Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
1.
Nature ; 571(7764): 275-278, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31181567

RESUMEN

Recently developed DNA base editing methods enable the direct generation of desired point mutations in genomic DNA without generating any double-strand breaks1-3, but the issue of off-target edits has limited the application of these methods. Although several previous studies have evaluated off-target mutations in genomic DNA4-8, it is now clear that the deaminases that are integral to commonly used DNA base editors often bind to RNA9-13. For example, the cytosine deaminase APOBEC1-which is used in cytosine base editors (CBEs)-targets both DNA and RNA12, and the adenine deaminase TadA-which is used in adenine base editors (ABEs)-induces site-specific inosine formation on RNA9,11. However, any potential RNA mutations caused by DNA base editors have not been evaluated. Adeno-associated viruses are the most common delivery system for gene therapies that involve DNA editing; these viruses can sustain long-term gene expression in vivo, so the extent of potential RNA mutations induced by DNA base editors is of great concern14-16. Here we quantitatively evaluated RNA single nucleotide variations (SNVs) that were induced by CBEs or ABEs. Both the cytosine base editor BE3 and the adenine base editor ABE7.10 generated tens of thousands of off-target RNA SNVs. Subsequently, by engineering deaminases, we found that three CBE variants and one ABE variant showed a reduction in off-target RNA SNVs to the baseline while maintaining efficient DNA on-target activity. This study reveals a previously overlooked aspect of off-target effects in DNA editing and also demonstrates that such effects can be eliminated by engineering deaminases.


Asunto(s)
ADN/genética , Edición Génica/métodos , Mutagénesis , Mutación , Nucleósido Desaminasas/genética , Ingeniería de Proteínas , ARN/genética , Adenina/metabolismo , Aminohidrolasas/genética , Aminohidrolasas/metabolismo , Citosina/metabolismo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Células HEK293 , Humanos , Nucleósido Desaminasas/metabolismo , Especificidad por Sustrato , Transfección
2.
BMC Cancer ; 19(1): 197, 2019 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-30832616

RESUMEN

BACKGROUND: The cytosine deaminase (CD)/5-fluorocytosine (5-FC) system is among the best explored enzyme/prodrug systems in the field of the suicide gene therapy. Recently, by the screening of the environmental metagenomic libraries we identified a novel isocytosine deaminase (ICD), termed Vcz, which is able of specifically converting a prodrug 5-fluoroisocytosine (5-FIC) into toxic drug 5-fluorouracil (5-FU). The aim of this study is to test the applicability of the ICD Vcz / 5-FIC pair as a potential suicide gene therapy tool. METHODS: Vcz-expressing human glioblastoma U87 and epithelial colorectal adenocarcinoma Caco-2 cells were treated with 5-FIC, and the Vcz-mediated cytotoxicity was evaluated by performing an MTT assay. In order to examine anti-tumor effects of the Vcz/5-FIC system in vivo, murine bone marrow-derived mesenchymal stem cells (MSC) were transduced with the Vcz-coding lentivirus and co-injected with 5-FIC or control reagents into subcutaneous GL261 tumors evoked in C57/BL6 mice. RESULTS: 5-FIC alone showed no significant toxic effects on U87 and Caco-2 cells at 100 µM concentration, whereas the number of cells of both cell lines that express Vcz cytosine deaminase gene decreased by approximately 60% in the presence of 5-FIC. The cytotoxic effects on cells were also induced by media collected from Vcz-expressing cells pre-treated with 5-FIC. The co-injection of the Vcz-transduced mesenchymal stem cells and 5-FIC have been shown to augment tumor necrosis and increase longevity of tumorized mice by 50% in comparison with control group animals. CONCLUSIONS: We have confirmed that the novel ICD Vcz together with the non-toxic prodrug 5-FIC has a potential of being a new enzyme/prodrug system for suicide gene therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Flucitosina/análogos & derivados , Fluorouracilo/farmacología , Genes Transgénicos Suicidas , Profármacos/farmacología , Adenocarcinoma , Animales , Antimetabolitos Antineoplásicos/metabolismo , Neoplasias Encefálicas , Células CACO-2 , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales , Citosina/análogos & derivados , Citosina/metabolismo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Flucitosina/metabolismo , Flucitosina/farmacología , Fluorouracilo/metabolismo , Terapia Genética , Vectores Genéticos , Glioblastoma , Humanos , Lentivirus , Células Madre Mesenquimatosas , Ratones , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Profármacos/metabolismo
3.
Plant Cell ; 31(3): 734-751, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30787180

RESUMEN

Plants can fully catabolize purine nucleotides. A firmly established central intermediate is the purine base xanthine. In the current widely accepted model of plant purine nucleotide catabolism, xanthine can be generated in various ways involving either inosine and hypoxanthine or guanosine and xanthosine as intermediates. In a comprehensive mutant analysis involving single and multiple mutants of urate oxidase, xanthine dehydrogenase, nucleoside hydrolases, guanosine deaminase, and hypoxanthine guanine phosphoribosyltransferase, we demonstrate that purine nucleotide catabolism in Arabidopsis (Arabidopsis thaliana) mainly generates xanthosine, but not inosine and hypoxanthine, and that xanthosine is derived from guanosine deamination and a second source, likely xanthosine monophosphate dephosphorylation. Nucleoside hydrolase 1 (NSH1) is known to be essential for xanthosine hydrolysis, but the in vivo function of a second cytosolic nucleoside hydrolase, NSH2, is unclear. We demonstrate that NSH1 activates NSH2 in vitro and in vivo, forming a complex with almost two orders of magnitude higher catalytic efficiency for xanthosine hydrolysis than observed for NSH1 alone. Remarkably, an inactive NSH1 point mutant can activate NSH2 in vivo, fully preventing purine nucleoside accumulation in nsh1 background. Our data lead to an altered model of purine nucleotide catabolism that includes an NSH heterocomplex as a central component.


Asunto(s)
Adenosina Monofosfato/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Guanosina Monofosfato/metabolismo , Ribonucleósidos/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Mutación , N-Glicosil Hidrolasas/genética , N-Glicosil Hidrolasas/metabolismo , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Plantas Modificadas Genéticamente , Xantinas
4.
Alcohol Clin Exp Res ; 41(7): 1271-1279, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28449374

RESUMEN

BACKGROUND: A substantial part of the risk for alcohol use disorder is determined by genetic factors. We previously used chromosome substitution (CSS) mice, to identify a quantitative trait loci (QTL) for alcohol preference on mouse chromosome 2. The aim of this study was to identify candidate genes within this QTL that confer the risk for alcohol preference. METHODS: In order to delineate the neurobiological underpinnings of alcohol consumption, we expanded on the QTL approach to identify candidate genes for high alcohol preference in mice. We narrowed down a QTL for alcohol preference on mouse chromosome 2, that we previously identified using CSS mice, to 4 candidate genes in silico. Expression levels of these candidate genes in prefrontal cortex, amygdala, and nucleus accumbens-brain regions implicated in reward and addiction-were subsequently compared for the CSS-2 and the C57BL/6J host strain. RESULTS: We observed increased expression of adenosine deaminase-like (Adal) in all 3 regions in CSS-2 mice. Moreover, we found that the adenosine deaminase inhibitor EHNA reduced the difference in alcohol preference between CSS-2 and C57BL/6J mice. CONCLUSIONS: This study identifies Adal as a genetically protective factor against alcohol consumption in mice, in which elevated Adal levels contribute to low alcohol preference.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Nucleósido Desaminasas/genética , Animales , Encéfalo/metabolismo , Masculino , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Receptores Colinérgicos/metabolismo
5.
Sci Rep ; 6: 38133, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27924834

RESUMEN

Follicular lymphoma (FL) is an uncurable cancer characterized by progressive severity of relapses. We analyzed sequence context specificity of mutations in the B cells from a large cohort of FL patients. We revealed substantial excess of mutations within a novel hybrid nucleotide motif: the signature of somatic hypermutation (SHM) enzyme, Activation Induced Deaminase (AID), which overlaps the CpG methylation site. This finding implies that in FL the SHM machinery acts at genomic sites containing methylated cytosine. We identified the prevalence of this hybrid mutational signature in many other types of human cancer, suggesting that AID-mediated, CpG-methylation dependent mutagenesis is a common feature of tumorigenesis.


Asunto(s)
Islas de CpG/genética , Metilación de ADN/genética , Linfoma Folicular/genética , Mutación/genética , Nucleósido Desaminasas/genética , Linfocitos B/metabolismo , Carcinogénesis/genética , Citosina/metabolismo , Humanos , Mutagénesis/genética , Nucleótidos/genética
6.
J Bacteriol ; 196(11): 1950-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24633874

RESUMEN

We show here that deoxycytidine deaminase (DCD)-deficient mutants of Escherichia coli are hypersensitive to killing by exogenous cytidine, adenosine, or guanosine, whereas wild-type cells are not. This hypersensitivity is reversed by exogenous thymidine. The mechanism likely involves the allosteric regulation of ribonucleotide reductase and severe limitations of the dTTP pools, resulting in thymineless death, the phenomenon of cell death due to thymidine starvation. We also report here that DCD-deficient mutants of E. coli are more sensitive to a series of different antibiotics, including vancomycin, and we show synergistic killing with the combination of vancomycin and cytidine. One possibility is that a very low, subinhibitory concentration of vancomycin enters Gram-negative cells and that this concentration is potentiated by chromosomal lesions resulting from the thymineless state. A second possibility is that the metabolic imbalance resulting from DCD deficiency affects the assembly of the outer membrane, which normally presents a barrier to drugs such as vancomycin. We consider these findings with regard to ideas of rendering Gram-negative bacteria sensitive to drugs such as vancomycin.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/enzimología , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Nucleósido Desaminasas/metabolismo , Vancomicina/farmacología , Adenosina/farmacología , Citidina/farmacología , Citidina Desaminasa , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Eliminación de Gen , Guanosina/farmacología , Nucleósido Desaminasas/genética
7.
Plant Cell ; 25(10): 4101-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24130159

RESUMEN

Purine nucleotide catabolism is common to most organisms and involves a guanine deaminase to convert guanine to xanthine in animals, invertebrates, and microorganisms. Using metabolomic analysis of mutants, we demonstrate that Arabidopsis thaliana uses an alternative catabolic route employing a highly specific guanosine deaminase (GSDA) not reported from any organism so far. The enzyme is ubiquitously expressed and deaminates exclusively guanosine and 2'-deoxyguanosine but no other aminated purines, pyrimidines, or pterines. GSDA belongs to the cytidine/deoxycytidylate deaminase family of proteins together with a deaminase involved in riboflavin biosynthesis, the chloroplastic tRNA adenosine deaminase Arg and a predicted tRNA-specific adenosine deaminase 2 in A. thaliana. GSDA is conserved in plants, including the moss Physcomitrella patens, but is absent in the algae and outside the plant kingdom. Our data show that xanthosine is exclusively generated through the deamination of guanosine by GSDA in A. thaliana, excluding other possible sources like the dephosphorylation of xanthosine monophosphate. Like the nucleoside hydrolases NUCLEOSIDE HYDROLASE1 (NSH1) and NSH2, GSDA is located in the cytosol, indicating that GMP catabolism to xanthine proceeds in a mostly cytosolic pathway via guanosine and xanthosine. Possible implications for the biosynthetic route of purine alkaloids (caffeine and theobromine) and ureides in other plants are discussed.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Nucleósido Desaminasas/metabolismo , Purinas/metabolismo , Ribonucleósidos/biosíntesis , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Clonación Molecular , ADN Bacteriano/genética , Metaboloma , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Nucleósido Desaminasas/genética , Plantas Modificadas Genéticamente/enzimología , Plantas Modificadas Genéticamente/genética , Xantinas
8.
Biochemistry ; 52(37): 6525-36, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23972005

RESUMEN

Proteins of unknown function belonging to cog1816 and cog0402 were characterized. Sav2595 from Steptomyces avermitilis MA-4680, Acel0264 from Acidothermus cellulolyticus 11B, Nis0429 from Nitratiruptor sp. SB155-2 and Dr0824 from Deinococcus radiodurans R1 were cloned, purified, and their substrate profiles determined. These enzymes were previously incorrectly annotated as adenosine deaminases or chlorohydrolases. It was shown here that these enzymes actually deaminate 6-aminodeoxyfutalosine. The deamination of 6-aminodeoxyfutalosine is part of an alternative menaquinone biosynthetic pathway that involves the formation of futalosine. 6-Aminodeoxyfutalosine is deaminated by these enzymes with catalytic efficiencies greater than 10(5) M(-1) s(-1), Km values of 0.9-6.0 µM, and kcat values of 1.2-8.6 s(-1). Adenosine, 2'-deoxyadenosine, thiomethyladenosine, and S-adenosylhomocysteine are deaminated at least an order of magnitude slower than 6-aminodeoxyfutalosine. The crystal structure of Nis0429 was determined and the substrate, 6-aminodeoxyfutalosine, was positioned in the active site on the basis of the presence of adventitiously bound benzoic acid. In this model, Ser-145 interacts with the carboxylate moiety of the substrate. The structure of Dr0824 was also determined, but a collapsed active site pocket prevented docking of substrates. A computational model of Sav2595 was built on the basis of the crystal structure of adenosine deaminase and substrates were docked. The model predicted a conserved arginine after ß-strand 1 to be partially responsible for the substrate specificity of Sav2595.


Asunto(s)
Nucleósido Desaminasas/metabolismo , Nucleósidos de Purina/metabolismo , Vitamina K 2/metabolismo , Actinomycetales/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Desaminación , Deinococcus/enzimología , Epsilonproteobacteria/enzimología , Epsilonproteobacteria/genética , Cinética , Modelos Moleculares , Simulación del Acoplamiento Molecular , Nucleósido Desaminasas/genética , Streptomyces/enzimología , Streptomyces/genética , Especificidad por Sustrato
9.
Anticancer Res ; 33(5): 1899-912, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23645737

RESUMEN

BACKGROUND/AIM: GS 9219 is a double prodrug of antiproliferative nucleotide analog 9-(2-Phosphonylmethoxyethyl)guanine (PMEG), with potent in vivo efficacy against various hematological malignancies. This study investigates the role of adenosine deaminase-like (ADAL) protein in the intracellular activation of GS-9219. MATERIALS AND METHODS: A cell line resistant to 9-(2-Phosphonylmethoxyethyl)-N(6)-cyclopropyl-2,6-diaminopurine (cPrPMEDAP), an intermediate metabolite of GS-9219, was generated and characterized. RESULTS: The resistant cell line was cross-resistant to cPrPMEDAP and GS-9219, due to a defect in the deamination of cPrPMEDAP to PMEG. Mutations in the ADAL gene (H286R and S180N) were identified in the resistant cells that adversely-affected its enzymatic activity. Introduction of the wild-type ADAL gene re-sensitized resistant cells to both cPrPMEDAP and GS-9219. CONCLUSION: The ADAL protein plays an essential role in the intracellular activation of GS-9219 by catalyzing the deamination of cPrPMEDAP metabolite to PMEG. Mutations affecting the activity of ADAL confer resistance to both GS-9219 and its metabolite cPrPMEDAP.


Asunto(s)
Adenina/análogos & derivados , Alanina/análogos & derivados , Resistencia a Antineoplásicos/genética , Mutación/genética , Nucleósido Desaminasas/genética , Purinas/farmacología , Neoplasias del Cuello Uterino/genética , Adenina/farmacología , Alanina/farmacología , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Western Blotting , Femenino , Humanos , Datos de Secuencia Molecular , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Profármacos/farmacología , Conformación Proteica , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/tratamiento farmacológico
10.
Angew Chem Int Ed Engl ; 51(44): 11166-9, 2012 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-23038402

RESUMEN

Checking for mistakes: By conjugating a catalytic domain with a guide RNA, deamination activity can be harnessed to repair a specific codon on mRNA. This method can be used for the highly selective repair of point mutations in mRNA by site-selective editing.


Asunto(s)
Nucleósido Desaminasas/química , Mutación Puntual , ARN/química , Adenosina Desaminasa/química , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Modelos Moleculares , Estructura Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , ARN/genética , ARN/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Ann N Y Acad Sci ; 1267: 79-85, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22954220

RESUMEN

Organisms minimize genetic damage through complex pathways of DNA repair. Yet a gene family--the AID/APOBECs--has evolved in vertebrates with the sole purpose of producing targeted damage in DNA/RNA molecules through cytosine deamination. They likely originated from deaminases involved in A>I editing in tRNAs. AID, the archetypal AID/APOBEC, is the trigger of the somatic diversification processes of the antibody genes. Its homologs may have been associated with the immune system even before the evolution of the antibody genes. The APOBEC3s, arising from duplication of AID, are involved in the restriction of exogenous/endogenous threats such as retroviruses and mobile elements. Another family member, APOBEC1, has (re)acquired the ability to target RNA while maintaining its ability to act on DNA. The AID/APOBECs have shaped the evolution of vertebrate genomes, but their ability to mutate nucleic acids is a double-edged sword: AID is a key player in lymphoproliferative diseases by triggering mutations and chromosomal translocations in B cells, and there is increasing evidence suggesting that other AID/APOBECs could be involved in cancer development as well.


Asunto(s)
Daño del ADN , Evolución Molecular , Genoma Humano , Nucleósido Desaminasas/fisiología , Animales , Humanos , Inmunoglobulinas/genética , Mutagénesis , Mutación , Neoplasias/genética , Nucleósido Desaminasas/genética , Filogenia , ARN/genética , ARN/metabolismo , Edición de ARN
12.
Curr Gene Ther ; 12(2): 77-91, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22384805

RESUMEN

Suicide gene therapy is an attractive strategy to selectively destroy cancer cells while minimizing unnecessary toxicity to normal cells. Since this idea was first introduced more than two decades ago, numerous studies have been conducted and significant developments have been made to further its application for mainstream cancer therapy. Major limitations of the suicide gene therapy strategy that have hindered its clinical application include inefficient directed delivery to cancer cells and the poor prodrug activation capacity of suicide enzymes. This review is focused on efforts that have been and are currently being pursued to improve the activity of individual suicide enzymes towards their respective prodrugs with particular attention to the application of nucleotide metabolizing enzymes in suicide cancer gene therapy. A number of protein engineering strategies have been employed and our discussion here will center on the use of mutagenesis approaches to create and evaluate nucleotide metabolizing enzymes with enhanced prodrug activation capacity and increased thermostability. Several of these studies have yielded clinically important enzyme variants that are relevant for cancer gene therapy applications because their utilization can serve to maximize cancer cell killing while minimizing the prodrug dose, thereby limiting undesirable side effects.


Asunto(s)
Enzimas/metabolismo , Terapia Genética/métodos , Neoplasias/terapia , Profármacos/uso terapéutico , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Enzimas/genética , Terapia Genética/tendencias , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Nucleotidasas/genética , Nucleotidasas/metabolismo , Nucleótidos/metabolismo , Profármacos/metabolismo , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/metabolismo
13.
Invest New Drugs ; 30(3): 1175-83, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21424698

RESUMEN

PURPOSE: Patients with metastatic pancreatic cancer have limited therapeutic options. The role of the Ras-Raf-MAPK pathway and of vascular endothelial growth factor in pancreatic carcinogenesis provided the rational to evaluate the efficacy of sorafenib with or without gemcitabine in a randomized phase II study. METHODS: Patients with metastatic pancreatic cancer were randomized to sorafenib alone (arm A) or sorafenib with gemcitabine (arm B). RESULTS: Arm A was closed to accrual at interim analysis due to the lack of objective response. Median PFS and OS were 2.3 and 4.3 months respectively. There was one partial response among the 37 patients in arm B. Median PFS and OS were 2.9 and 6.5 months respectively. There were more grade 3 and 4 toxicities in arm B with the most common being neutropenia (17%), thrombocytopenia (8%), alkaline phosphatase elevation (14%), venous thromboembolism (8%), diarrhea, hypokalemia and ALT elevation (5%) each. Several associations were noted between single nucleotide polymorphisms in ribonucleotide reductase, Cox-2, vascular endothelial growth factor and survival in patients treated with gemcitabine and sorafenib. CONCLUSIONS: Neither sorafenib alone or sorafenib in combination with gemcitabine manifested promising activity in metastatic pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclooxigenasa 2/genética , Nucleósido Desaminasas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bencenosulfonatos/administración & dosificación , Bencenosulfonatos/efectos adversos , Citidina Desaminasa , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Niacinamida/análogos & derivados , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Compuestos de Fenilurea , Polimorfismo de Nucleótido Simple , Piridinas/administración & dosificación , Piridinas/efectos adversos , Ribonucleósido Difosfato Reductasa , Sorafenib , Gemcitabina
14.
J Med Chem ; 54(16): 5902-14, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21755941

RESUMEN

Human N(6)-methyl-AMP/dAMP aminohydrolase has been shown to be involved in metabolism of pharmacologically important N(6)-substituted purine nucleosides and 5'-monophosphate prodrugs thereof. This enzyme was cloned and expressed in E. coli, and mass spectroscopic analysis followed by amino acid sequence analyses indicated that the protein was adenosine deaminase-like protein isoform 1 (ADAL1). An extensive structure-activity relationship study showed that ADAL1 was able to catalyze removal of different alkyl groups not only from N(6)-substituted purine or 2-aminopurine nucleoside monophosphates but also from O(6)-substituted compounds. The ADAL1 activity was susceptible to modifications in the phosphate moiety but not to changes in the sugar moiety. Overall, our data indicated that ADAL1 specifically acts at the 6-position of purine and 2-aminopurine nucleoside monophosphates. Our results may help designing of new therapeutic nucleoside/nucleotide prodrugs with desired metabolic profiles. Furthermore, amino acid sequence analysis in conjunction with crystallographic data and metal analysis suggested that ADAL1 contains a catalytic zinc ion. Finally, a potential physiological role of ADAL1 is discussed.


Asunto(s)
2-Aminopurina/metabolismo , Adenosina Monofosfato/metabolismo , Aminohidrolasas/metabolismo , Nucleósido Desaminasas/metabolismo , Purinas/metabolismo , 2-Aminopurina/química , Adenosina Monofosfato/química , Secuencia de Aminoácidos , Aminohidrolasas/química , Aminohidrolasas/genética , Biocatálisis , Línea Celular Tumoral , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Humanos , Hidrólisis , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Datos de Secuencia Molecular , Estructura Molecular , Peso Molecular , Nucleósido Desaminasas/genética , Fosforilación , Purinas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato , Zinc/química , Zinc/metabolismo
15.
Genes Cells ; 16(6): 714-27, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21605288

RESUMEN

Defects in the FANCJ/BRIP1 helicase gene are associated with genome instability disorders such as familial breast cancer or Fanconi anemia (FA). Although FANCJ has an in vitro activity to resolve G-quadruplex (G4) structures, and FANCJ ortholog in C. elegans prevents G4-associated deletions during replication, how FANCJ loss affects genome integrity in higher organisms remains unclear. Here, we report that FANCJ, but not other FA genes FANCD2 or FANCC, protected against large-scale genomic deletion that occurred frequently at the rearranged immunoglobulin heavy chain (IgH) locus in chicken DT40 cell line, suggesting that FancJ protects the genome independently of the FA ubiquitination pathway. In a more unbiased approach using array-comparative genomic hybridization, we identified de novo deletions as well as amplifications in fancj cells kept in culture for 2 months. A cluster of G4 sequence motifs was found near the breakpoint of one amplified region, but G4 sequence motifs were not detected at the breakpoints of two deleted regions. These results collectively suggest that, unlike in C. elegans, actions of vertebrate FANCJ to promote genome stability may not be limited to protection against the G4-mediated gene deletions.


Asunto(s)
Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Inestabilidad Genómica/genética , ARN Helicasas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Pollos , Hibridación Genómica Comparativa , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , G-Cuádruplex , Amplificación de Genes/genética , Conversión Génica/genética , Eliminación de Gen , Orden Génico , Reordenamiento Génico/genética , Marcación de Gen , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Datos de Secuencia Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , ARN Helicasas/genética , Alineación de Secuencia
16.
Biochemistry (Mosc) ; 76(1): 131-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21568845

RESUMEN

Enzymatic deamination of bases in DNA or RNA leads to an alteration of flow of genetic information. Adenosine deaminases edit RNA (ADARs, TADs). Specialized cytidine deaminases are involved in RNA/DNA editing in lipid metabolism (APOBEC1) and in innate (APOBEC3 family) and humoral (AID) immunity. APOBEC2 is required for proper muscle development and, along with AID, was implicated in demethylation of DNA. The functions of APOBEC4, APOBEC5, and other deaminases recently discovered by bioinformatics approaches are unknown. What is the basis for the diverse biological functions of enzymes with similar enzyme structure and the same principal enzymatic reaction? AID, APOBEC1, lamprey CDA1, and APOBEC3G enzymes cause uracil DNA glycosylase-dependent induction of mutations when overproduced ectopically in bacteria or yeast. APOBEC2, on the contrary, is nonmutagenic. We studied the effects of the expression of various deaminases in yeast and bacteria. The mutagenic specificities of four deaminases, hAID, rAPOBEC1, hAPOBEC3G, and lamprey CDA1, are strikingly different. This suggests the existence of an intrinsic component of deaminase targeting. The expression of yeast CDD1 and TAD2/TAD3, human APOBEC4, Xanthomonas oryzae APOBEC5, and deaminase encoded by Micromonas sp. gene MICPUN_56782 was nonmutagenic. A lack of a mutagenic effect for Cdd1 is expected because the enzyme functions in the salvage of pyrimidine nucleotides, and it is evolutionarily distant from RNA/DNA editing enzymes. The reason for inactivity of deaminases grouped with APOBEC2 is not obvious from their structures. This can not be explained by protein insolubility and peculiarities of cellular distribution and requires further investigation.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/genética , Proteínas Fúngicas/genética , Mutación , Nucleósido Desaminasas/genética , Levaduras/enzimología , Secuencias de Aminoácidos , Animales , Bacterias/química , Bacterias/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Humanos , Datos de Secuencia Molecular , Nucleósido Desaminasas/química , Nucleósido Desaminasas/metabolismo , Levaduras/química , Levaduras/genética
17.
Clin Chem ; 56(12): 1862-70, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20921267

RESUMEN

BACKGROUND: The nucleoside analog cytarabine (Ara-C [cytosine arabinoside]) is the key agent for treating acute myeloid leukemia (AML); however, up to 30% of patients fail to respond to treatment. Screening of patient blood samples to determine drug response before commencement of treatment is needed. This project aimed to construct and evaluate a self-bioluminescent reporter strain of Escherichia coli for use as an Ara-C biosensor and to design an in vitro assay to predict Ara-C response in clinical samples. METHODS: We used transposition mutagenesis to create a cytidine deaminase (cdd)-deficient mutant of E. coli MG1655 that responded to Ara-C. The strain was transformed with the luxCDABE operon and used as a whole-cell biosensor for development an 8-h assay to determine Ara-C uptake and phosphorylation by leukemic cells. RESULTS: Intracellular concentrations of 0.025 µmol/L phosphorylated Ara-C were detected by significantly increased light output (P < 0.05) from the bacterial biosensor. Results using AML cell lines with known response to Ara-C showed close correlation between the 8-h assay and a 3-day cytotoxicity test for Ara-C cell killing. In retrospective tests with 24 clinical samples of bone marrow or peripheral blood, the biosensor-based assay predicted leukemic cell response to Ara-C within 8 h. CONCLUSIONS: The biosensor-based assay may offer a predictor for evaluating the sensitivity of leukemic cells to Ara-C before patients undergo chemotherapy and allow customized treatment of drug-sensitive patients with reduced Ara-C dose levels. The 8-h assay monitors intracellular Ara-CTP (cytosine arabinoside triphosphate) levels and, if fully validated, may be suitable for use in clinical settings.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Técnicas Biosensibles , Citarabina/análisis , Escherichia coli , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/patología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Línea Celular Tumoral , Citarabina/farmacología , Citidina Desaminasa , Desoxicitidina Quinasa/biosíntesis , Desoxicitidina Quinasa/genética , Resistencia a Antineoplásicos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Humanos , Espacio Intracelular/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Mediciones Luminiscentes , Mutación , Nucleósido Desaminasas/genética , Fosforilación
18.
Nucleosides Nucleotides Nucleic Acids ; 29(4-6): 394-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20544525

RESUMEN

The deoxynucleoside analogs cytarabine (Ara-C) and gemcitabine (dFdC) are widely used in the treatment of cancer. Due to their hydrophilic nature they need the equilibrative (hENT) and concentrative (hCNT) nucleoside transporters to enter the cell. To bypass drug resistance due to decreased uptake, lipophilic 5'elaidic acid esters were synthesized, elacytarabine (CP-4055, from ara-C) and CP-4126 (from gemcitabine), which are currently in clinical development for solid and hematological tumors. We investigated whether resistance can be induced in vitro, and treated the CEM leukemic cell line with weekly increasing elacytarabine concentrations, up to 0.28 microM (10 times IC(50)). The IC(50) of the resistant CEM/CP-4055 was 35 microM, about 1,000 times that of the wildtype CEM, and comparable to that of CEM/dCK- (deoxycytidine kinase deficient) (22 microM). CEM/CP-4055 was also cross-resistant to Ara-C, gemcitabine and CP-4126 (28 and 33 microM, respectively). A low level of mRNA dCK was observed, and similar to CEM/dCK-, CEM/CP-4055 did not accumulate Ara-CTP after exposure to Ara-C or elacytarabine, which is consistent with a deficiency in dCK. In conclusion, elacytarabine induced resistance similar to Ara-C. This resistance was caused by downregulation of dCK.


Asunto(s)
Antineoplásicos/farmacología , Citarabina/análogos & derivados , Línea Celular Tumoral , Citarabina/farmacología , Citidina Desaminasa , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Leucemia/enzimología , Leucemia/genética , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/fisiología , Reacción en Cadena de la Polimerasa , Gemcitabina
19.
Oncol Rep ; 23(2): 471-5, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20043109

RESUMEN

The prognosis of pancreatic cancer remains poor, and the standard first-line chemotherapy with gemcitabine (GEM) has a response rate of less than 20%. Since expression of deoxycytidine kinase (dCK) seems important for improvement of GEM sensitivity, overexpression of dCK was investigated using pancreatic cancer cell lines (Panc-1, MIAPaCa-2 and BxPC-3). dCK gene was introduced into the cell lines by retrovirus and changes in IC50 were examined. Sensitivity of two pancreatic cancer cell lines to GEM elevated dramatically in comparison with control cells, but change of sensitivity remained at 1.8 times in BxPC-3. Since addition of tetrahydro uridine (THU), an inhibitor of deoxycytidine deaminase (CDA), increased the sensitivity 54-fold, overexpression of CDA seems to be the mechanism for improvement of the sensitivity. In conclusion, dCK is a key enzyme of GEM, but resistance of GEM is not improved in all pancreatic cancer cells by overexpression of dCK. Combination treatment based on expression of GEM metabolism-related gene may become an effective therapy in the future.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Desoxicitidina Quinasa/genética , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Antimetabolitos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Citidina Desaminasa , Desoxicitidina/uso terapéutico , Desoxicitidina Quinasa/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Modelos Biológicos , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/fisiología , Tetrahidrouridina/farmacología , Transfección , Regulación hacia Arriba/fisiología , Gemcitabina
20.
Pharmacogenomics ; 10(10): 1657-74, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19842938

RESUMEN

The mainstay of acute myeloid leukemia chemotherapy is the nucleoside analog cytarabine (ara-C). Numerous studies suggest that the intracellular concentrations of the ara-C active metabolite, ara-CTP, vary widely among patients and, in turn, are associated with variability in clinical response to acute myeloid leukemia treatment. Thus, genetic variation in key genes in the ara-C metabolic pathway--specifically, deoxycytidine kinase (a rate-limiting activating enzyme), 5 nucleotidase, cytidine deaminase and deoxycytidylate deaminase (all three are inactivating enzymes), human equilibrative nucleoside transporter (ara-C uptake transporter) and ribonucleotide reductase (RRM1 and RRM2--enzymes regulating intracellular deoxycytidine triphosphate pools)--form the molecular basis of the interpatient variability observed in intracellular ara-CTP concentrations and response to ara-C. Understanding genetic variants in the key candidate genes involved in the metabolic activation of ara-C, as well as the pharmacodynamic targets of ara-C, will provide an opportunity to identify patients at an increased risk of adverse reactions or decreased likelihood of response, based upon their genetic profile, which in future could help in dose optimization to reduce drug toxicity without compromising efficacy. The pharmacogenetic studies on ara-C would also be equally applicable to other nucleoside analogs, such as gemcitabine, decitabine, clofarabine and so on, which are metabolized by the same pathway.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Citarabina/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , 5'-Nucleotidasa/genética , Trifosfato de Arabinofuranosil Citosina/metabolismo , Citidina Desaminasa/genética , Desoxicitidina Quinasa/genética , Tranportador Equilibrativo 1 de Nucleósido/genética , Predicción , Variación Genética , Humanos , Leucemia Mieloide Aguda/genética , Nucleósido Desaminasas/genética , Farmacogenética/métodos , Ribonucleótido Reductasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA