Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 8): 1433-46, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23897467

RESUMEN

Arylamine N-acetyltransferase from Mycobacterium tuberculosis (TBNAT) plays an important role in the intracellular survival of the microorganism inside macrophages. Medicinal chemistry efforts to optimize inhibitors of the TBNAT enzyme have been hampered by the lack of a three-dimensional structure of the enzyme. In this paper, the first structure of TBNAT, determined using a lone crystal produced using cross-seeding with the homologous protein from M. marinum, is reported. Despite the similarity between the two enzymes (74% sequence identity), they show distinct physical and biochemical characteristics. The structure elegantly reveals the characteristic features of the protein surface as well as details of the active site of TBNAT relevant to drug-discovery efforts. The crystallographic analysis of the diffraction data presented many challenges, since the crystal was twinned and the habit possessed pseudo-translational symmetry.


Asunto(s)
Arilamina N-Acetiltransferasa/química , Arilamina N-Acetiltransferasa/metabolismo , Mycobacterium marinum/enzimología , Mycobacterium tuberculosis/enzimología , Arilamina N-Acetiltransferasa/aislamiento & purificación , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalización/métodos , Cristalografía por Rayos X , Estabilidad de Enzimas , Modelos Moleculares , Conformación Proteica , Dispersión del Ángulo Pequeño , Homología de Secuencia de Aminoácido
2.
Artículo en Inglés | MEDLINE | ID: mdl-22297998

RESUMEN

Arylamine N-acetyltransferases (NATs) are xenobiotic metabolizing enzymes (XMEs) that catalyze the acetylation of arylamines. All functional NATs described to date possess a strictly conserved Cys-His-Asp catalytic triad. Here, the purification, crystallization and preliminary X-ray characterization of Bacillus cereus arylamine N-acetyltransferase 3 [(BACCR)NAT3], a putative NAT isoenzyme that possesses a unique catalytic triad containing a glutamate residue, is reported. The crystal diffracted to 2.42 Å resolution and belonged to the monoclinic space group C121, with unit-cell parameters a = 90.44, b = 44.52, c = 132.98 Å, ß = 103.8°.


Asunto(s)
Arilamina N-Acetiltransferasa/química , Bacillus cereus/enzimología , Arilamina N-Acetiltransferasa/aislamiento & purificación , Cristalización , Cristalografía por Rayos X
3.
Protein Expr Purif ; 80(2): 246-52, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21767648

RESUMEN

Arylamine N-acetyltransferase from Mycobacterium tuberculosis (TBNAT) has been proposed as a drug target for latent tuberculosis treatment. The enzyme is essential for the survival of the mycobacterium in macrophages. However, TBNAT has been very difficult to generate as a soluble protein. In this work we describe production of soluble recombinant TBNAT at a reasonable yield achieved by subcloning the tbnat gene with a purification His-tag into the pVLT31 plasmid, and subsequent optimisation of the induction conditions. The expression system results in soluble protein optimised upon extended (60 h) low level isopropyl ß-D-1-thiogalactopyranoside level induction (100 µM) at a temperature of 15 °C. The level of TBNAT expression obtained in E. coli has been significantly improved from ∼2 mg to a final yield of up to 16 mg per litre of culture at a purity level suitable for structural studies. The molecular mass of 31310 Da was confirmed using mass spectroscopy and the oligomerisation state was determined. The stability of TBNAT in different buffer systems was investigated by thermal shift assays and sufficient protein is now available for the screening of chemical libraries for inhibitors.


Asunto(s)
Arilamina N-Acetiltransferasa/aislamiento & purificación , Proteínas Bacterianas/aislamiento & purificación , Mycobacterium tuberculosis/enzimología , Proteínas Recombinantes/aislamiento & purificación , Arilamina N-Acetiltransferasa/metabolismo , Proteínas Bacterianas/metabolismo , Tampones (Química) , Cromatografía en Gel , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Pruebas de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Histidina/metabolismo , Isopropil Tiogalactósido/farmacología , Peso Molecular , Plásmidos/genética , Plásmidos/metabolismo , Estabilidad Proteica , Proteínas Recombinantes/metabolismo , Solubilidad , Espectrometría de Masa por Ionización de Electrospray , Temperatura
4.
J Cell Biochem ; 103(2): 492-508, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17541948

RESUMEN

N-terminal acetylation is one of the most common modifications, occurring on the vast majority of eukaryotic proteins. Saccharomyces cerevisiae contains three major NATs, designated NatA, NatB, and NatC, with each having catalytic subunits Ard1p, Nat3p, and Mak3p, respectively. Gautschi et al. (Gautschi et al. [2003] Mol Cell Biol 23: 7403) previously demonstrated with peptide crosslinking experiments that NatA is bound to ribosomes. In our studies, biochemical fractionation in linear sucrose density gradients revealed that all of the NATs are associated with mono- and polyribosome fractions. However only a minor portion of Nat3p colocalized with the polyribosomes. Disruption of the polyribosomes did not cause dissociation of the NATs from ribosomal subparticles. The NAT auxiliary subunits, Nat1p and Mdm20p, apparently are required for efficient binding of the corresponding catalytic subunits to the ribosomes. Deletions of the genes corresponding to auxiliary subunits significantly diminish the protein levels of the catalytic subunits, especially Nat3p, while deletions of the catalytic subunits produced less effect on the stability of Nat1p and Mdm20p. Also two ribosomal proteins, Rpl25p and Rpl35p, were identified in a TAP-affinity purified NatA sample. Moreover, Ard1p copurifies with Rpl35p-TAP. We suggest that these two ribosomal proteins, which are in close proximity to the ribosomal exit tunnel, may play a role in NatA attachment to the ribosome.


Asunto(s)
Acetiltransferasas/metabolismo , N-Acetiltransferasa de Aminoácidos/metabolismo , Arilamina N-Acetiltransferasa/metabolismo , Mapeo de Interacción de Proteínas , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Acetiltransferasas/genética , Acetiltransferasas/aislamiento & purificación , N-Acetiltransferasa de Aminoácidos/genética , N-Acetiltransferasa de Aminoácidos/aislamiento & purificación , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Eliminación de Gen , Acetiltransferasa B N-Terminal , Acetiltransferasa C N-Terminal , Acetiltransferasas N-Terminal , Polirribosomas/química , Polirribosomas/metabolismo , Unión Proteica , Subunidades de Proteína , Proteínas Ribosómicas/aislamiento & purificación , Proteínas Ribosómicas/metabolismo , Ribosomas/química , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación
5.
J Mol Biol ; 375(1): 178-91, 2008 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-18005984

RESUMEN

Arylamine N-acetyltransferase (NAT) enzymes are widespread in nature. They serve to acetylate xenobiotics and/or endogenous substrates using acetyl coenzyme A (CoA) as a cofactor. Conservation of the architecture of the NAT enzyme family from mammals to bacteria has been demonstrated by a series of prokaryotic NAT structures, together with the recently reported structure of human NAT1. We report here the cloning, purification, kinetic characterisation and crystallographic structure determination of NAT from Mycobacterium marinum, a close relative of the pathogenic Mycobacterium tuberculosis. We have also determined the structure of M. marinum NAT in complex with CoA, shedding the first light on cofactor recognition in prokaryotic NATs. Surprisingly, the principal CoA recognition site in M. marinum NAT is located some 30 A from the site of CoA recognition in the recently deposited structure of human NAT2 bound to CoA. The structure explains the Ping-Pong Bi-Bi reaction mechanism of NAT enzymes and suggests mechanisms by which the acetylated enzyme intermediate may be protected. Recognition of CoA in a much wider groove in prokaryotic NATs suggests that this subfamily may accommodate larger substrates than is the case for human NATs and may assist in the identification of potential endogenous substrates. It also suggests the cofactor-binding site as a unique subsite to target in drug design directed against NAT in mycobacteria.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Coenzima A/metabolismo , Evolución Molecular , Variación Genética , Células Procariotas/enzimología , Secuencia de Aminoácidos , Arilamina N-Acetiltransferasa/química , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Clonación Molecular , Coenzima A/análisis , Secuencia Conservada , Cristalografía por Rayos X , Dimerización , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Espectrometría de Masas , Modelos Químicos , Modelos Moleculares , Datos de Secuencia Molecular , Mycobacterium/enzimología , Sistemas de Lectura Abierta , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
6.
Chem Res Toxicol ; 20(9): 1300-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17672512

RESUMEN

Arylamine N-acetyltransferases (NATs) are phase II xenobiotic metabolism enzymes that catalyze the detoxification of arylamines by N-acetylation and the bioactivation of N-arylhydroxylamines by O-acetylation. Endogenous and recombinant mammalian NATs with high specific activities are difficult to obtain in substantial quantities and in a state of homogeneity. This paper describes the overexpression of human wild-type NAT2 as a dihydrofolate reductase fusion protein containing a TEV protease-sensitive linker. Treatment of the partially purified fusion protein with TEV protease, followed by chromatographic purification, afforded 2.8 mg of homogeneous NAT2 from 2 L of cell culture. The kinetic specificity constants ( k cat/ K m) for N-acetylation of arylamine environmental contaminants, some of which are associated with bladder cancer risk, were determined with NAT2 and NAT1. The NAT1/NAT2 ratio of the specificity constants varied almost 1000-fold for monosubstituted and disubstituted alkylanilines containing methyl and ethyl ring substituents. 2-Alkyl substituents depressed N-acetylation rates but were more detrimental to catalysis by NAT1 than by NAT2. 3-Alkyl groups caused substrates to be preferentially N-acetylated by NAT2, and both 4-methyl- and 4-ethylaniline were better substrates for NAT1 than NAT2. NMR-based models were used to analyze the NAT binding site interactions of the alkylanilines. The selectivity of NAT1 for acetylation of 4-alkylanilines appears to be due to binding of the substituents to V216, which is replaced by S216 in NAT2. The contribution of 3-alkyl substituents to NAT2 substrate selectivity is attributed to multiple bonding interactions with F93, whereas a single bonding interaction occurs with V93 in NAT1. Unfavorable steric clashes between 2-methyl substituents and F125 of NAT1 may account for the selective NAT2-mediated N-acetylation of 2-alkylanilines; F125 is replaced by S125 in NAT2. These results provide insight into the structural basis for the substrate specificity of two NATs that play major roles in the biotransformation of genotoxic environmental arylamines.


Asunto(s)
Compuestos de Anilina/química , Arilamina N-Acetiltransferasa/química , Isoenzimas/biosíntesis , Acetilación , Arilamina N-Acetiltransferasa/biosíntesis , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Clonación Molecular , Contaminantes Ambientales/química , Humanos , Isoenzimas/genética , Isoenzimas/aislamiento & purificación , Espectroscopía de Resonancia Magnética , Modelos Biológicos , Estructura Molecular , Unión Proteica , Proteínas Recombinantes/genética , Especificidad por Sustrato
7.
J Biosci Bioeng ; 103(2): 147-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17368397

RESUMEN

A bacterium, strain 10-L-2, that was isolated from soil and identified as Bacillus cereus grew well on medium containing 4-phenylenediamine and Polypepton. Strain 10-L-2 converted a wide variety of anilines, including 4-phenylenediamine, to their corresponding acetanilides. Growing cells acetylated a single amino group of 4-phenylenediamine to form 4-aminoacetanilide with a 97% molar yield, as shown by mass spectrometry and HPLC. Cell extracts exhibited arylamine N-acetyltransferase (NAT) activity toward 4-phenylenediamine. Two NATs, namely, NAT-a and NAT-b, were separated by DE52 column chromatography and were further purified and characterized. The subunit molecular masses of NAT-a and NAT-b were 31.0 and 27.5 kDa, respectively, as determined by SDS-PAGE analysis. The two enzymes had similar pH- and thermo-stabilities and were similarly affected by pH, temperature, and several reagents. The enzymes showed peak activity toward 5-aminosalicylic acid of the substrates tested, but they differed in substrate specificity. Only NAT-a had activity toward sulfamethazine. Although other wild-type bacterial cultures also synthesize NAT, the ability of strain 10-L-2 to convert and detoxify 4-phenylenediamine is much higher. This report provides the first evidence of two NATs in a eubacterium.


Asunto(s)
Acetanilidas/metabolismo , Arilamina N-Acetiltransferasa/química , Bacillus cereus/enzimología , Fenilendiaminas/química , Secuencia de Aminoácidos , Compuestos de Anilina/química , Compuestos de Anilina/metabolismo , Arilamina N-Acetiltransferasa/aislamiento & purificación , Bacillus cereus/aislamiento & purificación , Biotransformación , Datos de Secuencia Molecular , Fenilendiaminas/metabolismo , Especificidad por Sustrato
8.
Protein J ; 24(2): 65-77, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16003948

RESUMEN

Human arylamine N-acetyltransferase 1 (NAT1) has been overexpressed in E. coli as a mutant dihydrofolic acid reductase (DHFR) fusion protein with a thrombin sensitive linker. An initial DEAE anion-exchange chromatography resulted in partial purification of the fusion protein. The fusion protein was cleaved with thrombin, and human rNAT1 was purified with a second DEAE column. A total of 8 mg of human rNAT1 from 2 1 of cell culture was purified to homogeneity with this methodology. Arylamine substrate specificities were determined for human rNATI and hamster rNAT2. With both NATs, the second order rate constants (k(cat)/ Kmb) for p-aminobenzoic acid (PABA) and 2-aminofluorene (2-AF) were several thousand-fold higher than those for procainamide (PA), consistent with the expected substrate specificities of the enzymes. However, p-aminosalicylic acid (PAS), previously reported to be a human NAT1 and hamster NAT2 selective substrate, exhibits 20-fold higher specificity for hamster rNAT2 (k(cat)/Kmb 3410 microM(-1) s(-1)) than for human rNAT1 (k(cat)/Kmb 169.4 microM(-1) s(-1)). p-aminobenzoyl-glutamic acid (pABglu) was acetylated 10-fold more efficiently by human rNAT1 than by hamster rNAT2. Inhibition studies of human rNAT1 and hamster rNAT2 revealed that folic acid and methotrexate (MTX) are competitive inhibitors of both the unacetylated and acetylated forms of the enzymes, with K(I) values in 50 - 300 micro range. Dihydrofolic acid (DHF) was a much poorer inhibitor of human rNAT1 than of hamster rNAT2. The combined results demonstrate that human rNAT1 and hamster rNAT2 have similar but distinct kinetic properties with certain substrates, and suggest that folic acid, at least in the non-polyglutamate form, may not have an effect on human NAT1 activity in vivo.


Asunto(s)
Arilamina N-Acetiltransferasa/genética , Animales , Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Arilamina N-Acetiltransferasa/aislamiento & purificación , Arilamina N-Acetiltransferasa/metabolismo , Secuencia de Bases , Cromatografía DEAE-Celulosa , Cromatografía Líquida de Alta Presión , Clonación Molecular , Cricetinae , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Ácido Fólico/análogos & derivados , Ácido Fólico/farmacología , Humanos , Plásmidos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato
9.
Ai Zheng ; 24(7): 893-7, 2005 Jul.
Artículo en Chino | MEDLINE | ID: mdl-16004823

RESUMEN

BACKGROUND & OBJECTIVE: Using DNA samples obtained from buccal cells for genetic polymorphism analysis in molecular epidemiological studies has been repeatedly reported, but whether DNA from food remnants in mouth influences the result is still concerned. This study was to compare genetic polymorphisms of buccal cell DNA with those of buffy coat DNA, and with plant and animal DNA from foods to rule out the possibility of interference from food remnants, to improve technique of buccal cell collection and elevate DNA yield. METHODS: Buccal cells were collected from mouthwash (40 ml/case) of 62 subjects, and fixed with isopropyl alcohol; buffy coats of peripheral blood were collected from 30 of these subjects. Common foods (rice, greengrocery, soybean, apple, pork, beef, chicken, and duck) were also collected. DNA of all samples was extracted by chloroform-phenol method. NAT2, GSTM1, GSTT1, CYP1A1, and CYP2E1 genetic polymorphisms were assayed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique. Alu (human mutual DNA sequence) was also tested. RESULTS: DNA yield of 62 individual mouthwash samples was (135.15+/-64.30) microg (22.36-330.70 microg); 30 individual mouthwash samples contained 75%-95% oral epithelial cells with DNA yield of (143.44+/-61.64) microg (51.01-283.58 microg). DNA yield of 30 buffy coat samples was (91.19+/-38.01) microg (30.83-178.63 microg). Electrophoresis showed that all 62 buccal cell samples and 30 buffy coat samples contained DNA fragments in high molecular weight; beta-globin, Alu, NAT2, GSTM1, GSTT1, CYP1A1, and CYP2E1 gene fragments were successfully amplified from 61 buccal cells samples and 30 buffy coat samples, which showed no difference between the 2 kinds of samples from individual collections; these gene fragments were not amplified from all food DNA samples. CONCLUSIONS: The majority of DNA from mouthwash is human-origin. A little amount of food remnants would not influence the measurements of genetic polymorphisms. The genetic polymorphisms show no difference between buccal cell samples and buffy coat samples.


Asunto(s)
ADN/genética , Genoma Humano , Mucosa Bucal/citología , Polimorfismo Genético , Anciano , Elementos Alu/genética , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , ADN/aislamiento & purificación , Sondas de ADN , Estudios Epidemiológicos , Frecuencia de los Genes , Glutatión Transferasa/genética , Glutatión Transferasa/aislamiento & purificación , Humanos , Masculino , Antisépticos Bucales/análisis , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción
10.
Chem Res Toxicol ; 18(2): 183-97, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15720122

RESUMEN

Arylamine N-acetyltransferases (NATs) catalyze the N-acetylation of arylamines, the O-acetylation of N-arylhydroxylamines, and the conversion of N-(aryl)acetohydroxamic acids to N-acetoxyarylamines. NATs also undergo irreversible inactivation in the presence of N-(aryl)acetohydroxamic acids. We previously established that inactivation of hamster NAT1 by N-hydroxy-2-acetylaminofluorene is the result of sulfinamide adduct formation with Cys68. The purpose of this research was to determine the kinetics of inactivation of hamster NAT1, hamster NAT2, and human NAT1 by N-hydroxy-4-acetylaminobiphenyl (N-OH-4-AABP), to identify the amino acids that are modified upon NAT-catalyzed bioactivation of N-OH-4-AABP, to characterize the adducts and to identify factors that influence the propensity of NATs to undergo inactivation by N-arylhydroxamic acids. Mass spectrometric analysis of the NATs, after incubation with N-OH-4-AABP, revealed that the principal adduct of each protein was a (4-biphenyl)sulfinamide. Proteolysis of the adducted NATs caused hydrolysis of the sulfinamides to sulfinic acids. Tandem mass spectrometric analysis of the modified peptides revealed that each NAT isozyme contained a sulfinic acid on the Cys68 side chain. Minor adducts were identified as 4-aminobiphenyl conjugates of tyrosines. Hamster NAT1 was more rapidly inactivated by N-OH-4-AABP than either hamster NAT2 or human NAT1, and it was demonstrated that 4-nitrosoobiphenyl, which forms the sulfinamide adducts, accumulates during incubation of N-OH-4-AABP with hamster NAT2 and human NAT1 but not during incubations with hamster NAT1. Steady state kinetic analysis of the hydrolysis of acetylated NATs revealed that the half-lives of acetylated hamster NAT2 and human NAT1 are 7-8-fold greater than that of acetylated hamster NAT1. These results support the proposal that the mechanism of inactivation of NATs by N-OH-4-AABP involves initial deacetylation to produce N-OH-4-aminobiphenyl, which after oxidative conversion to 4-nitrosobiphenyl reacts with Cys68 to form a sulfinamide. The relatively short half-life of the acetylated form of hamster NAT1 contributes to its greater susceptibility to inactivation by N-OH-4-AABP.


Asunto(s)
Compuestos de Aminobifenilo/farmacología , Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Animales , Arilamina N-Acetiltransferasa/biosíntesis , Arilamina N-Acetiltransferasa/aislamiento & purificación , Catálisis , Cricetinae , Relación Dosis-Respuesta a Droga , Humanos , Hidrólisis , Isoenzimas/antagonistas & inhibidores , Isoenzimas/biosíntesis , Isoenzimas/aislamiento & purificación , Estructura Molecular , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Factores de Tiempo
11.
Biochem Pharmacol ; 69(2): 347-59, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15627487

RESUMEN

Arylamine N-acetyltransferases (NAT; EC 2.3.1.5) catalyse the transfer of acetyl groups from acetylCoA to xenobiotics, including drugs and carcinogens. The enzyme is found extensively in both eukaryotes and prokaryotes, yet the endogenous roles of NATs are still unclear. In order to study the properties of eukaryotic NATs, high-throughput substrate and inhibitor screens have been developed using pure soluble recombinant Syrian hamster NAT2 (shNAT2) protein. The assay can be used with a wide range of compounds and was used to determine substrate specificity of shNAT2. We describe the expression and characterisation of shNAT2 and also purified recombinant human NAT1 and NAT2, including the use of the assay to explore the substrate specificities of each of the enzymes. Hamster NAT2 has similar substrate specificity to human NAT1, acetylating para-aminobenzoate but not arylhydrazine and hydralazine compounds. The overlapping but distinct substrate-specific activity profiles of human NAT1 and NAT2 were clearly observed from the screen. Naturally occurring compounds were tested as substrates or inhibitors of shNAT2 and succinylCoA was found to be a potent inhibitor of shNAT2.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Células Eucariotas/enzimología , Animales , Arilamina N-Acetiltransferasa/aislamiento & purificación , Cromatografía en Gel , Cricetinae , Células Eucariotas/química , Humanos , Cinética , Luz , Conejos , Proteínas Recombinantes/análisis , Proteínas Recombinantes/metabolismo , Dispersión de Radiación , Especificidad por Sustrato/fisiología
12.
Artículo en Inglés | MEDLINE | ID: mdl-16508079

RESUMEN

The arylamine N-acetyltransferase (NAT) enzymes have been found in a broad range of both eukaryotic and prokaryotic organisms. The NAT enzymes catalyse the transfer of an acetyl group from acetyl Co-enzyme A onto the terminal nitrogen of a range of arylamine, hydrazine and arylhydrazine compounds. Recently, several NAT structures have been reported from different prokaryotic sources including Salmonella typhimurium, Mycobacterium smegmatis and Pseudomonas aeruginosa. Bioinformatics analysis of the Mesorhizobium loti genome revealed two NAT paralogues, the first example of multiple NAT isoenzymes in a eubacterial organism. The M. loti NAT 1 enzyme was recombinantly expressed and purified for X-ray crystallographic studies. The purified enzyme was crystallized in 0.5 M Ca(OAc)2, 16% PEG 3350, 0.1 M Tris-HCl pH 8.5 using the sitting-drop vapour-diffusion method. A data set diffracting to 2.0 A was collected from a single crystal at 100 K. The crystal belongs to the orthorhombic spacegroup P2(1)2(1)2(1), with unit-cell parameters a = 53.2, b = 97.3, c = 114.3 A. The structure was refined to a final free-R factor of 24.8%. The structure reveals that despite low sequence homology, M. loti NAT1 shares the common fold as reported in previous NAT structures and exhibits the same catalytic triad of residues (Cys-His-Asp) in the active site.


Asunto(s)
Alphaproteobacteria/enzimología , Arilamina N-Acetiltransferasa/química , Secuencia de Aminoácidos , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Sitios de Unión , Clonación Molecular , Modelos Moleculares , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
13.
J Microbiol Immunol Infect ; 37(4): 208-15, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15340647

RESUMEN

Arylamine N-acetyltransferase (NAT) enzymes have been found in laboratory animals, humans, microorganisms (fungi, bacteria and parasites), and in plants. But the characteristics of NAT from Klebsiella pneumoniae are not clear. NAT activities with p-aminobenzoic acid (PABA) and 2-aminofluorene (AF) as substrates were examined in the cytosol of K. pneumoniae. NAT activity (N-acetylation of substrates) was determined using an acetyl coenzyme A recycling assay and high performance liquid chromatography for determining the amounts of acetylated or non-acetylated PABA or AF. NAT activities from a number of K. pneumoniae isolates were found to be 0.72 +/- 0.08 nmol/min/mg protein for AF, and 0.49 +/- 0.04 nmol/min/mg protein for PABA. The kinetic parameters of apparent Michaelis constant (Km) and maximum velocity (Vmax) obtained were 2.92 +/- 0.48 mM and 7.89 +/- 0.82 nmol/min/mg protein, respectively, for AF and 2.42 +/- 0.28 mM and 9.87 +/- 0.64 nmol/min/mg protein, respectively, for PABA. The optimal pH value for the NAT activity was 7.0 for AF and PABA. The optimal temperature for NAT activity was 37 degrees C for both substrates. The NAT activity was inhibited by 50% with 0.25 mM iodoacetamide, and by more than 90% at 1.0 mM. Among a series of divalent cations and salts, Cu2+ and Zn2+ were the most potent inhibitors of NAT activity.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Klebsiella pneumoniae/enzimología , Ácido 4-Aminobenzoico/metabolismo , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Secuencia de Bases , ADN Bacteriano/genética , Fluorenos/metabolismo , Concentración de Iones de Hidrógeno , Yodoacetamida/farmacología , Cinética , Klebsiella pneumoniae/genética , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales (Química)/farmacología , Especificidad por Sustrato , Temperatura
14.
Chem Res Toxicol ; 17(3): 275-86, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15025497

RESUMEN

Arylamine N-acetyltransferases (NATs) are expressed in most mammalian tissues. NATs catalyze the N-acetylation of primary arylamines, the O-acetylation of N-arylhydroxylamines, and the N,O-transacetylation of N-arylhydroxamic acids. The latter two reactions result in formation of reactive, electrophilic N-acetoxyarylamines, which are considered to be the ultimate carcinogenic metabolites of certain environmental and dietary arylamines. Incubation of various N-(aryl)acetohydroxamic acids, such as N-hydroxy-2-acetylaminofluorene (N-OH-AAF) with hamster NAT1, results in time-dependent, concentration-dependent, and kinetically first-order irreversible inactivation of the enzyme. N-OH-AAF also causes in vivo inactivation of NAT1. The purpose of this research was to investigate the molecular mechanism of NAT1 inactivation by identifying the amino acid residues that undergo covalent modification upon NAT1-catalyzed bioactivation of N-OH-AAF and by characterizing the chemical structures of the adducts. Electrospray ionization quadrupole time-of-flight mass spectrometric analysis of NAT1 that had been incubated with N-OH-AAF revealed that the mass of the major adduct (+195 Da) was consistent with a (2-fluorenyl)sulfinamide modification. The major adduct underwent hydrolysis to yield a protein with a molecular mass that corresponded to a sulfinic acid-modified NAT1. Treatment of NAT1 with 2-nitrosofluorene resulted in a modification (+195 Da) that was identical in mass to that obtained with N-OH-AAF-inactivated enzyme. Matrix-assisted laser desorption-ionization quadrupole time-of flight tandem mass spectrometric (MALDI Q-TOF MS/MS) analysis revealed that the modified residue was the catalytically essential Cys68. MALDI Q-TOF MS/MS sequencing of peptides from protease digests of inactivated NAT1 also identified two minor adducts at Tyr17 and Tyr186, each of which was covalently conjugated with 2-aminofluorene. Thus, the mechanism of inactivation of NAT1 by N-OH-AAF involves NAT1-catalyzed deacetylation to afford N-hydroxy-2-aminofluorene, which after oxidative conversion to 2-nitrosofluorene, forms a sulfinamide adduct by reacting with Cys68. GSH had little effect on the inactivation of NAT1 by N-OH-AAF, although high concentrations of cysteine attenuated both the extent of inactivation and the sulfinamide adduct formation.


Asunto(s)
Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hidroxiacetilamino Fluoreno/farmacología , Animales , Arilamina N-Acetiltransferasa/aislamiento & purificación , Cricetinae , Relación Dosis-Respuesta a Droga , Proteínas Recombinantes , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Especificidad por Sustrato
15.
Bioorg Med Chem ; 11(7): 1227-34, 2003 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-12628650

RESUMEN

Arylamine N-acetyltransferases (NATs) catalyse the acetylation of arylamine, arylhydrazine and arylhydroxylamine substrates by acetyl Coenzyme A. NAT has been discovered in a wide range of eukaryotic and prokaryotic species. Although prokaryotic NATs have been implicated in xenobiotic metabolism, to date no endogenous role has been identified for the arylamine N-acetyl transfer reaction in prokaryotes. Investigating the substrate specificity of these enzymes is one approach to determining a possible endogenous role for prokaryotic NATs. We describe an accurate and efficient assay for NAT activity that is suitable for high-throughput screening of potential NAT ligands. This assay has been utilised to identify novel substrates for pure NAT from Salmonella typhimurium and Mycobacterium smegmatis which show a relationship between the lipophilicity of the arylamine and its activity as a substrate. The lipophilic structure/activity relationship observed is proposed to depend on the topology of the active site using docking studies of the crystal structures of these NAT isoenzymes. The evidence suggests an endogenous role of NAT in the protection of bacteria from aromatic and lipophilic toxins.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Bacterias/enzimología , Acetilación , Arilamina N-Acetiltransferasa/aislamiento & purificación , Coenzima A/metabolismo , Cristalografía por Rayos X , Ácido Ditionitrobenzoico , Escherichia coli/metabolismo , Hidralazina/farmacología , Hidrólisis , Cinética , Modelos Moleculares , Mycobacterium smegmatis/enzimología , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/enzimología , Relación Estructura-Actividad , Especificidad por Sustrato
16.
Insect Biochem Mol Biol ; 32(1): 15-22, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11719065

RESUMEN

We separated two forms of arylalkylamine N-acetyltransferase (AANAT) from various organs of the American cockroach, Periplaneta americana. Both forms of the enzyme had an equivalent molecular mass of 28 kDa. One form isolated from the testicular accessory glands had high enzyme activity at acidic pHs. The isoelectric point was 5-6 and the substrate specificity was wider than the other type. The other isolated form from female midguts had a higher level of enzyme activity at basic pHs. These findings suggested that P. americana contains polymorphic AANAT, as is the case in Drosophila melanogaster. These forms differed not only in pH specificity, and substrate specificity but in chromatographic behavior and kinetic properties. Most of the organs we examined contained a mixture of the two forms since two types of AANAT activity were separated in different chromatographic fractions when two pH conditions were used for activity measurement.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Periplaneta/enzimología , Polimorfismo Genético , Animales , Arilamina N-Acetiltransferasa/aislamiento & purificación , Cromatografía Liquida/métodos , Sistema Digestivo/enzimología , Femenino , Cinética , Masculino , Periplaneta/genética , Especificidad por Sustrato
17.
Trends Pharmacol Sci ; 22(3): 140-6, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11239577

RESUMEN

Arylamine N-acetyltransferases (NATs) catalyse the transfer of an acetyl group from acetyl CoA to the terminal nitrogen of hydrazine and arylamine drugs and carcinogens. These enzymes are polymorphic and have an important place in the history of pharmacogenetics, being first identified as responsible for the polymorphic inactivation of the anti-tubercular drug isoniazid. NAT has recently been identified within Mycobacterium tuberculosis itself and is an important candidate for modulating the response of mycobacteria to isoniazid. The first three-dimensional structure of the unique NAT family shows the active-site cysteine to be aligned with conserved histidine and aspartate residues to form a catalytic triad, thus providing an activation mechanism for transfer of the acetyl group from acetyl CoA to cysteine. The unique fold could allow different members of the NAT family to play a variety of roles in endogenous and xenobiotic metabolism.


Asunto(s)
Arilamina N-Acetiltransferasa , Alelos , Animales , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/aislamiento & purificación , Arilamina N-Acetiltransferasa/metabolismo , Arilamina N-Acetiltransferasa/fisiología , Humanos , Ratones , Mutación , Mycobacterium smegmatis/enzimología , Mycobacterium tuberculosis/enzimología , Polimorfismo Genético , Conejos , Ratas
18.
DNA Cell Biol ; 19(11): 697-705, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11098219

RESUMEN

In insects, amine acetylation by the enzyme arylalkylamine N-acetyltransferase (AANAT) is involved in melatonin formation, sclerotization, and neurotransmitter inactivation. This wide spectrum of activities suggests that several AANAT enzymes are present. We recently purified a protein fraction with AANAT activity from Drosophila melanogaster and cloned the corresponding gene, aaNAT1. Following the same strategy, we now report the purification of an additional AANAT from D. melanogaster, AANAT2, and the cloning of the corresponding cDNA. The isolated protein differs from AANAT1a and AANAT1b in its molecular weight and isoelectric point. The AANAT2 shares about 30% identity with the products of the aaNAT1 gene. The enzyme does not follow one-site Michaelis-Menten kinetics when assayed with various concentrations of the arylalkylamine tryptamine and a constant concentration (0.5 mM) of the cofactor acetyl coenzyme A. The data can be interpreted in terms of an enzyme with two kinetic regimes (K(m1) = 7.2 microM, K(m2) = 0.6 mM, and v(max2) = 2.7 v(max1)) that are governed by binding of the substrate to a regulatory site (K(r) = 6.2 mM). These findings demonstrate the presence of a second expressed gene encoding an AANAT in D. melanogaster. Northern blot analysis revealed no diurnal variation of aaNAT2 transcription, similar to the results obtained for aaNAT1a and aaNAT1b.


Asunto(s)
Arilamina N-Acetiltransferasa/genética , Drosophila melanogaster/genética , Acetilación , Secuencia de Aminoácidos , Animales , Arilamina N-Acetiltransferasa/aislamiento & purificación , Arilamina N-Acetiltransferasa/metabolismo , Secuencia de Bases , Células COS , Ritmo Circadiano , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , ADN Recombinante/genética , ADN Recombinante/metabolismo , Drosophila melanogaster/enzimología , Regulación Enzimológica de la Expresión Génica , Cabeza , Isoenzimas/genética , Cinética , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
19.
J Biol Chem ; 275(12): 8794-805, 2000 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-10722724

RESUMEN

Arylalkylamine N-acetyltransferase (AANAT) catalyzes the reaction of serotonin with acetyl-CoA to form N-acetylserotonin and plays a major role in the regulation of the melatonin circadian rhythm in vertebrates. In the present study, the human cloned enzyme has been expressed in bacteria, purified, cleaved, and characterized. The specificity of the human enzyme toward substrates (natural as well as synthetic arylethylamines) and cosubstrates (essentially acyl homologs of acetyl-CoA) has been investigated. Peptide combinatorial libraries of tri-, tetra-, and pentapeptides with various amino acid compositions were also screened as potential sources of inhibitors. We report the findings of several peptides with low micromolar inhibitory potency. For activity measurement as well as for specificity studies, an original and rapid method of analysis was developed. The assay was based on the separation and detection of N-[(3)H]acetylarylethylamine formed from various arylethylamines and tritiated acetyl-CoA, by means of high performance liquid chromatography with radiochemical detection. The assay proved to be robust and flexible, could accommodate the use of numerous synthetic substrates, and was successfully used throughout this study. We also screened a large number of pharmacological bioamines among which only one, tranylcypromine, behaved as a substrate. The synthesis and survey of simple arylethylamines also showed that AANAT has a large recognition pattern, including compounds as different as phenyl-, naphthyl-, benzothienyl-, or benzofuranyl-ethylamine derivatives. An extensive enzymatic study allowed us to pinpoint the amino acid residue of the pentapeptide inhibitor, S 34461, which interacts with the cosubstrate-binding site area, in agreement with an in silico study based on the available coordinates of the hAANAT crystal.


Asunto(s)
Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Arilamina N-Acetiltransferasa/metabolismo , Acilcoenzima A/farmacología , Aminas/metabolismo , Animales , N-Acetiltransferasa de Arilalquilamina , Arilamina N-Acetiltransferasa/aislamiento & purificación , Dominio Catalítico , Cromatografía Líquida de Alta Presión/métodos , Escherichia coli/genética , Humanos , Espectrometría de Masas , Modelos Moleculares , Oligopéptidos/farmacología , Ovinos , Especificidad de la Especie , Relación Estructura-Actividad , Especificidad por Sustrato
20.
Anal Biochem ; 275(1): 93-7, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10542113

RESUMEN

Three polymorphic sites in the N-acetyltransferase 2 (NAT2) gene were detected using rapid cycle DNA amplification with allele-specific fluorescent probes and melting curve analysis. Two fluorogenic adjacent hybridization probes were designed to NAT2*5A (C(481)T), NAT2*6A (G(590)A), and NAT2*7A (G(857)A). During amplification, probe hybridization is observed as fluorescence resonance energy transfer. The fluorescence increases every cycle as the product accumulates during amplification. A single base mismatch resulted in a melting temperature shift (T(m)) of 5 to 6 degrees C, allowing for the easy distinction of a wild-type allele from the mutant allele. The protocol is rapid, requiring 40 min for the completion of 45 cycles including the melting curves. It is also a simple and flexible method, since DNA templates prepared from different sources, including DNA from serum and paraffin-embedded tissue sections, could be used without adverse effects. Fluorescence genotyping of all three polymorphisms in a total of 155 DNA samples correlated perfectly with our previously validated genotyping by restriction enzyme digestion (PCR-RFLP). This new facile approach allows for the easy detection of NAT2 polymorphisms in hundreds of samples in only a day.


Asunto(s)
Arilamina N-Acetiltransferasa/aislamiento & purificación , Reacción en Cadena de la Polimerasa/métodos , Arilamina N-Acetiltransferasa/genética , Colorantes Fluorescentes , Genotipo , Humanos , Polimorfismo Genético , Polimorfismo de Longitud del Fragmento de Restricción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...