Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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.
FEBS Lett ; 590(12): 1704-12, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27155231

RESUMO

Sml1 is an intrinsically disordered protein inhibitor of Saccharomyces cerevisiae ribonucleotide reductase (ScRR1), but its inhibition mechanism is poorly understood. RR reduces ribonucleoside diphosphates to their deoxy forms, and balances the nucleotide pool. Multiple turnover kinetics show that Sml1 inhibition of dGTP/ADP- and ATP/CDP-bound ScRR follows a mixed inhibition mechanism. However, Sml1 cooperatively binds to the ES complex in the dGTP/ADP form, whereas with ATP/CDP, Sml1 binds weakly and noncooperatively. Gel filtration and mutagenesis studies indicate that Sml1 does not alter the oligomerization equilibrium and the CXXC motif is not involved in the inhibition. The data suggest that Sml1 is an allosteric inhibitor.


Assuntos
Ribonucleotídeo Redutases/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Regulação Alostérica/fisiologia , Motivos de Aminoácidos , Ligação Proteica/fisiologia , Multimerização Proteica/fisiologia , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
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
7.
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
8.
Biochim Biophys Acta ; 1864(1): 65-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26277093

RESUMO

Historically, the study of proteins has relied heavily on characterizing the activity of a single purified protein isolated from other cellular components. This classic approach allowed scientists to unambiguously define the intrinsic kinetic and chemical properties of that protein. The ultimate hope was to extrapolate this information toward understanding how the enzyme or receptor behaves within its native cellular context. These types of detailed in vitro analyses were necessary to reduce the innate complexities of measuring the singular activity and biochemical properties of a specific enzyme without interference from other enzymes and potential competing substrates. However, recent developments in fields encompassing cell biology, molecular imaging, and chemical biology now provide the unique chemical tools and instrumentation to study protein structure, function, and regulation in their native cellular environment. These advancements provide the foundation for a new field, coined physiological enzymology, which quantifies the function and regulation of enzymes and proteins at the cellular level. In this Special Edition, we explore the area of Physiological Enzymology and Protein Function through a series of review articles that focus on the tools and techniques used to measure the cellular activity of proteins inside living cells. This article is part of a Special Issue entitled: Physiological Enzymology and Protein Functions.


Assuntos
Técnicas de Química Analítica/métodos , Enzimas/metabolismo , Espaço Intracelular/enzimologia , Proteínas/metabolismo , Biocatálise , Biologia Celular/tendências , Técnicas de Química Analítica/tendências , Cristalografia por Raios X , Ensaios Enzimáticos/métodos , Enzimas/química , Cinética , Conformação Proteica , Proteínas/química , Análise Espectral/métodos
10.
PLoS Genet ; 11(9): e1005507, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26352807

RESUMO

Translesion DNA synthesis (TLS) by specialized DNA polymerases (Pols) is a conserved mechanism for tolerating replication blocking DNA lesions. The actions of TLS Pols are managed in part by ring-shaped sliding clamp proteins. In addition to catalyzing TLS, altered expression of TLS Pols impedes cellular growth. The goal of this study was to define the relationship between the physiological function of Escherichia coli Pol IV in TLS and its ability to impede growth when overproduced. To this end, 13 novel Pol IV mutants were identified that failed to impede growth. Subsequent analysis of these mutants suggest that overproduced levels of Pol IV inhibit E. coli growth by gaining inappropriate access to the replication fork via a Pol III-Pol IV switch that is mechanistically similar to that used under physiological conditions to coordinate Pol IV-catalyzed TLS with Pol III-catalyzed replication. Detailed analysis of one mutant, Pol IV-T120P, and two previously described Pol IV mutants impaired for interaction with either the rim (Pol IVR) or the cleft (Pol IVC) of the ß sliding clamp revealed novel insights into the mechanism of the Pol III-Pol IV switch. Specifically, Pol IV-T120P retained complete catalytic activity in vitro but, like Pol IVR and Pol IVC, failed to support Pol IV TLS function in vivo. Notably, the T120P mutation abrogated a biochemical interaction of Pol IV with Pol III that was required for Pol III-Pol IV switching. Taken together, these results support a model in which Pol III-Pol IV switching involves interaction of Pol IV with Pol III, as well as the ß clamp rim and cleft. Moreover, they provide strong support for the view that Pol III-Pol IV switching represents a vitally important mechanism for regulating TLS in vivo by managing access of Pol IV to the DNA.


Assuntos
Dano ao DNA , DNA Polimerase beta/metabolismo , Reparo do DNA , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Seleção Genética , Domínio Catalítico , DNA Polimerase beta/genética , Replicação do DNA , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Ligação Proteica
11.
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
12.
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
13.
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
14.
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
15.
Chembiochem ; 14(4): 489-98, 2013 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-23404822

RESUMO

This report describes the use of several isosteric non-natural nucleotides as probes to evaluate the roles of nucleobase shape, size, solvation energies, and π-electron interactions as forces influencing key kinetic steps of the DNA polymerization cycle. Results are provided using representative high- and low-fidelity DNA polymerases. Results generated with the E. coli Klenow fragment reveal that this high-fidelity polymerase utilizes hydrophobic nucleotide analogues with higher catalytic efficiencies compared to hydrophilic analogues. These data support a major role for nucleobase desolvation during nucleotide selection and insertion. In contrast, the low-fidelity HIV-1 reverse transcriptase discriminates against hydrophobic analogues and only tolerates non-natural nucleotides that are capable of hydrogen-bonding or π-stacking interactions. Surprisingly, hydrophobic analogues that function as efficient substrates for the E. coli Klenow fragment behave as noncompetitive or uncompetitive inhibitors against HIV-1 reverse transcriptase. In these cases, the mode of inhibition depends upon the absence or presence of a templating nucleobase. Molecular modeling studies suggest that these analogues bind to the active site of reverse transcriptase as well as to a nearby hydrophobic binding pocket. Collectively, the studies using these non-natural nucleotides reveal important mechanistic differences between representative high- and low-fidelity DNA polymerases during nucleotide selection and incorporation.


Assuntos
DNA Polimerase I/metabolismo , DNA/metabolismo , Escherichia coli/enzimologia , Transcriptase Reversa do HIV/metabolismo , HIV-1/enzimologia , Nucleotídeos/metabolismo , Sítios de Ligação , DNA/química , DNA Polimerase I/química , Elétrons , Escherichia coli/química , Transcriptase Reversa do HIV/química , HIV-1/química , Interações Hidrofóbicas e Hidrofílicas , Simulação de Acoplamento Molecular , Nucleotídeos/química , Polimerização
16.
Biochim Biophys Acta ; 1834(1): 34-45, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22959853

RESUMO

High fidelity DNA polymerases maintain genomic fidelity through a series of kinetic steps that include nucleotide binding, conformational changes, phosphoryl transfer, polymerase translocation, and nucleotide excision. Developing a comprehensive understanding of how these steps are coordinated during correct and pro-mutagenic DNA synthesis has been hindered due to lack of spectroscopic nucleotides that function as efficient polymerase substrates. This report describes the application of a non-natural nucleotide designated 5-naphthyl-indole-2'-deoxyribose triphosphate which behaves as a fluorogenic substrate to monitor nucleotide incorporation and excision during the replication of normal DNA versus two distinct DNA lesions (cyclobutane thymine dimer and an abasic site). Transient fluorescence and rapid-chemical quench experiments demonstrate that the rate constants for nucleotide incorporation vary as a function of DNA lesion. These differences indicate that the non-natural nucleotide can function as a spectroscopic probe to distinguish between normal versus translesion DNA synthesis. Studies using wild-type DNA polymerase reveal the presence of a fluorescence recovery phase that corresponds to the formation of a pre-excision complex that precedes hydrolytic excision of the non-natural nucleotide. Rate constants for the formation of this pre-excision complex are dependent upon the DNA lesion, and this suggests that the mechanism of exonuclease proofreading is regulated by the nature of the formed mispair. Finally, spectroscopic evidence confirms that exonuclease proofreading competes with polymerase translocation. Collectively, this work provides the first demonstration for a non-natural nucleotide that functions as a spectroscopic probe to study the coordinated efforts of polymerization and exonuclease proofreading during correct and translesion DNA synthesis.


Assuntos
Bacteriófago T4/enzimologia , DNA Polimerase Dirigida por DNA/química , DNA/biossíntese , Exonucleases/química , Proteínas Virais/química , DNA/química , DNA/genética , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Exonucleases/genética , Exonucleases/metabolismo , Espectrometria de Fluorescência , Proteínas Virais/genética , Proteínas Virais/metabolismo
17.
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
18.
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
19.
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
20.
Nucleic Acids Res ; 40(5): 2357-67, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22086959

RESUMO

The misreplication of damaged DNA is an important biological process that produces numerous adverse effects on human health. This report describes the synthesis and characterization of a non-natural nucleotide, designated 3-ethynyl-5-nitroindolyl-2'-deoxyriboside triphosphate (3-Eth-5-NITP), as a novel chemical reagent that can probe and quantify the misreplication of damaged DNA. We demonstrate that this non-natural nucleotide is efficiently inserted opposite an abasic site, a commonly formed and potentially mutagenic non-instructional DNA lesion. The strategic placement of the ethynyl moiety allows the incorporated nucleoside triphosphate to be selectively tagged with an azide-containing fluorophore using 'click' chemistry. This reaction provides a facile way to quantify the extent of nucleotide incorporation opposite non-instructional DNA lesions. In addition, the incorporation of 3-Eth-5-NITP is highly selective for an abasic site, and occurs even in the presence of a 50-fold molar excess of natural nucleotides. The biological applications of using 3-Eth-5-NITP as a chemical probe to monitor and quantify the misreplication of non-instructional DNA lesions are discussed.


Assuntos
Dano ao DNA , Indóis/química , Nucleotídeos/química , Química Click , DNA/química , Replicação do DNA , Indóis/síntese química , Cinética , Nucleosídeos/síntese química , Nucleosídeos/química , Nucleotídeos/síntese química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...