition of Mtb DnaG and the inhibitory activity against in vitro growth of Mtb and M. smegmatis (Msm) by other anthracyclines, daunorubicin and idarubicin, as well as by less cytotoxic DNA intercalators: aloe-emodin, rhein and a mitoxantrone derivative. Generally, low-µM inhibition of Mtb DnaG by the anthracyclines was correlated with their low-µM minimum inhibitory concentrations. Aloe-emodin displayed threefold weaker potency than doxorubicin against Mtb DnaG and similar inhibition of Msm (but not Mtb) in the mid-µM range, whereas rhein (a close analog of aloe-emodin) and a di-glucosylated mitoxantrone derivative did not show significant inhibition of Mtb DnaG or antimycobacterial activity. Taken together, these observations strongly suggest that several clinically used anthracyclines and aloe-emodin target mycobacterial primase, setting the stage for a more extensive exploration of this enzyme as an antibacterial target.


Assuntos
Antituberculosos/farmacologia , DNA Primase/antagonistas & inibidores , Substâncias Intercalantes/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/crescimento & desenvolvimento
6.
Expert Opin Ther Targets ; 17(10): 1119-32, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23930666

RESUMO

INTRODUCTION: The Herpesviridae are responsible for debilitating acute and chronic infections, and some members of this family are associated with human cancers. Conventional anti-herpesviral therapy targets the viral DNA polymerase and has been extremely successful; however, the emergence of drug-resistant virus strains, especially in neonates and immunocompromised patients, underscores the need for continued development of anti-herpes drugs. In this article, we explore an alternative target for antiviral therapy, the HSV helicase/primase complex. AREAS COVERED: This review addresses the current state of knowledge of HSV DNA replication and the important roles played by the herpesvirus helicase- primase complex. In the last 10 years several helicase/primase inhibitors (HPIs) have been described, and in this article, we discuss and contrast these new agents with established inhibitors. EXPERT OPINION: The outstanding safety profile of existing nucleoside analogues for α-herpesvirus infection make the development of new therapeutic agents a challenge. Currently used nucleoside analogues exhibit few side effects and have low occurrence of clinically relevant resistance. For HCMV, however, existing drugs have significant toxicity issues and the frequency of drug resistance is high, and no antiviral therapies are available for EBV and KSHV. The development of new anti-herpesvirus drugs is thus well worth pursuing especially for immunocompromised patients and those who develop drug-resistant infections. Although the HPIs are promising, limitations to their development into a successful drug strategy remain.


Assuntos
Antivirais/uso terapêutico , DNA Helicases/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Infecções por Herpesviridae/tratamento farmacológico , Proteínas Virais/antagonistas & inibidores , Animais , Replicação do DNA , DNA Viral , Herpesviridae/fisiologia , Infecções por Herpesviridae/enzimologia , Humanos , Replicação Viral
7.
J Nat Prod ; 76(2): 142-9, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23387796

RESUMO

Three novel hydrazides, geralcins C-E (1-3), were isolated from Streptomyces sp. LMA-545, together with MH-031 and geralcins A and B. This unusual family of compounds was isolated from liquid-state and agar-supported fermentation using Amberlite XAD-16 solid-phase extraction during the cultivation step. The use of such neutral resin during the cultivation step allowed the specific adsorption of microbial secondary metabolites, avoiding any contamination of the crude extracts by the constituents of the culture medium. The trapped compounds were eluted from the resin with methanol, and their structures elucidated using (1)H, (13)C, and (15)N NMR spectroscopic analysis and high-resolution mass spectrometry. Molecular modeling calculations were applied in order to support structural attributions. No antimicrobial, cytotoxic, or DnaG-inhibition activities were detected for geralcins D and E. Geralcin C has no antimicrobial activity but exhibited an IC(50) of 0.8 µM against KB and HCT116 cancer cell lines. Furthermore, geralcin C inhibited the E. coli DnaG primase, a Gram-negative antimicrobial target, with an IC(50) of 0.7 mM.


Assuntos
Hidrazinas/isolamento & purificação , Streptomyces/química , 4-Butirolactona/análogos & derivados , DNA Primase/antagonistas & inibidores , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Escherichia coli/efeitos dos fármacos , Células HCT116 , Humanos , Hidrazinas/química , Hidrazinas/farmacologia , Células KB , Testes de Sensibilidade Microbiana , Ressonância Magnética Nuclear Biomolecular
8.
Drug Resist Updat ; 14(1): 45-51, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21183396

RESUMO

A new class of chemical inhibitors has been discovered that interferes with the process of herpesvirus DNA replication. To date, the majority of useful herpesvirus antivirals are nucleoside analogues that block herpesvirus DNA replication by targeting the DNA polymerase. The new helicase-primase inhibitors (HPI) target a different enzyme complex that is also essential for herpesvirus DNA replication. This review will place the HPI in the context of previous work on the nucleoside analogues. Several promising highly potent HPI will be described with a particular focus on the identification of drug-resistance mutations. Several HPI have good pharmacological profiles and are now at the outset of phase II clinical trials. Provided there are no safety issues to stop their progress, this new class of compound will be a major advance in the herpesvirus antiviral field. Furthermore, HPI are likely to have a major impact on the therapy and prevention of herpes simplex virus and varicella zoster in both immunocompetent and immunocompromised patients alone or in combination with current nucleoside analogues. The possibility of acquired drug-resistance to HPI will then become an issue of great practical importance.


Assuntos
Antivirais/farmacologia , DNA Helicases/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Animais , Farmacorresistência Viral , Inibidores Enzimáticos/farmacologia , Herpes Genital/tratamento farmacológico , Herpes Genital/enzimologia , Herpes Simples/tratamento farmacológico , Herpes Simples/enzimologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 2/enzimologia , Humanos , Replicação Viral/efeitos dos fármacos
9.
FEBS J ; 276(12): 3382-91, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19438708

RESUMO

The antibiotic heliquinomycin, which inhibits cellular DNA replication at a half-maximal inhibitory concentration (IC(50)) of 1.4-4 microM, was found to inhibit the DNA helicase activity of the human minichromosome maintenance (MCM) 4/6/7 complex at an IC(50) value of 2.4 microM. In contrast, 14 microM heliquinomycin did not inhibit significantly either the DNA helicase activity of the SV40 T antigen and Werner protein or the oligonucleotide displacement activity of human replication protein A. At IC(50) values of 25 and 6.5 microM, heliquinomycin inhibited the RNA priming and DNA polymerization activities, respectively, of human DNA polymerase-alpha/primase. Thus, of the enzymes studied, the MCM4/6/7 complex was the most sensitive to heliquinomycin; this suggests that MCM helicase is one of the main targets of heliquinomycin in vivo. It was observed that heliquinomycin did not inhibit the ATPase activity of the MCM4/6/7 complex to a great extent in the absence of single-stranded DNA. In contrast, heliquinomycin at an IC(50) value of 5.2 microM inhibited the ATPase activity of the MCM4/6/7 complex in the presence of single-stranded DNA. This suggests that heliquinomycin interferes with the interaction of the MCM4/6/7 complex with single-stranded DNA.


Assuntos
Proteínas de Ciclo Celular/metabolismo , DNA Helicases/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/metabolismo , Benzoquinonas/farmacologia , Transporte Biológico/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , DNA Helicases/metabolismo , DNA Polimerase I/antagonistas & inibidores , DNA Polimerase I/metabolismo , DNA Primase/antagonistas & inibidores , DNA Primase/metabolismo , Relação Dose-Resposta a Droga , Exodesoxirribonucleases/antagonistas & inibidores , Exodesoxirribonucleases/metabolismo , Células HeLa , Humanos , Componente 4 do Complexo de Manutenção de Minicromossomo , Componente 6 do Complexo de Manutenção de Minicromossomo , Componente 7 do Complexo de Manutenção de Minicromossomo , Complexos Multiproteicos/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA/genética , RNA/metabolismo , RecQ Helicases/antagonistas & inibidores , RecQ Helicases/metabolismo , Proteína de Replicação A/metabolismo , Compostos de Espiro/farmacologia , Helicase da Síndrome de Werner
10.
Oncol Rep ; 19(2): 535-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202804

RESUMO

The polyamines are aliphatic cations which are present in millimolar concentrations in all mammalian cells, and are required for optimal growth of almost all cell types. In this study, the roles of polyamines in DNA replication in vitro and the mechanism by which polyamines affected DNA replication were examined using simian virus 40 DNA replication system in vitro. We found that polyamines inhibited DNA replication, but it is not clear at which stage this occurs. Spermidine inhibited the DNA cleavage by topoisomerase I at 8.0 mM, but stimulated its activity at 1.0 mM. Spermine also inhibited its activity at 4.0 mM, but stimulated at 1.0 mM. The ssDNA binding activity of replication protein A was slightly affected by polyamines. Polyamines, especially spermine, also significantly reduced polymerase alpha-primase activity at 133 microM. Taken together, we suggest that the major inhibition of SV40 DNA replication may be due to the inhibition of pol alpha-primase activity, and possible roles for polyamines in the initiation process are discussed.


Assuntos
DNA Polimerase I/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Replicação do DNA , Espermidina/fisiologia , Espermina/fisiologia , Clivagem do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA de Cadeia Simples/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Humanos , Poliaminas/farmacologia , Proteína de Replicação A/antagonistas & inibidores , Vírus 40 dos Símios/efeitos dos fármacos , Vírus 40 dos Símios/genética , Espermidina/farmacologia , Espermina/farmacologia , Inibidores da Topoisomerase I
11.
Oncol Rep ; 19(2): 527-34, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202803

RESUMO

p-Tyrosol is a phenolic compound present in different dietary sources that can exert mild antioxidant properties based on in vitro and in vivo studies. In our study, two p-tyrosol derivatives (p-tyrosyl gallate and p-tyrosyl acetate) were synthesized and compared together with p-tyrosol and gallic acid for their cytotoxic activities on human cancer cells. p-Tyrosyl gallate had the most potent cytotoxicity and the major cytotoxic mechanism of its action was studied. We found that in HeLa cells, p-tyrosyl gallate can effectively induce cell cycle arrest during S phase and inhibited in vitro simian virus (SV40 DNA) replication. In addition, p-tyrosyl gallate can inhibit three important functional replication proteins (topoisomerase I, RPA and pol alpha-primase), especially pol alpha-primase. These results suggest that p-tyrosyl gallate-induced cell cycle arrest during S phase correlates with the inhibition of DNA replication. Pol alpha-primase may be the main target molecule. Taken together, we suggest that p-tyrosyl gallate is a strong anticancer drug candidate that warrants further investigation.


Assuntos
Antineoplásicos/farmacologia , DNA Polimerase I/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Replicação do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Álcool Feniletílico/análogos & derivados , Antineoplásicos/síntese química , Antineoplásicos/química , Ciclo Celular/efeitos dos fármacos , DNA de Cadeia Simples/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Células HeLa , Humanos , Álcool Feniletílico/síntese química , Álcool Feniletílico/química , Álcool Feniletílico/farmacologia , Vírus 40 dos Símios/efeitos dos fármacos
12.
Yao Xue Xue Bao ; 41(10): 978-84, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17184117

RESUMO

AIM: To investigate apoptosis induced by 3,3'-diethyl-9-methylthia-carbocyanine iodide (DMTCCI), an inhibitor of DNA primase found in our previous study, and the mechanism of DMTCCI in human myelogenous leukemia HL-60 cells. METHODS: HL-60 cells were cultured in RPMI-1640 medium and treated with different concentrations of DMTCCI. MTT assay was used to detect growth inhibition. Flow cytometry and DNA ladders were used to detect apoptosis. Western blotting was used to observe the expression of survivin, Bcl-xL, Bad, Bax, Bcl-2, caspase-9, caspase-3, caspase-6, PARP, DFF45 and lamin B protein. Caspase-3 activity was measured by ApoAlert Caspase-3 Assay Kit. RESULTS: DMTCCI inhibited proliferation of human leukemia HL-60 cells with IC50 value of 0.24 micromol x L(-1). The results of flow cytometry and DNA ladders showed that DMTCCI could induce apoptosis of HL-60 cells. The expression levels of protein survivin and Bcl-xL were down-regulated, Bad and Bax were up-regulated, while Bcl-2 protein had no change in response to DMTCCI treatment in HL-60 cells. Treatment of HL-60 cells with DMTCCI induced the proteolytic cleavage of caspase-9, caspase-3, caspase-6, PARP, DFF45 and lamin B protein. Caspase-3 activity apparently increased at 3 h and reached a peak at 12 h after exposure to 1 micromol x L(-1) of DMTCCI in HL-60 cells. CONCLUSION: DMTCCI inhibited proliferation and induced apoptosis of human leukemia HL-60 cells. Bcl-2 family proteins, survivin and caspases family proteins might play a role in the apoptosis process induced by DMTCCI.


Assuntos
Apoptose/efeitos dos fármacos , Carbocianinas/farmacologia , DNA Primase/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Fragmentação do DNA/efeitos dos fármacos , Citometria de Fluxo , Células HL-60 , Humanos , Proteínas Inibidoras de Apoptose , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Survivina , Proteína X Associada a bcl-2/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo , Proteína bcl-X/metabolismo
13.
Bioorg Med Chem Lett ; 16(18): 4879-83, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16814545

RESUMO

Infection by human cytomegalovirus (hCMV) remains a potent threat to susceptible people throughout the world. We have discovered a series of imidazolyl-pyrimidine compounds, which were found to be irreversible inhibitors of the hCMV UL70 primase based on results from radiolabeling and SAR studies. Two promising analogs are described that rival ganciclovir and cidofovir in antiviral potency and possess improved cytotoxicity profiles.


Assuntos
Citomegalovirus/efeitos dos fármacos , Citomegalovirus/enzimologia , DNA Primase/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Antivirais/síntese química , Antivirais/química , Antivirais/farmacologia , Medula Óssea/efeitos dos fármacos , Linhagem Celular , DNA Primase/metabolismo , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Pirimidinas/química , Relação Estrutura-Atividade
14.
Curr Pharm Des ; 12(11): 1315-38, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16611118

RESUMO

Helicases are promising antiviral drug targets because their enzymatic activities are essential for viral genome replication, transcription, and translation. Numerous potent inhibitors of helicases encoded by herpes simplex virus, severe acute respiratory syndrome coronavirus, hepatitis C virus, Japanese encephalitis virus, West Nile virus, and human papillomavirus have been recently reported in the scientific literature. Some inhibitors have also been shown to decrease viral replication in cell culture and animal models. This review discusses recent progress in understanding the structure and function of viral helicases to help clarify how these potential antiviral compounds function and to facilitate the design of better inhibitors. The above helicases and all related viral proteins are classified here based on their evolutionary and functional similarities, and the key mechanistic features of each group are noted. All helicases share a common motor function fueled by ATP hydrolysis, but differ in exactly how the motor moves the protein and its cargo on a nucleic acid chain. The helicase inhibitors discussed here influence rates of helicase-catalyzed DNA (or RNA) unwinding by preventing ATP hydrolysis, nucleic acid binding, nucleic acid release, or by disrupting the interaction of a helicase with a required cofactor.


Assuntos
Antivirais/farmacologia , DNA Helicases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , RNA Helicases/antagonistas & inibidores , DNA Helicases/química , DNA Helicases/fisiologia , DNA Primase/antagonistas & inibidores , Papillomaviridae/efeitos dos fármacos , Papillomaviridae/enzimologia , RNA Helicases/química , RNA Helicases/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/enzimologia , Serina Endopeptidases , Simplexvirus/efeitos dos fármacos , Simplexvirus/enzimologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores
15.
BMC Cancer ; 4: 70, 2004 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15456519

RESUMO

BACKGROUND: SV40 DNA replication system is a very useful tool to understand the mechanism of replication, which is a tightly regulated process. Many environmental and cellular factors can induce cell cycle arrest or apoptosis by inhibiting DNA replication. In the course of our search for bioactive metabolites from the marine sponges, psammaplin A was found to have some anticancer properties, the possible mechanism of which was studied. METHODS: Cell viability was determined by Cell Counting Kit-8 (CCK-8) to count living RAW264.7 cells by combining 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium (WST-8) and 1-methoxy-phenazine methosulfate (1-methoxy-PMS). The effect of psammaplin A on DNA replication was carried out in SV40 DNA replication system in vitro. The activities of topoisomerase I and polymerase alpha-primase were measured by the relaxation of superhelical plasmid DNA and the incorporation of [3H]dTTP to the template respectively. The ssDNA binding activity of RPA was assessed by Gel Mobility Shift Assay (GMSA). RESULTS: We have found that psammaplin A delivers significant cytotoxic activity against the RAW264.7 cell line. It was also found that psammaplin A could substantially inhibit SV40 DNA replication in vitro, in which polymerase alpha-primase is one of its main targets. CONCLUSION: Taken together, we suggest that psammaplin A-induced cytotoxicity may correlate with its inhibition on DNA replication. Psammaplin A has the potential to be developed as an anticancer drug.


Assuntos
DNA Polimerase I/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Replicação do DNA/efeitos dos fármacos , Dissulfetos/farmacologia , Poríferos/química , Inibidores da Topoisomerase II , Tirosina/análogos & derivados , Tirosina/farmacologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Vírus 40 dos Símios/efeitos dos fármacos , Vírus 40 dos Símios/fisiologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
16.
World J Gastroenterol ; 10(4): 514-20, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14966908

RESUMO

AIM: To evaluate the effects of 3,3'-diethyl-9-methylthia-carbocyanine iodide (DMTCCI) on DNA primase activity and on apoptosis of human hepatocellular carcinoma BEL-7402 cells. METHODS: DNA primase assay was used to investigate DNA primase activity. MTT assay was applied to determine cell proliferation. Flow cytometric analysis, transmission electron microscopy, DNA fragmentation assay were performed to detect DMTCCI-induced apoptosis. Expression levels of p53, Bcl-2, Bcl-xL, Bad, Bax, survivin, Caspase-3 and poly (ADP-ribose) polymerase (PARP) were evaluated by immunoblot analysis. Caspase-3 activity was assessed with ApoAlert Caspase-3 colorimetric assay kit. RESULTS: DMTCCI had inhibitory effects on eukaryotic DNA primase activity with IC(50) value of 162.2 nmol/L. It also inhibited proliferation of human hepatocellular carcinoma BEL-7402 cells with IC(50) value of 2.09 micromol/L. Furthermore, DMTCCI-induced BEL-7402 cell apoptosis was confirmed by DNA fragmentation (DNA ladders and sub-G1 formation) and transmission electron microscopy (apoptotic bodies formation). During the induction of apoptosis, expression of Bcl-2, Bcl-xL and survivin was decreased, and that of p53, Bad and Bax was increased. Caspase-3 was activated and poly (ADP-ribose) polymerase (PARP) was cleaved in BEL-7402 cells treated with DMTCCI. CONCLUSION: The present data suggest that DMTCCI has inhibitory effects on eukaryotic DNA primase and can induce apoptosis of BEL-7402 cells. The modulation of expression of p53 and Bcl-2 family proteins, and activation of Caspase-3 might be involved in the induction of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Carbocianinas/farmacologia , Carcinoma Hepatocelular , DNA Primase/antagonistas & inibidores , Neoplasias Hepáticas , Apoptose/fisiologia , Carbocianinas/química , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , DNA Primase/metabolismo , Citometria de Fluxo , Humanos , Proteínas Inibidoras de Apoptose , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias , Nucleossomos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Survivina , Proteína Supressora de Tumor p53/metabolismo
17.
FASEB J ; 14(10): 1318-24, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10877824

RESUMO

p12(DOC-1) is a growth suppressor identified and isolated from normal keratinocytes. Ectopic expression of p12(DOC-1) in squamous carcinoma cells led to the reversion of in vitro transformation phenotypes including anchorage independence, doubling time, and morphology. Here we report that p12(DOC-1) associates with DNA polymerase alpha/primase (pol-alpha:primase) in vitro and in cells. The pol-alpha:primase binding domain in p12(DOC-1) is mapped to the amino-terminal six amino acid (MSYKPN). The biological effect of p12(DOC-1) on pol-alpha:primase was examined using in vitro DNA replication assays. Using the SV40 DNA replication assay, p12(DOC-1) suppresses DNA replication, leveling at approximately 50%. Similar results were obtained using the M13 single-stranded DNA synthesis assay. Analysis of the DNA replication products revealed that p12(DOC-1) affects the initiation step, not the elongation phase. The p12(DOC-1) suppression of DNA replication is likely to be mediated either by a direct inhibitory effect on pol-alpha:primase or by its effect on cyclin-dependent kinase 2 (CDK2), a recently identified p12(DOC-1)-associated protein known to stimulate DNA replication by phosphorylating pol-alpha:primase. p12(DOC-1) suppresses CDK2-mediated phosphorylation of pol-alpha:primase. These data support a role of p12(DOC-1) as a regulator of DNA replication by direct inhibition of pol-alpha:primase or by negatively regulating the CDK2-mediated phosphorylation of pol-alpha:primase.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Inibidores do Crescimento/metabolismo , Proteínas/metabolismo , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , DNA Polimerase I/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Replicação do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores do Crescimento/química , Inibidores do Crescimento/farmacologia , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas/química , Proteínas/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
18.
Biochemistry ; 38(31): 10126-34, 1999 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-10433721

RESUMO

We examined the effects of Mn(2+) on eukaryotic DNA primase both in the presence and absence of 5 mM Mg(2+). In the absence of Mg(2+), Mn(2+)-supported primase activity to a level 4-fold greater than that obtained with Mg(2+) alone, and adding low levels of Mn(2+) (100 microM) to assays containing 5 mM Mg(2+) greatly stimulated primase. Increased activity was primarily due to more efficient utilization of NTPs, as reflected in a lower K(M) for NTPs. Under conditions of saturating NTPs, Mn(2+) had minimal effects on both the rate of initiation (i.e., dinucleotide synthesis) and processivity. The effects of Mn(2+) involve multiple metal binding sites on primase and may involve both the catalytic p49 subunit as well as the p58 subunit. Physiological levels of salt can inhibit primase activity due to the presence of an anion binding site and low levels of Mn(2+) significantly decreased this salt sensitivity. The implications of these results with respect to the biological role of primase are discussed.


Assuntos
DNA Primase/antagonistas & inibidores , DNA Primase/metabolismo , Manganês/química , Iniciação Traducional da Cadeia Peptídica , Ânions , Sequência de Bases , Catálise , DNA Primase/química , Fosfatos de Dinucleosídeos/metabolismo , Fosfatos de Dinucleosídeos/fisiologia , Ativação Enzimática/fisiologia , Guanosina Trifosfato/metabolismo , Humanos , Magnésio/química , Magnésio/fisiologia , Manganês/metabolismo , Manganês/fisiologia , Dados de Sequência Molecular , Elongação Traducional da Cadeia Peptídica/fisiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Moldes Genéticos
19.
Biochem Mol Biol Int ; 47(5): 795-801, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10365250

RESUMO

The effects of persimmon extract (Diospyros kaki) and related polyphenols on eukaryotic DNA polymerase alpha were examined. It was found that persimmon extract, epigallocatechin gallate, and epicatechin gallate strongly inhibited the activity of DNA polymerase alpha purified from calf thymus. Among these polyphenols, persimmon extract had the most potent effect on DNA polymerase alpha activity and the concentration of persimmon extract producing 50% inhibition of the activity was 0.191 microM. Persimmon extract showed a weaker effect on DNA polymerase beta and slightly inhibited primase and DNA polymerase I. The inhibition of DNA polymerase alpha by persimmon extract was competitive with the template-primer and noncompetitive with dTTP substrate. The Ki value of DNA polymerase alpha for persimmon extract was estimated to be 70 nM. Moreover, persimmon extract inhibited [3H]thymidine incorporation of human peripheral lymphocyte cells stimulated by PHA.


Assuntos
DNA Polimerase I/antagonistas & inibidores , Flavonoides , Fenóis/farmacologia , Proteínas de Plantas/farmacologia , Polímeros/farmacologia , DNA Polimerase I/sangue , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/sangue , DNA Primase/antagonistas & inibidores , DNA Primase/sangue , Células Eucarióticas/metabolismo , Humanos , Concentração Inibidora 50 , Cinética , Polifenóis
20.
Biochem J ; 337 ( Pt 3): 585-90, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9895305

RESUMO

We demonstrate that l-ATP is recognized by some enzymes that are involved in the synthesis of nucleotides and nucleic acids. l-ATP, as well as its natural d-enantiomer, acts as a phosphate donor in the reaction catalysed by human deoxycytidine kinase, whereas it is not recognized by either enantioselective human thymidine kinase or non-enantioselective herpes virus thymidine kinase. l-ATP strongly inhibits (Ki 80 microM) the synthesis of RNA primers catalysed by DNA primase associated with human DNA polymerase alpha, whereas RNA synthesis catalysed by Escherichia coli RNA polymerase is completely unaffected. Moreover, l-ATP competitively inhibits ATP-dependent T4 DNA ligase (Ki 25 microM), suggesting that it interacts with the ATP-binding site of the enzyme. Kinetic studies demonstrated that l-ATP cannot be used as a cofactor in the ligase-catalysed joining reaction. On the other hand, l-AMP is used by T4 DNA ligase to catalyse the reverse reaction, even though a high level of intermediate circular nicked DNA molecules accumulates. Our results suggest that a lack of enantioselectivity of enzymes is more common than was believed a few years ago, and, given the absence of selective constraints against l-nucleosides in Nature, this may depend on chance more than on evolutionary strategy.


Assuntos
Trifosfato de Adenosina/química , DNA/biossíntese , Monofosfato de Adenosina/química , DNA Ligases/química , DNA Primase/antagonistas & inibidores , DNA Primase/química , RNA Polimerases Dirigidas por DNA/química , Desoxicitidina Quinase/química , Escherichia coli/enzimologia , Humanos , Cinética , Simplexvirus/enzimologia , Estereoisomerismo , Timidina Quinase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA