Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
DNA Repair (Amst) ; 21: 165-70, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24986640

RESUMO

The ability of the radiomimetic anti-tumor enediyne C-1027 to induce DNA inter-strand crosslinks (ICLs), in addition to the expected DNA strand breaks, is unique among traditional DNA targeted cancer therapies. Importantly, radiation therapy and most radiomimetic drugs have diminished effect in hypoxic environments due to decreased induction of DNA strand breaks, which is an oxygen requiring process. However, C-1027's induction of ICLs is enhanced under hypoxia and it is actually more potent against hypoxic cells, overcoming this common tumor resistance mechanism. In this study, an analog of C-1027, 20'-deschloro-C-1027 was examined for its ability to induce DNA ICLs under hypoxic conditions. Deschloro-induced ICLs were detected under hypoxic cell-free conditions, with a concomitant reduction in the induction of DNA strand breaks. In cells deschloro behaved similarly, inducing cellular ICLs under hypoxic conditions with a reduction in DNA breaks. The cytotoxicity of deschloro treatment was similar in normoxic and hypoxic cells, suggesting that the ICL induction allows deschloro to retain its cytotoxic activity under hypoxia. It appears that rational engineering of the C-1027 family of radiomimetics holds promise toward overcoming the radioresistance associated with the hypoxic environment associated with solid tumors.


Assuntos
Aminoglicosídeos/toxicidade , Antibióticos Antineoplásicos/toxicidade , Reagentes de Ligações Cruzadas/toxicidade , Quebras de DNA , Enedi-Inos/toxicidade , Tolerância a Radiação , Aminoglicosídeos/uso terapêutico , Hipóxia Celular , Enedi-Inos/uso terapêutico , Células HCT116 , Humanos
2.
Bioorg Med Chem ; 20(15): 4744-50, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22748380

RESUMO

C1027 is a potent antitumor agent that damages DNA. It has the unusual ability to produce double strand breaks and interstrand cross-links (ICLs) intracellularly, which enable it to initiate concurrent ataxia-telangiestasia mutated (ATM) and Rad-3 related (ATR) independent damage responses. The latter form of damage is not well characterized. We have examined the effect of DNA sequence on C1027 reactivity and found it to be more diverse than previously thought. In addition, analysis of the chemical stability of ICLs suggests that they result from reaction with the deoxyribose ring on one strand but direct addition to a nucleobase on the opposite strand.


Assuntos
Aminoglicosídeos/farmacologia , DNA/efeitos dos fármacos , Enedi-Inos/farmacologia , Hidrogênio/química , Aminoglicosídeos/química , Dano ao DNA , Enedi-Inos/química , Estrutura Molecular , Plasmídeos
3.
Biotechniques ; 46(2): 127-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19317657

RESUMO

We report an extraction-free assay in which the same slot blot membrane can be used to assess total genomic DNA damage (i.e., crosslinks or strand breaks) and DNA replication (i.e., bromodeoxyuridine incorporation) or protein levels (i.e., gamma-H2AX). 14C-thymidine radiolabeling of HCT116 cells loaded directly on a Hybond N+ membrane slot blot enables the quantitation of DNA interstrand crosslinks and DNA breaks, while bromodeoxyuridine incorporation or levels of gamma-H2AX can be assessed by incubating blots with primary monoclonal antibodies followed by detection with horseradish peroxidase (HRP) secondary antibodies. Uniform Ponceau staining of all samples on the membrane indicates that protein binding to the membrane is independent of DNA damage or elution. The use of a single membrane to assay levels of DNA damage and concomitant changes in damage response proteins or replication allows the direct quantitation of diverse parameters under identical conditions.


Assuntos
Dano ao DNA , Replicação do DNA/genética , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , DNA/química , DNA/genética , Mapeamento de Nucleotídeos/métodos , Manejo de Espécimes/métodos , Extração em Fase Sólida
4.
Cancer Res ; 69(2): 593-8, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147573

RESUMO

The hypoxic nature of cells within solid tumors limits the efficacy of anticancer therapies such as ionizing radiation and conventional radiomimetics because their mechanisms require oxygen to induce lethal DNA breaks. For example, the conventional radiomimetic enediyne neocarzinostatin is 4-fold less cytotoxic to cells maintained in low oxygen (hypoxic) compared with normoxic conditions. By contrast, the enediyne C-1027 was nearly 3-fold more cytotoxic to hypoxic than to normoxic cells. Like other radiomimetics, C-1027 induced DNA breaks to a lesser extent in cell-free, or cellular hypoxic, compared with normoxic environments. However, the unique DNA interstrand cross-linking ability of C-1027 was markedly enhanced under the same hypoxic conditions that reduced its DNA break induction. Although the unique chemistry of C-1027 allows it to concurrently generate both DNA breaks and cross-links in normoxic cells, a low oxygen environment represses the former and promotes the latter. Thus, treatment with C-1027 offers a facile approach for overcoming the radioresistance associated with poorly oxygenated cells.


Assuntos
Aminoglicosídeos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Dano ao DNA , Enedi-Inos/farmacologia , Hipóxia Celular/fisiologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , DNA Super-Helicoidal/efeitos dos fármacos , Células HCT116 , Histonas/metabolismo , Humanos , Oxigênio/metabolismo , Plasmídeos/efeitos dos fármacos , Plasmídeos/genética , Transdução de Sinais/efeitos dos fármacos
5.
Proc Natl Acad Sci U S A ; 104(45): 17632-7, 2007 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-17978180

RESUMO

The ability of the radiomimetic anticancer enediyne C-1027 to induce ataxia-telangiectasia mutated (ATM) and ATM and Rad3-related (ATR)-independent damage responses was discovered to reside in its unique ability to concurrently generate robust amounts of double-strand breaks (DSBs) and interstrand cross-links (ICLs) in cellular DNA. Furthermore, a single substitution to the chromophore's benzoxazolinate moiety shifted DNA damage to primarily ICLs and an ATR- but not ATM-dependent damage response. In contrast, single substitutions of the chromophore's beta-amino acid component shifted DNA damage to primarily DSBs, consistent with its induction of conventional ATM-dependent damage responses of the type generated by ionizing radiation and other radiomimetics. Thus, phosphatidylinositol 3-kinase-like protein kinase regulation of DNA damage responses is dictated by the relative proportions of DSBs and ICLs.


Assuntos
Reagentes de Ligações Cruzadas/síntese química , Dano ao DNA , Enedi-Inos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Ataxia Telangiectasia/genética , DNA/química , DNA Viral/genética , Enedi-Inos/síntese química , Regulação da Expressão Gênica , Humanos , Cinética , Mutação , Conformação de Ácido Nucleico , Plasmídeos , Vírus 40 dos Símios/genética
6.
Cancer Res ; 67(2): 773-81, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234789

RESUMO

The radiomimetic enediyne C-1027 induces almost exclusively DNA double-strand breaks (DSB) and is extremely cytotoxic. Unique among radiomimetics, ataxia-telangiectasia mutated (ATM) is dispensable for cellular responses to C-1027-induced DNA damage. This study explores the biological activity of three recently bioengineered C-1027 analogues: 7''-desmethyl-C-1027 (desmethyl), 20'-deschloro-C-1027 (deschloro), and 22'-deshydroxy-C-1027 (deshydroxy). Each compound maintains the characteristic ability of radiomimetics to cleave DNA in cell-free systems, varying in activity from 2-fold (deschloro) to 55-fold (desmethyl) less than C-1027. The induction of cellular DNA breaks based on pulsed field gel electrophoresis, comet analysis, and gammaH2AX activation was in the same rank order as cell-free DNA break induction, although the amount of breaks induced by desmethyl is greatly reduced compared with the other analogues. Despite the disparity in inducing DNA DSBs, all of the analogues produced G2-M cell cycle arrest and activated DNA DSB damage response proteins, such as p53-Ser15 and Chk2-Thr68, at concentrations in concordance with their ability to inhibit cell growth. Interestingly, of the three analogues, only the desmethyl-induced DNA damage response was similar to C-1027, as it did not cause hypersensitive cell growth inhibition in the absence of ATM nor require the kinase to phosphorylate p53 or Chk2. These findings show that simple modifications of the chromophore of C-1027 can result in varied induction of, and cellular response to, DNA DSBs.


Assuntos
Aminoglicosídeos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/genética , Dano ao DNA , Proteínas de Ligação a DNA/genética , DNA/efeitos dos fármacos , Enedi-Inos/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/genética , Aminoglicosídeos/química , Antibióticos Antineoplásicos/química , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular , Sistema Livre de Células , DNA/metabolismo , Enedi-Inos/química , Fibroblastos , Humanos , Mutação , Relação Estrutura-Atividade
7.
J Mol Biol ; 361(5): 898-919, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16887143

RESUMO

To understand how bulky adducts might perturb DNA helicase function, three distinct DNA-binding agents were used to determine the effects of DNA alkylation on a DNA helicase. Adozelesin, ecteinascidin 743 (Et743) and hedamycin each possess unique structures and sequence selectivity. They bind to double-stranded DNA and alkylate one strand of the duplex in cis, adding adducts that alter the structure of DNA significantly. The results show that Et743 was the most potent inhibitor of DNA unwinding, followed by adozelesin and hedamycin. Et743 significantly inhibited unwinding, enhanced degradation of DNA, and completely eliminated the ability of the translocating RecBCD enzyme to recognize and respond to the recombination hotspot chi. Unwinding of adozelesin-modified DNA was accompanied by the appearance of unwinding intermediates, consistent with enzyme entrapment or stalling. Further, adozelesin also induced "apparent" chi fragment formation. The combination of enzyme sequestering and pseudo-chi modification of RecBCD, results in biphasic time-courses of DNA unwinding. Hedamycin also reduced RecBCD activity, albeit at increased concentrations of drug relative to either adozelesin or Et743. Remarkably, the hedamycin modification resulted in constitutive activation of the bottom-strand nuclease activity of the enzyme, while leaving the ability of the translocating enzyme to recognize and respond to chi largely intact. Finally, the results show that DNA alkylation does not significantly perturb the allosteric interaction that activates the enzyme for ATP hydrolysis, as the efficiency of ATP utilization for DNA unwinding is affected only marginally. These results taken together present a unique response of RecBCD enzyme to bulky DNA adducts. We correlate these effects with the recently determined crystal structure of the RecBCD holoenzyme bound to DNA.


Assuntos
Antineoplásicos Alquilantes/farmacologia , DNA/química , Escherichia coli/enzimologia , Exodesoxirribonuclease V/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Sítio Alostérico/efeitos dos fármacos , Antraquinonas/química , Antraquinonas/farmacologia , Antineoplásicos Alquilantes/química , Benzofuranos , Catálise/efeitos dos fármacos , Ácidos Cicloexanocarboxílicos/química , Ácidos Cicloexanocarboxílicos/farmacologia , Cicloexenos , DNA Helicases/antagonistas & inibidores , Duocarmicinas , Exodesoxirribonuclease V/metabolismo , Fluorescência , Hidrólise/efeitos dos fármacos , Indóis/química , Indóis/farmacologia , Modelos Moleculares , Conformação de Ácido Nucleico/efeitos dos fármacos , Recombinação Genética/efeitos dos fármacos
8.
J Cell Biol ; 173(4): 533-44, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16717128

RESUMO

The activity of the p53 gene product is regulated by a plethora of posttranslational modifications. An open question is whether such posttranslational changes act redundantly or dependently upon one another. We show that a functional interference between specific acetylated and phosphorylated residues of p53 influences cell fate. Acetylation of lysine 320 (K320) prevents phosphorylation of crucial serines in the NH(2)-terminal region of p53; only allows activation of genes containing high-affinity p53 binding sites, such as p21/WAF; and promotes cell survival after DNA damage. In contrast, acetylation of K373 leads to hyperphosphorylation of p53 NH(2)-terminal residues and enhances the interaction with promoters for which p53 possesses low DNA binding affinity, such as those contained in proapoptotic genes, leading to cell death. Further, acetylation of each of these two lysine clusters differentially regulates the interaction of p53 with coactivators and corepressors and produces distinct gene-expression profiles. By analogy with the "histone code" hypothesis, we propose that the multiple biological activities of p53 are orchestrated and deciphered by different "p53 cassettes," each containing combination patterns of posttranslational modifications and protein-protein interactions.


Assuntos
Ciclo Celular/genética , Regulação da Expressão Gênica/genética , Processamento de Proteína Pós-Traducional/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Sequência de Aminoácidos/fisiologia , Apoptose/genética , Sítios de Ligação/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Genes cdc/fisiologia , Humanos , Lisina/metabolismo , Fosforilação , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Estrutura Terciária de Proteína/fisiologia , Elementos Reguladores de Transcrição/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p53/química
9.
Biochemistry ; 45(11): 3747-54, 2006 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-16533058

RESUMO

Cells lacking the protein kinase ataxia telangiectasia mutated (ATM) have defective responses to DNA double-strand breaks (DSBs), including an inability to activate damage response proteins such as p53. However, we previously showed that cells lacking ATM robustly activate p53 in response to DNA strand breaks induced by the radiomimetic enediyne C-1027. To gain insight into the nature of C-1027-induced ATM-independent damage responses to DNA DSBs, we further examined the molecular mechanisms underlying the cellular response to this unique radiomimetic agent. Like ionizing radiation (IR) and other radiomimetics, breaks induced by C-1027 efficiently activate ATM by phosphorylation at Ser1981, yet unlike other radiomimetics and IR, DNA breaks induced by C-1027 result in normal phosphorylation of p53 and the cell cycle checkpoint kinases (Chk1 and Chk2) in the absence of ATM. In the presence of ATM, but under ATM and Rad3-related kinase (ATR) deficient conditions, C-1027 treatment resulted in a decrease in the level of Chk1 phosphorylation but not in the level of p53 and Chk2 phosphorylation. Only when cells were deficient in both ATM and ATR was there a reduction in the level of phosphorylation of each of these DNA damage response proteins. This reduction was also accompanied by an increased level of cell death in comparison to that of wild-type cells or cells lacking either ATM or ATR. Our findings demonstrate a unique cellular response to C-1027-induced DNA DSBs in that DNA damage response proteins are unaffected by the absence of ATM, as long as ATR is present.


Assuntos
Aminoácidos Cíclicos/farmacologia , Aminoglicosídeos/farmacologia , Dano ao DNA/efeitos dos fármacos , Aminoácidos Cíclicos/metabolismo , Aminoglicosídeos/metabolismo , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Células Cultivadas , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Proteínas de Ligação a DNA/metabolismo , Enedi-Inos , Fibroblastos/metabolismo , Humanos , Raios Infravermelhos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Serina/metabolismo , Treonina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Zinostatina/metabolismo , Zinostatina/farmacologia
10.
Mol Cancer Ther ; 4(8): 1175-85, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16093433

RESUMO

Genotoxic treatments, such as UV light, camptothecin, and adozelesin, stall DNA replication and subsequently generate DNA strand breaks. Typically, DNA breaks are reflected by an increase in ataxia and Rad-related kinase (ATR)-regulated phosphorylation of H2AX (gammaH2AX) and require replication fork movement. This study examined the potential of the monofunctional DNA alkylating agent hedamycin, a powerful inhibitor of DNA replication, to induce DNA strand breaks, phosphorylated H2AX (gammaH2AX) foci, and chromosome aberrations. Hedamycin treatment of HCT116 carcinoma cells resulted in a rapid induction of DNA strand breaks accompanied by increasing H2AX phosphorylation and focalization. Unlike many other treatments that also stall replication, such as UV, camptothecin, and adozelesin, gammaH2AX formation was not suppressed in ATR-compromised cells but actually increased. Similarly, hedamycin induction of gammaH2AX is not dependent on ataxia telangiectasia mutated or DNA-protein kinase, and pretreatment of cells with the phosphatidylinositol 3-kinase-related kinase inhibitor caffeine did not substantially reduce induction of H2AX phosphorylation by hedamycin. Furthermore, the DNA replication inhibitor aphidicolin only modestly depressed hedamycin-induced gammaH2AX formation, indicating that hedamycin-induced DNA double-strand breaks are not dependent on fork progression. In contrast, camptothecin- and adozelesin-induced gammaH2AX was strongly suppressed by aphidicolin. Moreover, after 24 hours following a short-term hedamycin treatment, cells displayed high levels of breaks in interphase nuclear DNA and misjoined chromosomes in metaphase cells. Finally, focalization of a tightly bound form of Ku80 was observed in interphase cells, consistent with the subsequent appearance of chromosomal aberrations via abnormal nonhomologous end joining. Overall, this study has revealed a disparate type of DNA damage response to stalled replication induced by a bulky DNA adduct inducer, hedamycin, that seems not to be highly dependent on ATR or DNA replication.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos Alquilantes/farmacologia , Aberrações Cromossômicas/induzido quimicamente , Replicação do DNA/efeitos dos fármacos , Histonas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Antígenos Nucleares/análise , Antígenos Nucleares/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/farmacologia , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Humanos , Autoantígeno Ku , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Células Tumorais Cultivadas
11.
Cancer Res ; 65(12): 5344-51, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15958582

RESUMO

This study examined the extent of chromosome instability induced in cultured human colon carcinoma HCT116 cells by the antitumor radiomimetic enediyne antibiotic C-1027. Spectral karyotype analysis showed frequent intrachromosomal fusions and fragmentations 26 hours after addition of as little as 0.035 nmol/L C-1027. When the concentration was increased to 0.14 nmol/L C-1027, 92% of cells showed chromosomal aberrations compared with only 2.9% after treatment with an equivalent growth inhibitory dose of ionizing radiation (20 Gy). Thus, chromosome misrejoining was associated to a much greater extent with C-1027-induced than with ionizing radiation-induced cell growth inhibition. Despite these aberrations, a large fraction of C-1027-treated cells progressed into G1. Comet analysis showed that these extensive chromosomal anomalies were not due to increased induction or reduced repair of C-1027-induced compared with ionizing radiation-induced strand breaks. Fluorescence in situ hybridization analysis showed that misrejoining of telomere repeats (i.e., chromosomes joined end to end at their telomeres or fused together after complete loss of telomere sequences) was observed within 26 hours of C-1027 addition. The extreme cytotoxicity of C-1027 may reflect both induction and erroneous repair of DNA double-strand break in the whole genome and/or in subgenomic targets such as telomere sequences.


Assuntos
Aminoglicosídeos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Aberrações Cromossômicas/induzido quimicamente , Telômero/efeitos dos fármacos , Ensaio Cometa , Enedi-Inos , Células HCT116 , Humanos , Cariotipagem , Telômero/fisiologia
12.
J Biol Chem ; 280(10): 9745-51, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15637054

RESUMO

Expression of the antiapoptotic protein survivin is associated with cancer cell viability and drug resistance. Thus, control of its expression in cancer cells has significant consequences for cancer therapeutics. Here we have shown that hedamycin, a GC-rich DNA binding drug, down-regulated survivin expression. Using a series of survivin promoter-luciferase constructs, we have identified an 86-bp GC-rich DNA element (-124 to -39) that mediates the ability of hedamycin to down-regulate survivin expression. Furthermore, both in vivo foot-printing and in vitro gel mobility shift experiments revealed that hedamycin bound to a 21-bp GC-rich DNA element (-115 to -95) in the survivin promoter. This drug-DNA interaction abrogated the binding of Sp-1 or Sp1-like proteins to the 21-bp cis-acting DNA element, and mutagenesis of this region consistently diminished survivin promoter activity. Finally, down-regulation of survivin transcription by hedamycin modulated the viability of cancer cells. These data suggest that abrogation of Sp-1 or Sp1-like protein binding to the 21-bp DNA element in the survivin promoter contributes at least in part to the inhibitory effect of hedamycin on survivin gene transcription. Drug-induced modulation of survivin gene expression may provide novel approaches for cancer therapeutics.


Assuntos
Antraquinonas/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Transcrição Gênica/efeitos dos fármacos , Apoptose , Pareamento de Bases , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo , Pegada de DNA , DNA de Neoplasias/química , DNA de Neoplasias/genética , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Dados de Sequência Molecular , Proteínas de Neoplasias , Reação em Cadeia da Polimerase , Survivina
13.
Mol Pharmacol ; 67(3): 877-82, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15576630

RESUMO

Polyamides are a class of synthetic molecules that exhibit high-affinity, sequence-specific reversible binding in the DNA minor groove but are incapable of inducing DNA damage. In cell-free systems, polyamides have been shown to regulate gene expression by activation, repression, and antirepression. However, effectiveness in cell culture has met with limited success and seems to be cell-dependent. By combining a polyamide with a moiety of a DNA-alkylating agent of the cyclopropylpyrroloindole (CPI) family, a conjugate molecule [polyamide 1-CBI (1-(chloromethyl)-5-hydroxyl-1,2-dihydro-3H-benz[e]indole) conjugate] capable of sequence-specific DNA alkylation was shown to exhibit cellular activity (i.e., cell-growth inhibition and cell-cycle arrest) in mammalian cells. These effects, however, occur at concentrations several orders of magnitude higher than those of its parent CPI agent adozelesin. In addition, 1-CBI is able to interact sequence-specifically with viral DNA and inhibit SV40 DNA replication in infected BSC-1 (African green monkey kidney epithelial) cells, albeit at a greatly reduced ability compared with its CPI parent. On the basis of results from previous studies, we tested whether pretreatment of virus with 1-CBI, compared with direct treatment of infected cells, would enhance its cellular activity. Therefore, using SV40 virions as a model system, we examined the ability of this conjugate molecule to penetrate SV40 virions and damage viral DNA. Our results demonstrate that 1-CBI is able to damage encapsidated SV40 DNA. Both DNA replication and virus production are effectively inhibited in a concentration-dependent manner after infection of BSC-1 cells with 1-CBI-pretreated virions. It is surprising that, unlike in mammalian cells, the relative activity of 1-CBI in SV40 virions is comparable with that of the highly cytotoxic CPI agent adozelesin. Because 1-CBI is able to efficiently penetrate virions and damage DNA, these findings may provide the framework for the development of polyamide-based antiviral agents with enhanced sequence-preference capabilities.


Assuntos
Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Indóis/farmacologia , Pirróis/farmacologia , Vírus 40 dos Símios/genética , DNA Viral/efeitos dos fármacos , DNA Viral/genética , Cinética , Vírus 40 dos Símios/efeitos dos fármacos , Vírion/efeitos dos fármacos , Vírion/genética
14.
Biochemistry ; 43(44): 14228-37, 2004 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-15518573

RESUMO

Ecteinascidin 743 (Et743) is a highly cytotoxic anticancer agent isolated from the squirt Ecteinascidia turbinate, which alkylates DNA in the minor groove at GC-rich sequences resulting in an unusual bending toward the major groove. The ability of Et743 to block DNA replication was studied using the well-established simian virus (SV40) model for mammalian DNA replication in cells and cell-free extracts. Intracellular SV40 DNA isolated from Et743-treated BSC-1 cells was analyzed by native, two-dimensional agarose gel electrophoresis. A low frequency of Et743 adducts detected at 30-100 nM drug concentrations inhibited SV40 origin activity and induced formation of unusual DNA replication intermediates. Under cell-free conditions, only a high Et743 adduct frequency reduced SV40 DNA synthesis. Comparative studies involving related DNA alkylators, tomamycin and saframycin A, revealed inhibition of SV40 DNA replication in cells at concentrations approximately 10 times higher than Et743. Under cell-free conditions tomamycin- or saframycin-A-adducted DNA templates inhibited DNA synthesis similarly to Et743. Et743 appears to be unusual among other alkylators, because its adducts strongly inhibit intracellular SV40 DNA replication but are relatively weak as cis inhibitors as measured under cell-free conditions.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Antivirais/farmacologia , Replicação do DNA/efeitos dos fármacos , DNA Viral/antagonistas & inibidores , DNA Viral/biossíntese , Dioxóis/farmacologia , Isoquinolinas/farmacologia , Vírus 40 dos Símios/genética , Replicação Viral/efeitos dos fármacos , Animais , Benzodiazepinas/farmacologia , Linhagem Celular , Sistema Livre de Células/efeitos dos fármacos , Chlorocebus aethiops , Adutos de DNA/análise , DNA Viral/análise , Exodesoxirribonucleases/química , Células HCT116 , Humanos , Líquido Intracelular/química , Pirróis/farmacologia , Vírus 40 dos Símios/efeitos dos fármacos , Endonucleases Específicas para DNA e RNA de Cadeia Simples/química , Moldes Genéticos , Tetra-Hidroisoquinolinas , Trabectedina , Urocordados
15.
Mol Cancer Ther ; 3(5): 577-85, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15141015

RESUMO

Long-term exposure (72 h) to hedamycin, a monofunctional DNA alkylator of the pluramycin class of antitumor antibiotics, decreased growth of mammalian cells by 50% at subnanomolar concentrations. Short-term treatment (4 h) rapidly reduced DNA synthesis by 50% also at subnanomolar concentrations, but substantially higher levels were needed to block RNA synthesis while protein synthesis even at very high hedamycin concentrations remained unaffected. Hedamycin treatment at concentrations below its growth IC(50) induced only a transient and temporary accumulation of cells in G(2). Somewhat higher concentrations resulted in substantial S-phase arrest, and at increasing concentrations, complete cell cycle arrest in G(1) was observed without the appearance of a sub-G(1) cell population. Neither inhibition of cell growth nor cell cycle arrest appeared to be dependent on ataxia and Rad-related kinase expression. DNA damage checkpoint proteins including p53, chk1, and chk2 were differentially activated by hedamycin depending on the concentration and duration of treatment. The level of downstream cell cycle regulators such as cdc25A, E2F1, cyclin E, and p21 were also altered under conditions that induced cell cycle arrest, but atypically, p21 overexpression was observed only in S-phase-arrested cells. Apoptotic indicators were only observed at moderate hedamycin concentrations associated with S-phase arrest, while increasing concentrations, when cells were arrested in G(1), resulted in a reduction of these signals. Taken together, the responses of cells to hedamycin are distinct with regard to its effect on cell cycle but also in the unusual concentration-dependent manner of activation of DNA damage and cell cycle checkpoint proteins as well as the induction of apoptotic-associated events.


Assuntos
Alquilantes/farmacologia , Aminoglicosídeos/farmacologia , Antraquinonas/farmacologia , Antibióticos Antineoplásicos/farmacologia , Citotoxinas/farmacologia , Dano ao DNA/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Ciclina E/metabolismo , DNA/biossíntese , DNA Nucleotidilexotransferase/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Células HCT116 , Humanos , Concentração Inibidora 50 , Poli(ADP-Ribose) Polimerases/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , RNA/biossíntese , Fase S/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fosfatases cdc25/metabolismo
16.
Mol Cancer Ther ; 2(7): 651-9, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12883038

RESUMO

As members of the cyclopropylpyrroloindole family, adozelesin and bizelesin cause genomic DNA lesions by alkylating DNA. Adozelesin induces single-strand DNA lesions, whereas bizelesin induces both single-strand lesions and double-strand DNA cross-links. At equivalent cytotoxic concentrations, these agents caused different biological responses. Low adozelesin concentrations (e.g., 0.5 nM) induced a transient S-phase block and cell cycle arrest in G(2)-M, as well as increased induction of p53 and p21, whereas a high drug concentration (e.g., 2.5 nM) caused apoptosis but no p21 induction. In contrast, both low and high bizelesin concentrations enhanced p53 and p21 induction and triggered G(2)-M cell cycle arrest and eventual senescence without significant apoptotic cell death. However, in cells lacking p21, bizelesin, as well as adozelesin, triggered apoptosis, indicating that p21 was crucial to sustained bizelesin-induced G(2)-M arrest. Thus, despite similar abilities to alkylate DNA, the chemotherapeutic agents adozelesin and bizelesin caused a decrease in HCT116 tumor cell proliferation by different pathways (i.e., adozelesin induced apoptosis, and bizelesin induced senescence).


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias do Colo/patologia , Ácidos Cicloexanocarboxílicos/farmacologia , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Indóis/farmacologia , Ureia/análogos & derivados , Ureia/farmacologia , Apoptose/efeitos dos fármacos , Benzofuranos , Ciclo Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Cicloexenos , Relação Dose-Resposta a Droga , Duocarmicinas , Humanos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Proteína Supressora de Tumor p53/metabolismo
17.
Nucleic Acids Res ; 31(4): 1208-15, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12582240

RESUMO

A prototype of a novel class of DNA alkylating agents, which combines the DNA crosslinking moiety chlorambucil (Chl) with a sequence-selective hairpin pyrrole-imidazole polyamide ImPy-beta-ImPy-gamma-ImPy-beta-Dp (polyamide 1), was evaluated for its ability to damage DNA and induce biological responses. Polyamide 1-Chl conjugate (1-Chl) alkylates and interstrand crosslinks DNA in cell-free systems. The alkylation occurs predominantly at 5'-AGCTGCA-3' sequence, which represents the polyamide binding site. Conjugate-induced lesions were first detected on DNA treated for 1 h with 0.1 micro M 1-Chl, indicating that the conjugate is at least 100-fold more potent than Chl. Prolonged incubation allowed for DNA damage detection even at 0.01 micro M concentration. Treatment with 1-Chl decreased DNA template activity in simian virus 40 (SV40) in vitro replication assays. 1-Chl inhibited mammalian cell growth, genomic DNA replication and cell cycle progression, and arrested cells in the G2/M phase. Moreover, cellular effects were observed at 1-Chl concentrations similar to those needed for DNA damage in cell-free systems. Neither of the parent compounds, unconjugated Chl or polyamide 1, demonstrated any cellular activity in the same concentration range. The conjugate molecule 1-Chl possesses the sequence-selectivity of a polyamide and the enhanced DNA reactivity of Chl.


Assuntos
Clorambucila/química , DNA/química , Nylons/química , Alquilação , Animais , Sequência de Bases , Sítios de Ligação , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Clorambucila/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , DNA/biossíntese , DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA Viral/química , DNA Viral/genética , Células HeLa , Humanos , Células Jurkat , Estrutura Molecular , Nylons/farmacologia , Biossíntese de Proteínas , Proteínas/efeitos dos fármacos , RNA/biossíntese , RNA/efeitos dos fármacos , Vírus 40 dos Símios/genética , Células Tumorais Cultivadas
18.
Mol Cancer Ther ; 2(1): 41-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12533671

RESUMO

Adozelesin is an alkylating minor groove DNA binder that is capable of rapidly inhibiting DNA replication in treated cells through a trans-acting mechanism and preferentially arrests cells in S phase. It has been shown previously that in cells treated with adozelesin, replication protein A (RPA) activity is deficient, and the middle subunit of RPA is hyperphosphorylated. The adozelesin-induced RPA hyperphosphorylation can be blocked by the replicative DNA polymerase inhibitor, aphidicolin, suggesting that adozelesin-triggered cellular DNA damage responses require active DNA replication forks. These data imply that cellular DNA damage responses to adozelesin treatment are preferentially induced in S phase. Here, we show that RPA hyperphosphorylation, RPA intranuclear focalization, and gamma-H2AX intranuclear focalization induced by adozelesin treatment are all dependent on DNA replication fork progression, and focalization is only induced in S phase cells. These findings are similar to those seen with the S phase-specific DNA-damaging agent, camptothecin. Conversely, all three DNA damage responses are independent of either S phase or replication fork progression when induced by treatment with the DNA strand scission agent, C-1027. Furthermore, we demonstrate that adozelesin-induced RPA and gamma-H2AX intranuclear foci appear to colocalize within the nuclei of S phase cells.


Assuntos
Aminoglicosídeos , Antineoplásicos Alquilantes/toxicidade , Camptotecina/toxicidade , Ácidos Cicloexanocarboxílicos/toxicidade , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , Indóis , Antibacterianos/toxicidade , Antibióticos Antineoplásicos/toxicidade , Afidicolina/farmacologia , Benzofuranos , Cicloexenos , Duocarmicinas , Enedi-Inos , Células HeLa , Humanos , Fase S/efeitos dos fármacos
19.
J Biol Chem ; 277(45): 42431-7, 2002 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-12196541

RESUMO

Alkylating agents are generally highly reactive with DNA but demonstrate limited DNA sequence selectivity. In contrast, synthetic pyrrole-imidazole polyamides recognize specific DNA sequences with high affinity but are unable to permanently damage DNA. An eight-ring hairpin polyamide conjugated to the alkylating moiety cyclopropylpyrroloindole, related to the natural product CC-1065, affords a conjugate 1-CBI (polyamide 1-CBI (1-(chloromethyl)-5-hydroxyl-1,2-dihydro-3H-benz[e]indole) conjugate), which binds to specific sequences in the minor groove of DNA and alkylates a single adenine flanking the polyamide binding site. In this study, we show that 1-CBI alkylates DNA in both plasmid and intracellular minichromosomal form and inhibits DNA replication under both cell-free and cellular conditions. In addition, it inhibits cell growth and arrests cells in the G2/M phase of the cell cycle.


Assuntos
Alquilantes/química , Dano ao DNA , DNA/química , Indóis/síntese química , Conformação de Ácido Nucleico , Nylons , Pirróis/síntese química , Alquilantes/farmacologia , Sequência de Bases , Neoplasias da Mama , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , DNA Viral/química , DNA Viral/efeitos dos fármacos , Feminino , Humanos , Indóis/química , Indóis/farmacologia , Oligodesoxirribonucleotídeos/química , Pirróis/química , Pirróis/farmacologia , Vírus 40 dos Símios/genética , Células Tumorais Cultivadas
20.
Bioorg Med Chem ; 10(10): 3313-8, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12150878

RESUMO

The cellular uptake and localization properties of DNA binding N-methylpyrrole/N-methylimidazole polyamide-dye conjugates in a variety of living cells have been examined by confocal laser scanning microscopy. With the exception of certain T-cell lines, polyamide-dye conjugates localize mainly in the cytoplasm and not in the nucleus. Reagents such as methanol typically used to fix cells for microscopy significantly alter the cellular localization of these DNA-binding ligands.


Assuntos
Corantes Fluorescentes/farmacocinética , Imidazóis/farmacocinética , Nylons/farmacocinética , Pirróis/farmacocinética , Compostos de Boro/farmacocinética , Compartimento Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Microscopia Confocal , Linfócitos T/citologia , Linfócitos T/metabolismo , Linfócitos T/ultraestrutura , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...