Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Biol ; 17(4): e3000204, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30951520

RESUMO

Telomerase, a unique reverse transcriptase that specifically extends the ends of linear chromosomes, is up-regulated in the vast majority of cancer cells. Here, we show that an indole nucleotide analog, 5-methylcarboxyl-indolyl-2'-deoxyriboside 5'-triphosphate (5-MeCITP), functions as an inhibitor of telomerase activity. The crystal structure of 5-MeCITP bound to the Tribolium castaneum telomerase reverse transcriptase reveals an atypical interaction, in which the nucleobase is flipped in the active site. In this orientation, the methoxy group of 5-MeCITP extends out of the canonical active site to interact with a telomerase-specific hydrophobic pocket formed by motifs 1 and 2 in the fingers domain and T-motif in the RNA-binding domain of the telomerase reverse transcriptase. In vitro data show that 5-MeCITP inhibits telomerase with a similar potency as the clinically administered nucleoside analog reverse transcriptase inhibitor azidothymidine (AZT). In addition, cell-based studies show that treatment with the cell-permeable nucleoside counterpart of 5-MeCITP leads to telomere shortening in telomerase-positive cancer cells, while resulting in significantly lower cytotoxic effects in telomerase-negative cell lines when compared with AZT treatment.


Assuntos
Nucleosídeos/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/fisiologia , Animais , Domínio Catalítico/efeitos dos fármacos , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Nucleosídeos/síntese química , Nucleosídeos/fisiologia , Nucleotídeos/síntese química , Nucleotídeos/metabolismo , RNA/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Telômero , Tribolium/genética , Tribolium/metabolismo , Zidovudina/metabolismo , Zidovudina/farmacologia
2.
Cell Rep ; 23(10): 3031-3041, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29874588

RESUMO

Telomerase, the end-replication enzyme, is reactivated in malignant cancers to drive cellular immortality. While this distinction makes telomerase an attractive target for anti-cancer therapies, most approaches for inhibiting its activity have been clinically ineffective. As opposed to inhibiting telomerase, we use its activity to selectively promote cytotoxicity in cancer cells. We show that several nucleotide analogs, including 5-fluoro-2'-deoxyuridine (5-FdU) triphosphate, are effectively incorporated by telomerase into a telomere DNA product. Administration of 5-FdU results in an increased number of telomere-induced foci, impedes binding of telomere proteins, activates the ATR-related DNA-damage response, and promotes cell death in a telomerase-dependent manner. Collectively, our data indicate that telomerase activity can be exploited as a putative anti-cancer strategy.


Assuntos
Neoplasias/enzimologia , Neoplasias/patologia , Nucleosídeos/administração & dosagem , Telomerase/metabolismo , Aminopeptidases/metabolismo , Morte Celular , Linhagem Celular Tumoral , DNA/metabolismo , Dano ao DNA , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Ligação Proteica , Pirimidinas/metabolismo , RNA Interferente Pequeno/metabolismo , Serina Proteases/metabolismo , Complexo Shelterina , Telômero/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Timidina/metabolismo , Tripeptidil-Peptidase 1
3.
Front Mol Biosci ; 4: 78, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29201867

RESUMO

Inhibiting DNA synthesis is an important therapeutic strategy that is widely used to treat a number of hyperproliferative diseases including viral infections, autoimmune disorders, and cancer. This chapter describes two major categories of therapeutic agents used to inhibit DNA synthesis. The first category includes purine and pyrmidine nucleoside analogs that directly inhibit DNA polymerase activity. The second category includes DNA damaging agents including cisplatin and chlorambucil that modify the composition and structure of the nucleic acid substrate to indirectly inhibit DNA synthesis. Special emphasis is placed on describing the molecular mechanisms of these inhibitory effects against chromosomal and mitochondrial DNA polymerases. Discussions are also provided on the mechanisms associated with resistance to these therapeutic agents. A primary focus is toward understanding the roles of specialized DNA polymerases that by-pass DNA lesions produced by DNA damaging agents. Finally, a section is provided that describes emerging areas in developing new therapeutic strategies targeting specialized DNA polymerases.

4.
J Mol Biol ; 429(15): 2308-2323, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28601494

RESUMO

Translesion DNA synthesis (TLS) is the ability of DNA polymerases to incorporate nucleotides opposite and beyond damaged DNA. TLS activity is an important risk factor for the initiation and progression of genetic diseases such as cancer. In this study, we evaluate the ability of a high-fidelity DNA polymerase to perform TLS with 8-oxo-guanine (8-oxo-G), a highly pro-mutagenic DNA lesion formed by reactive oxygen species. Results of kinetic studies monitoring the incorporation of modified nucleotide analogs demonstrate that the binding affinity of the incoming dNTP is controlled by the overall hydrophobicity of the nucleobase. However, the rate constant for the polymerization step is regulated by hydrogen-bonding interactions made between the incoming nucleotide with 8-oxo-G. Results generated here for replicating the miscoding 8-oxo-G are compared to those published for the replication of the non-instructional abasic site. During the replication of both lesions, binding of the nucleotide substrate is controlled by energetics associated with nucleobase desolvation, whereas the rate constant for the polymerization step is influenced by the physical nature of the DNA lesion, that is, miscoding versus non-instructional. Collectively, these studies highlight the importance of nucleobase desolvation as a key physical feature that enhances the misreplication of structurally diverse DNA lesions.


Assuntos
Dano ao DNA , DNA Polimerase Dirigida por DNA/metabolismo , DNA/biossíntese , 8-Hidroxi-2'-Desoxiguanosina , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Cinética , Nucleotídeos/química , Nucleotídeos/metabolismo
5.
Nucleic Acids Res ; 44(3): 1022-35, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26717984

RESUMO

This report evaluates the pro-mutagenic behavior of 8-oxo-guanine (8-oxo-G) by quantifying the ability of high-fidelity and specialized DNA polymerases to incorporate natural and modified nucleotides opposite this lesion. Although high-fidelity DNA polymerases such as pol δ and the bacteriophage T4 DNA polymerase replicating 8-oxo-G in an error-prone manner, they display remarkably low efficiencies for TLS compared to normal DNA synthesis. In contrast, pol η shows a combination of high efficiency and low fidelity when replicating 8-oxo-G. These combined properties are consistent with a pro-mutagenic role for pol η when replicating this DNA lesion. Studies using modified nucleotide analogs show that pol η relies heavily on hydrogen-bonding interactions during translesion DNA synthesis. However, nucleobase modifications such as alkylation to the N2 position of guanine significantly increase error-prone synthesis catalyzed by pol η when replicating 8-oxo-G. Molecular modeling studies demonstrate the existence of a hydrophobic pocket in pol η that participates in the increased utilization of certain hydrophobic nucleotides. A model is proposed for enhanced pro-mutagenic replication catalyzed by pol η that couples efficient incorporation of damaged nucleotides opposite oxidized DNA lesions created by reactive oxygen species. The biological implications of this model toward increasing mutagenic events in lung cancer are discussed.


Assuntos
Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Mutagênicos/toxicidade , Nucleotídeos/metabolismo , Biocatálise , Cinética , Modelos Moleculares , Conformação de Ácido Nucleico , Nucleotídeos/química
6.
Biochim Biophys Acta ; 1864(1): 165-76, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26004088

RESUMO

Nucleosides and their corresponding mono-, di-, and triphosphates play important roles in maintaining cellular homeostasis. In addition, perturbations in this homeostasis can result in dysfunctional cellular processes that cause pathological conditions such as cancer and autoimmune diseases. This review article discusses contemporary research areas applying nucleoside analogs to probe the mechanistic details underlying the complexities of nucleoside metabolism at the molecular and cellular levels. The first area describes classic and contemporary approaches used to quantify the activity of nucleoside transporters, an important class of membrane proteins that mediate the influx and efflux of nucleosides and nucleobases. A focal point of this section is describing how biophotonic nucleosides are replacing conventional assays employing radiolabeled substrates to study the mechanism of these proteins. The second section describes approaches to understand the utilization of nucleoside triphosphates by cellular DNA polymerases during DNA synthesis. Emphasis here is placed on describing how novel nucleoside analogs such as 5-ethynyl-2'-deoxyuridine are being used to quantify DNA synthesis during normal replication as well as during the replication of damaged DNA. In both sections, seminal research articles relevant to these areas are described to highlight how these novel probes are improving our understanding of these biological processes. This article is part of a Special Issue entitled: Physiological Enzymology and Protein Functions.


Assuntos
Replicação do DNA , Ácidos Nucleicos/química , Nucleosídeos/química , Nucleotídeos/química , Sequência de Bases , Química Click , Humanos , Modelos Químicos , Estrutura Molecular , Ácidos Nucleicos/genética , Ácidos Nucleicos/metabolismo , Proteínas de Transporte de Nucleosídeos/metabolismo , Nucleosídeos/metabolismo , Nucleotídeos/metabolismo
7.
J Biol Chem ; 290(15): 9714-26, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25713072

RESUMO

Nucleoside transport is an essential process that helps maintain the hyperproliferative state of most cancer cells. As such, it represents an important target for developing diagnostic and therapeutic agents that can effectively detect and treat cancer, respectively. This report describes the development of a metal-containing nucleoside designated Ir(III)-PPY nucleoside that displays both therapeutic and diagnostic properties against the human epidermal carcinoma cell line KB3-1. The cytotoxic effects of Ir(III)-PPY nucleoside are both time- and dose-dependent. Flow cytometry analyses validate that the nucleoside analog causes apoptosis by blocking cell cycle progression at G2/M. Fluorescent microscopy studies show rapid accumulation in the cytoplasm within 4 h. However, more significant accumulation is observed in the nucleus and mitochondria after 24 h. This localization is consistent with the ability of the metal-containing nucleoside to influence cell cycle progression at G2/M. Mitochondrial depletion is also observed after longer incubations (Δt ∼48 h), and this effect may produce additional cytotoxic effects. siRNA knockdown experiments demonstrate that the nucleoside transporter, hENT1, plays a key role in the cellular entry of Ir(III)-PPY nucleoside. Collectively, these data provide evidence for the development of a metal-containing nucleoside that functions as a combined therapeutic and diagnostic agent against cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Metais/metabolismo , Nucleosídeos/metabolismo , Nucleosídeos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citosol/metabolismo , Relação Dose-Resposta a Droga , Transportador Equilibrativo 1 de Nucleosídeo/genética , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Irídio/metabolismo , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Necrose , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Fatores de Tempo
8.
Expert Opin Drug Discov ; 9(2): 167-81, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24372166

RESUMO

INTRODUCTION: Ionizing radiation (IR) is an important therapeutic modality used in approximately 50% of all cancer patients and is particularly effective against solid tumors that cannot be removed by surgery or that are refractory to standard anticancer agents. IR is often combined with other chemotherapeutic agents with the goal of sensitizing cancer cells to the cytotoxic effects of IR to produce a synergistic cell-killing effect. AREAS COVERED: This review article describes current and emerging therapeutic agents that are designed to increase the therapeutic efficacy of IR. This includes a discussion of how IR causes cell death by damaging nucleic acid. The involvement of various DNA repair pathways, cell-cycle-dependent kinases and apoptotic pathways is also described. This mechanistic information provides the framework to understand how combining therapeutic modalities with IR produces synergistic effects as well as to explain how emerging therapeutic strategies are being designed to inhibit or activate these pathways. Biochemical mechanisms and clinical applications of these chemical entities are discussed. Finally, brief descriptions are provided for several emerging chemical entities that show promise as potential adjunctive agents to sensitize cells to the effects of IR. EXPERT OPINION: Using DNA damaging agents or kinase inhibitors to potentiate the cytotoxic effects of IR has significantly improved patient outcomes. However, several advancements in instrumentation as well as new molecular targets are changing the landscape of applying IR as a therapeutic modality.


Assuntos
Neoplasias/radioterapia , Radiação Ionizante , Animais , Antineoplásicos/uso terapêutico , Morte Celular , Terapia Combinada , Dano ao DNA , Humanos , Neoplasias/tratamento farmacológico , Resultado do Tratamento
9.
Chemistry ; 19(47): 15924-32, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24222393

RESUMO

Fundamental study of enzymatic nucleoside transport suffers for lack of optical probes that can be tracked noninvasively. Nucleoside transporters are integral membrane glycoproteins that mediate the salvage of nucleosides and their passage across cell membranes. The substrate recognition site is the deoxyribose sugar, often with little distinction among nucleobases. Reported here are nucleoside analogues in which emissive, cyclometalated iridium(III) complexes are "clicked" to C-1 of deoxyribose in place of canonical nucleobases. The resulting complexes show visible luminescence at room temperature and 77 K with microsecond-length triplet lifetimes. A representative complex is crystallographically characterized. Transport and luminescence are demonstrated in cultured human carcinoma (KB3-1) cells.


Assuntos
Complexos de Coordenação/química , Desoxirribose/química , Irídio/química , Linhagem Celular Tumoral , Química Click , Complexos de Coordenação/síntese química , Complexos de Coordenação/metabolismo , Cristalografia por Raios X , Ciclização , Humanos , Medições Luminescentes , Microscopia Confocal , Conformação Molecular , Teoria Quântica , Temperatura
10.
ACS Chem Biol ; 8(11): 2452-65, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23992753

RESUMO

Nucleoside analogs are an important class of anticancer agent that historically show better efficacy against hematological cancers versus solid tumors. This report describes the development and characterization of a new class of nucleoside analog that displays anticancer effects against both hematological and adherent cancer cell lines. These new analogs lack canonical hydrogen-bonding groups yet are effective nucleotide substrates for several high-fidelity DNA polymerases. Permutations in the position of the non-hydrogen-bonding functional group greatly influence the kinetic behavior of these nucleosides. One particular analog designated 4-nitroindolyl-2'-deoxynucleoside triphosphate (4-NITP) is unique as it is incorporated opposite C and T with high catalytic efficiencies. In addition, this analog functions as a nonobligate chain terminator of DNA synthesis, since it is poorly elongated. Consistent with this mechanism, the corresponding nucleoside, 4-nitroindolyl-2'-deoxynucleoside (4-NIdR), produces antiproliferative effects against leukemia cells. 4-NIdR also produces cytostatic and cytotoxic effects against several adherent cancer cell lines, especially those that are deficient in mismatch repair and p53. Cell death in this case appears to occur via mitotic catastrophe, a specialized form of apoptosis. Mass spectroscopy experiments performed on nucleic acid isolated from cells treated with 4-NIdR validate that the non-natural nucleoside is stably incorporated into DNA. Xenograft mouse studies demonstrate that administration of 4-NIdR delays tumor growth without producing adverse side effects such as anemia and thrombocytopenia. Collectively, the results of in vitro, cell-based, and animal studies provide evidence for the development of a novel nucleoside analog that shows enhanced effectiveness against solid tumors.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Desoxirribonucleotídeos/síntese química , Desoxirribonucleotídeos/farmacologia , Nucleosídeos/síntese química , Nucleosídeos/farmacologia , Animais , Antineoplásicos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desoxirribonucleotídeos/química , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias/tratamento farmacológico , Nucleosídeos/química
11.
Future Med Chem ; 4(11): 1461-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22857534

RESUMO

Nucleoside transporters play important physiological roles by regulating intra- and extra-cellular concentrations of purine and pyrimidine (deoxy)nucleosides. This review describes the biological function and activity of the two major families of membrane nucleoside transporters that exist in mammalian cells. These include equilibrative nucleoside transporters that transport nucleosides in a gradient-dependent fashion and concentrative nucleoside transporters that import nucleosides against a gradient by coupling movement with sodium transport. Particular emphasis is placed on describing the roles of nucleoside transport in normal physiological processes, including inflammation, cardiovascular function and nutrient transport across the blood-brain barrier. In addition, the role of nucleoside transport in pathological conditions such as cardiovascular disease and cancer are discussed. The potential therapeutic applications of manipulating nucleoside transport activities are discussed, focusing on nucleoside analogs as anti-neoplastic agents. Finally, we discuss future directions for the development of novel chemical entities to measure nucleoside transport activity at the cellular and organismal level.


Assuntos
Proteínas de Transporte de Nucleobases, Nucleosídeos, Nucleotídeos e Ácidos Nucleicos/metabolismo , Nucleosídeos/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Doenças Cardiovasculares/tratamento farmacológico , Humanos , Neoplasias/tratamento farmacológico , Proteínas de Transporte de Nucleobases, Nucleosídeos, Nucleotídeos e Ácidos Nucleicos/antagonistas & inibidores , Nucleosídeos/farmacologia , Nucleosídeos/uso terapêutico , Relação Estrutura-Atividade
12.
ACS Chem Biol ; 7(6): 988-98, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22390204

RESUMO

Acute lymphoblastic leukemia (ALL) is the most common type of childhood cancer, presenting with approximately 5,000 new cases each year in the United States. An interesting enzyme implicated in this disease is terminal deoxynucleotidyl transferase (TdT), a specialized DNA polymerase involved in V(D)J recombination. TdT is an excellent biomarker for ALL as it is overexpressed in ~90% of ALL patients, and these higher levels correlate with a poor prognosis. These collective features make TdT an attractive target to design new selective anti-cancer agents against ALL. In this report, we evaluate the anti-leukemia activities of two non-natural nucleotides designated 5-nitroindolyl-2'-deoxynucleoside triphosphate (5-NITP) and 3-ethynyl-5-nitroindolyl-2'-deoxynucleoside triphosphate (3-Eth-5-NITP). Using purified TdT, we demonstrate that both non-natural nucleotides are efficiently utilized as TdT substrates. However, 3-Eth-5-NITP is poorly elongated, and this observation validates its activity as a chain-terminator for blunt-end DNA synthesis. Cell-based experiments validate that the corresponding non-natural nucleoside produces robust cytostatic and cytotoxic effects against leukemia cells that overexpress TdT. The strategic placement of the ethynyl moiety allows the incorporated nucleoside triphosphate to be selectively tagged with an azide-containing fluorophore via "click" chemistry. This reaction allows the extent of nucleotide incorporation to be quantified such that the anti-cancer effects of the corresponding non-natural nucleoside can be self-assessed. The applications of this novel nucleoside are discussed, focusing on its use as a "theranostic" agent that can improve the accuracy of dosing regimens and accelerate clinical decisions regarding therapeutic intervention.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , DNA Nucleotidilexotransferase/metabolismo , Nucleosídeos/química , Nucleosídeos/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico
13.
J Med Chem ; 55(5): 2437-51, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-22289037

RESUMO

This report describes the design and application of several distinct gold-containing indoles as anticancer agents. When used individually, all gold-bearing compounds display cytostatic effects against leukemia and adherent cancer cell lines. However, two gold-bearing indoles show unique behavior by increasing the cytotoxic effects of clinically relevant levels of ionizing radiation. Quantifying the amount of DNA damage demonstrates that each gold-indole enhances apoptosis by inhibiting DNA repair. Both Au(I)-indoles were tested for inhibitory effects against various cellular targets including thioredoxin reductase, a known target of several gold compounds, and various ATP-dependent kinases. While neither compound significantly inhibits the activity of thioreoxin reductase, both showed inhibitory effects against several kinases associated with cancer initiation and progression. The inhibition of these kinases provides a possible mechanism for the ability of these Au(I)-indoles to potentiate the cytotoxic effects of ionizing radiation. Clinical applications of combining Au(I)-indoles with ionizing radiation are discussed as a new strategy to achieve chemosensitization of cancer cells.


Assuntos
Antineoplásicos/síntese química , Ouro , Indóis/síntese química , Compostos Organometálicos/síntese química , Fosfinas/síntese química , Radiossensibilizantes/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Raios gama , Humanos , Indóis/química , Indóis/farmacologia , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Fosfinas/química , Fosfinas/farmacologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia , Relação Estrutura-Atividade , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores
14.
PLoS One ; 6(4): e18824, 2011 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-21533111

RESUMO

Pseudomonas aeruginosa is especially adept at colonizing the airways of individuals afflicted with the autosomal recessive disease cystic fibrosis (CF). CF patients suffer from chronic airway inflammation, which contributes to lung deterioration. Once established in the airways, P. aeruginosa continuously adapts to the changing environment, in part through acquisition of beneficial mutations via a process termed pathoadaptation. MutS and DinB are proposed to play opposing roles in P. aeruginosa pathoadaptation: MutS acts in replication-coupled mismatch repair, which acts to limit spontaneous mutations; in contrast, DinB (DNA polymerase IV) catalyzes error-prone bypass of DNA lesions, contributing to mutations. As part of an ongoing effort to understand mechanisms underlying P. aeruginosa pathoadaptation, we characterized hydrogen peroxide (H(2)O(2))-induced phenotypes of isogenic P. aeruginosa strains bearing different combinations of mutS and dinB alleles. Our results demonstrate an unexpected epistatic relationship between mutS and dinB with respect to H(2)O(2)-induced cell killing involving error-prone repair and/or tolerance of oxidized DNA lesions. In striking contrast to these error-prone roles, both MutS and DinB played largely accurate roles in coping with DNA lesions induced by ultraviolet light, mitomycin C, or 4-nitroquinilone 1-oxide. Models discussing roles for MutS and DinB functionality in DNA damage-induced mutagenesis, particularly during CF airway colonization and subsequent P. aeruginosa pathoadaptation are discussed.


Assuntos
Dano ao DNA , Epistasia Genética , Proteína MutS de Ligação de DNA com Erro de Pareamento/fisiologia , Pseudomonas aeruginosa/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Catálise , Peróxido de Hidrogênio/metabolismo , Mutagênese , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
15.
ACS Chem Biol ; 6(8): 781-8, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21520912

RESUMO

Lon and ClpXP are the only soluble ATP-dependent proteases within the mammalian mitochondria matrix, which function in protein quality control by selectively degrading misfolded, misassembled, or damaged proteins. Chemical tools to study these proteases in biological samples have not been identified, thereby hindering a clear understanding of their respective functions in normal and disease states. In this study, we applied a proteolytic site-directed approach to identify a peptide reporter substrate and a peptide inhibitor that are selective for Lon but not ClpXP. These chemical tools permit quantitative measurements that distinguish Lon-mediated proteolysis from that of ClpXP in biochemical assays with purified proteases, as well as in intact mitochondria and mitochondrial lysates. This chemical biology approach provides needed tools to further our understanding of mitochondrial ATP-dependent proteolysis and contributes to the future development of diagnostic and pharmacological agents for treating diseases associated with defects in mitochondrial protein quality.


Assuntos
Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/metabolismo , Peptídeos/metabolismo , Protease La/antagonistas & inibidores , Protease La/metabolismo , Trifosfato de Adenosina/metabolismo , Domínio Catalítico , Endopeptidase Clp/antagonistas & inibidores , Endopeptidase Clp/metabolismo , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/metabolismo , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Células HeLa , Humanos , Mitocôndrias/metabolismo , Peptídeos/análise , Proteólise
16.
Biochemistry ; 49(8): 1640-8, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20104904

RESUMO

Multidrug resistance during cancer chemotherapy is commonly acquired by overexpression of the ATP binding cassette transporter, P-glycoprotein (P-gp). As such, inhibitors that target P-gp activity represent potential therapeutic agents against this form of drug resistance. This study evaluated the ability of various non-natural nucleosides that mimic the core structure of adenosine to modulate drug resistance by inhibiting the ATPase activity to P-gp. Of several analogues tested, only one novel non-natural nucleoside, 5-cyclohexylindolyl-2'-deoxyribose (5-CHInd), behaves as a P-gp inhibitor. Although 5-CHInd is an adenosine analogue that should block the binding of ATP, the non-natural nucleoside surprisingly stimulates the ATPase activity of P-gp in vitro. However, 5-CHInd is not an exportable substrate for P-gp as it is not transported across an MDCK-MDR1 monolayer. In addition, 5-CHInd differentially modulates MDR by decreasing or increasing the cytotoxicity of several chemotherapeutic agents. Although 5-CHInd displays variable activity in modulating the efflux of various drugs by P-gp, there is a correlation between changes observed in the drug-stimulated ATPase catalytic efficiency induced by 5-CHInd and its effect on drug efflux. The paradoxical behavior of 5-CHInd is rationalized within the context of contemporary models of P-gp function. In addition, the data are used to develop a predictive in vitro model for rapidly identifying potential drug-drug interactions with P-gp.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Desoxirribose/química , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Nucleosídeos/química , Nucleosídeos/farmacologia , Adenosina Trifosfatases/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cães , Ativação Enzimática/efeitos dos fármacos , Humanos , Cinética , Modelos Biológicos , Estrutura Molecular , Nucleosídeos/efeitos adversos
17.
Biochim Biophys Acta ; 1804(5): 1151-66, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19596089

RESUMO

Nearly every DNA polymerase characterized to date exclusively catalyzes the incorporation of mononucleotides into a growing primer using a DNA or RNA template as a guide to direct each incorporation event. There is, however, one unique DNA polymerase designated terminal deoxynucleotidyl transferase that performs DNA synthesis using only single-stranded DNA as the nucleic acid substrate. In this chapter, we review the biological role of this enigmatic DNA polymerase and the biochemical mechanism for its ability to perform DNA synthesis in the absence of a templating strand. We compare and contrast the molecular events for template-independent DNA synthesis catalyzed by terminal deoxynucleotidyl transferase with other well-characterized DNA polymerases that perform template-dependent synthesis. This includes a quantitative inspection of how terminal deoxynucleotidyl transferase binds DNA and dNTP substrates, the possible involvement of a conformational change that precedes phosphoryl transfer, and kinetic steps that are associated with the release of products. These enzymatic steps are discussed within the context of the available structures of terminal deoxynucleotidyl transferase in the presence of DNA or nucleotide substrate. In addition, we discuss the ability of proteins involved in replication and recombination to regulate the activity of the terminal deoxynucleotidyl transferase. Finally, the biomedical role of this specialized DNA polymerase is discussed focusing on its involvement in cancer development and its use in biomedical applications such as labeling DNA for detecting apoptosis.


Assuntos
DNA Nucleotidilexotransferase/metabolismo , DNA Polimerase Dirigida por DNA/fisiologia , DNA/metabolismo , Animais , Humanos , Conformação de Ácido Nucleico , Conformação Proteica
19.
Biochemistry ; 47(32): 8253-60, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18642851

RESUMO

Numerous pathological states, including cancer, autoimmune diseases, and viral/bacterial infections, are often attributed to uncontrollable DNA replication. Inhibiting this essential biological process provides an obvious therapeutic target against these diseases. A logical target is the DNA polymerase, the enzyme responsible for catalyzing the addition of mononucleotides to a growing polymer using a DNA or RNA template as a guide for directing each incorporation event. This review provides a summary of therapeutic agents that target polymerase activity. A discussion of the biological function and mechanism of polymerases is first provided to illustrate the strategy for therapeutic intervention as well as the rational design of various nucleoside analogues that inhibit various polymerases associated with viral infections and cancer. The development of nucleoside and non-nucleoside inhibitors as antiviral agents is discussed with particular emphasis on their mechanism of action, structure-activity relationships, toxicity, and mechanism of resistance. In addition, commonly used anticancer agents are described to illustrate the similarities and differences associated with various nucleoside analogues as therapeutic agents. Finally, new therapeutic approaches that include the inhibition of selective polymerases involved in DNA repair and/or translesion DNA synthesis as anticancer agents are discussed.


Assuntos
DNA Polimerase Dirigida por DNA/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Inibidores da Síntese de Ácido Nucleico , Animais , Antivirais/administração & dosagem , Antivirais/química , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Inibidores da Transcriptase Reversa/administração & dosagem , Inibidores da Transcriptase Reversa/química , Relação Estrutura-Atividade
20.
Biochemistry ; 46(48): 13752-61, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-17983244

RESUMO

Abasic sites are mutagenic DNA lesions formed as a consequence of inappropriate modifications to the functional groups present on purines and pyrimidines. In this paper we quantify the ability of the high-fidelity bacteriophage T4 DNA polymerase to incorporate various promutagenic alkylated nucleotides opposite and beyond this class of non-instructional DNA lesions. Kinetic analyses reveal that modified nucleotides such as N6-methyl-dATP and O6-methyl-dGTP are incorporated opposite an abasic site far more effectively than their unmodified counterparts. The enhanced incorporation is caused by a 10-fold increase in kpol values that correlates with an increase in hydrophobicity as well as changes in the tautomeric form of the nucleobase to resemble adenine. These biophysical features lead to enhanced base-stacking properties that also contribute toward their ability to be easily extended when paired opposite the non-instructional DNA lesion. Surprisingly, misincorporation opposite templating DNA is not enhanced by the increased base-stacking properties of most modified purines. The dichotomy in promutagenic DNA synthesis catalyzed by a high-fidelity polymerase indicates that the dynamics for misreplicating a miscoding versus a non-instructional DNA lesion are different. The collective data set is used to propose models accounting for synergistic enhancements in mutagenesis and the potential to develop treatment-related malignancies as a consequence of utilizing DNA-damaging agents as chemotherapeutic agents.


Assuntos
Trifosfato de Adenosina/química , Dano ao DNA , Replicação do DNA , Guanosina Trifosfato/química , Mutagênicos/toxicidade , DNA/efeitos dos fármacos , Cinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA