Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Leukemia ; 30(12): 2322-2331, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27211273

RESUMEN

The splicing factor SF3B1 is the most frequently mutated gene in myelodysplastic syndromes (MDS), and is strongly associated with the presence of ring sideroblasts (RS). We have performed a systematic analysis of cryptic splicing abnormalities from RNA sequencing data on hematopoietic stem cells (HSCs) of SF3B1-mutant MDS cases with RS. Aberrant splicing events in many downstream target genes were identified and cryptic 3' splice site usage was a frequent event in SF3B1-mutant MDS. The iron transporter ABCB7 is a well-recognized candidate gene showing marked downregulation in MDS with RS. Our analysis unveiled aberrant ABCB7 splicing, due to usage of an alternative 3' splice site in MDS patient samples, giving rise to a premature termination codon in the ABCB7 mRNA. Treatment of cultured SF3B1-mutant MDS erythroblasts and a CRISPR/Cas9-generated SF3B1-mutant cell line with the nonsense-mediated decay (NMD) inhibitor cycloheximide showed that the aberrantly spliced ABCB7 transcript is targeted by NMD. We describe cryptic splicing events in the HSCs of SF3B1-mutant MDS, and our data support a model in which NMD-induced downregulation of the iron exporter ABCB7 mRNA transcript resulting from aberrant splicing caused by mutant SF3B1 underlies the increased mitochondrial iron accumulation found in MDS patients with RS.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Síndromes Mielodisplásicos/genética , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Secuencia de Bases , Cicloheximida/farmacología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hierro/metabolismo , Mitocondrias/metabolismo , Empalme del ARN , Células Tumorales Cultivadas
2.
Leukemia ; 27(3): 610-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22936014

RESUMEN

Gene expression profiling signatures may be used to classify the subtypes of Myelodysplastic syndrome (MDS) patients. However, there are few reports on the global methylation status in MDS. The integration of genome-wide epigenetic regulatory marks with gene expression levels would provide additional information regarding the biological differences between MDS and healthy controls. Gene expression and methylation status were measured using high-density microarrays. A total of 552 differentially methylated CpG loci were identified as being present in low-risk MDS; hypermethylated genes were more frequent than hypomethylated genes. In addition, mRNA expression profiling identified 1005 genes that significantly differed between low-risk MDS and the control group. Integrative analysis of the epigenetic and expression profiles revealed that 66.7% of the hypermethylated genes were underexpressed in low-risk MDS cases. Gene network analysis revealed molecular mechanisms associated with the low-risk MDS group, including altered apoptosis pathways. The two key apoptotic genes BCL2 and ETS1 were identified as silenced genes. In addition, the immune response and micro RNA biogenesis were affected by the hypermethylation and underexpression of IL27RA and DICER1. Our integrative analysis revealed that aberrant epigenetic regulation is a hallmark of low-risk MDS patients and could have a central role in these diseases.


Asunto(s)
Biomarcadores de Tumor/genética , Islas de CpG/genética , Metilación de ADN , Perfilación de la Expresión Génica , Genoma Humano , Síndromes Mielodisplásicos/genética , Estudios de Casos y Controles , ARN Helicasas DEAD-box/genética , ADN de Neoplasias/genética , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Pronóstico , Proteína Proto-Oncogénica c-ets-1/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptores de Interleucina/genética , Ribonucleasa III/genética , Factores de Riesgo , Células Tumorales Cultivadas
3.
Biochemistry ; 40(42): 12712-8, 2001 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-11601996

RESUMEN

The fosfomycin resistance protein, FosA, catalyzes the Mn(2+)-dependent addition of glutathione to the antibiotic fosfomycin, (1R,2S)-epoxypropylphosphonic acid, rendering the antibiotic inactive. The enzyme is a homodimer of 16 kDa subunits, each of which contains a single mononuclear metal site. Stopped-flow absorbance/fluorescence spectrometry provides evidence suggesting a complex kinetic mechanism for the acquisition of Mn(2+) by apoFosA. The binding of Mn(H(2)O)(6)(2+) to apoFosA alters the UV absorption and intrinsic fluorescence characteristics of the protein sufficiently to provide sensitive spectroscopic probes of metal binding. The acquisition of metal is shown to be a multistep process involving rapid preequilibrium formation of an initial complex with release of approximately two protons (k(obsd) > or = 800 s(-1)). The initial complex either rapidly dissociates or forms an intermediate coordination complex (k > 300 s(-1)) with rapid isomerization (k > or = 20 s(-1)) to a set of tight protein-metal complexes. The observed bimolecular rate constant for formation of the intermediate coordination complex is 3 x 10(5) M(-1) s(-1). The release of Mn(2+) from the protein is slow (k approximately 10(-2) s(-1)). The kinetic results suggest a more complex chelate effect than is typically observed for metal binding to simple multidentate ligands. Although the addition of the substrate, fosfomycin, has no appreciable effect on the association kinetics of enzyme and metal, it significantly decreases the dissociation rate, suggesting that the substrate interacts directly with the metal center.


Asunto(s)
Proteínas Bacterianas , Fosfomicina/metabolismo , Glutatión Transferasa/metabolismo , Manganeso/metabolismo , Metaloproteínas/metabolismo , Sitios de Unión/efectos de los fármacos , Quelantes/metabolismo , Farmacorresistencia Microbiana , Ácido Edético/metabolismo , Fosfomicina/farmacología , Cinética , Sustancias Macromoleculares , Protones , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta
4.
Chem Res Toxicol ; 14(8): 1107-17, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11511185

RESUMEN

Glutathione (GSH) transferases are generally involved in the detoxication of xenobiotic chemicals. However, conjugation can also activate compounds and result in DNA modification. Activation of 1,2-dihaloethanes (BrCH(2)CH(2)Br, BrCH(2)CH(2)Cl, and ClCH(2)CH(2)Cl) was investigated using two mammalian theta class GSH transferases (rat GST 5-5 and human GST T1) and a bacterial dichloromethane dehalogenase (DM11). Although the literature suggests that the bacterial dehalogenase does not catalyze reactions with CH(3)Cl, ClCH(2)CH(2)Cl, or CH(3)CHCl(2), we found a higher enzyme efficiency for DM11 than for the mammalian GSH transferases in conjugating CH(3)Cl, CH(3)CH(2)Cl, and CH(3)CH(2)Br. Enzymatic rates of activation of 1,2-dihaloethanes were determined in vitro by measuring S,S-ethylene-bis-GSH, the major product trapped by nonenzymatic reaction with the substrate GSH. Salmonella typhimurium TA 1535 systems expressing each of these GSH transferases were used to determine mutagenicity. Rates of formation of S,S-ethylene-bis-GSH by the GSH transferases correlated with the mutagenicity determined in the reversion assays for the three 1,2-dihaloethanes, consistent with the view that half-mustards are the mutagenic products of the GSH transferase reactions. Half-mustards [S-(2-haloethyl)GSH] containing either F, Cl, or Br (as the leaving group) were tested for their abilities to induce revertants in S. typhimurium, and rates of hydrolysis were also determined. GSH transferases do not appear to be involved in the breakdown of the half-mustard intermediates. A halide order (Br > Cl) was observed for both GSH transferase-catalyzed mutagenicity and S,S-ethylene-bis-GSH formation from 1,2-dihaloethanes, with the single exception (both assays) of BrCH(2)CH(2)Cl reaction with DM11, which was unexpectedly high. The lack of substrate saturation seen for conjugation of dihalomethanes with GSTs 5-5 and T1 was also observed with the mono- and 1,2-dihaloethanes [Wheeler, J. B., Stourman, N. V., Thier, R., Dommermuth, A., Vuilleumier, S., Rose, J. A., Armstrong, R. N., and Guengerich, F. P. (2001) Chem. Res. Toxicol. 14, 1118-1127], indicative of an inherent difference in the catalytic mechanisms of the bacterial and mammalian GSH transferases.


Asunto(s)
Alcanos/metabolismo , Etano/análogos & derivados , Glutatión Transferasa/metabolismo , Animales , Bacterias , Catálisis , Etano/química , Hidrocarburos Halogenados , Hidrólisis , Cinética , Mamíferos , Compuestos de Mostaza/química , Pruebas de Mutagenicidad , Salmonella typhimurium/genética
5.
Chem Res Toxicol ; 14(8): 1118-27, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11511186

RESUMEN

A primary route of metabolism of dihalomethanes occurs via glutathione (GSH) transferase-catalyzed conjugation. Mammalian theta class GSH transferases and a group of bacterial dichloromethane dehalogenases are able to catalyze the hydrolytic dehalogenation of dihalomethanes via GSH conjugation and subsequent formation of HCHO. Dihalomethanes have been shown to induce revertants in Salmonella typhimurium TA 1535 expressing theta class GSH transferases. Two mammalian theta class GSH transferases (rat GST 5-5 and human GST T1) and the bacterial dehalogenase DM11 were compared in the in vitro conjugation of CH(3)Cl and using in vitro assays (HCHO formation) and the S. typhimurium mutagenesis assay with the dihalomethanes CH(2)Cl(2), CH(2)Br(2), CH(2)BrCl, CH(2)ICl, CH(2)I(2), and CH(2)ClF. GSTs 5-5 and T1 had similar characteristics and exhibited first-order rather than Michaelis-Menten kinetics for HCHO formation over the range of dihalomethane concentrations tested. In contrast, the DM11 enzyme displayed typical hyperbolic Michaelis-Menten kinetics for all of the compounds tested. A similar pattern was observed for the conjugation of CH(3)Cl. The reversion tests with S. typhimurium expressing DM11 or GST 5-5 showed a concentration-dependent increase in revertants for most of the dihalomethanes, and DM11 produced revertants at dihalomethane concentrations lower than GST 5-5. Collectively, the results indicate that rates of conversion of dihalomethanes to HCHO are not correlated with mutagenicity and that GSH conjugates are genotoxic. The results are compared with the conjugation and genotoxicity of haloethanes in the preceding paper in this issue [Wheeler, J. B., Stourman, N. V., Armstrong, R. N., and Guengerich, F. P. (2001) Chem. Res. Toxicol. 14, 1107-1117]. The halide order appears most important in the dihalomethane conjugation reactions catalyzed by GST 5-5 and less so in GST T1 and DM11, probably due to changes in the rate-limiting steps.


Asunto(s)
Glutatión Transferasa/metabolismo , Hidrocarburos Halogenados/química , Metano/análogos & derivados , Animales , Bacterias , Catálisis , Relación Dosis-Respuesta a Droga , Hidrolasas/metabolismo , Hidrólisis , Cinética , Mamíferos , Metano/química , Pruebas de Mutagenicidad , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética
6.
Biochemistry ; 40(11): 3378-84, 2001 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-11258959

RESUMEN

An important aspect of the catalytic mechanism of microsomal glutathione transferase (MGST1) is the activation of the thiol of bound glutathione (GSH). GSH binding to MGST1 as measured by thiolate anion formation, proton release, and Meisenheimer complex formation is a slow process that can be described by a rapid binding step (K(GSH)d = 47 +/- 7 mM) of the peptide followed by slow deprotonation (k2 = 0.42 +/- 0.03 s(-1). Release of the GSH thiolate anion is very slow (apparent first-order rate k(-2) = 0.0006 +/- 0.00002 s(-)(1)) and thus explains the overall tight binding of GSH. It has been known for some time that the turnover (kcat) of MGST1 does not correlate well with the chemical reactivity of the electrophilic substrate. The steady-state kinetic parameters determined for GSH and 1-chloro-2,4-dinitrobenzene (CDNB) are consistent with thiolate anion formation (k2) being largely rate-determining in enzyme turnover (kcat = 0.26 +/- 0.07 s(-1). Thus, the chemical step of thiolate addition is not rate-limiting and can be studied as a burst of product formation on reaction of halo-nitroarene electrophiles with the E.GS- complex. The saturation behavior of the concentration dependence of the product burst with CDNB indicates that the reaction occurs in a two-step process that is characterized by rapid equilibrium binding ( = 0.53 +/- 0.08 mM) to the E.GS- complex and a relatively fast chemical reaction with the thiolate (k3 = 500 +/- 40 s(-1). In a series of substrate analogues, it is observed that log k3 is linearly related (rho value 3.5 +/- 0.3) to second substrate reactivity as described by Hammett sigma- values demonstrating a strong dependence on chemical reactivity that is similar to the nonenzymatic reaction (rho = 3.4). Microsomal glutathione transferase 1 displays the unusual property of being activated by sulfhydryl reagents. When the enzyme is activated by N-ethylmaleimide, the rate of thiolate anion formation is greatly enhanced, demonstrating for the first time the specific step that is activated. This result explains earlier observations that the enzyme is activated only with more reactive substrates. Taken together, the observations show that the kinetic mechanism of MGST1 can be described by slow GSH binding/thiolate formation followed by a chemical step that depends on the reactivity of the electrophilic substrate. As the chemical reactivity of the electrophile becomes lower the rate-determining step shifts from thiolate formation to the chemical reaction.


Asunto(s)
Glutatión Transferasa/metabolismo , Glutatión/metabolismo , Microsomas Hepáticos/enzimología , Animales , Aniones/química , Aniones/metabolismo , Sitios de Unión , Catálisis , Dinitroclorobenceno/química , Activación Enzimática , Etilmaleimida/química , Glutatión/química , Glutatión Transferasa/química , Cinética , Ratas , Especificidad por Sustrato
7.
J Bacteriol ; 183(7): 2380-3, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11244082

RESUMEN

We demonstrate that the Bacillus subtilis fosB(yndN) gene encodes a fosfomycin resistance protein. Expression of fosB requires sigma(W), and both fosB and sigW mutants are fosfomycin sensitive. FosB is a metallothiol transferase related to the FosA class of Mn(2+)-dependent glutathione transferases but with a preference for Mg(2+) and L-cysteine as cofactors.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/fisiología , Cisteína/farmacología , Fosfomicina/farmacología , Factor sigma/fisiología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Farmacorresistencia Microbiana , Escherichia coli/efectos de los fármacos , Glutatión Transferasa/fisiología , Magnesio/farmacología , Manganeso/farmacología , Datos de Secuencia Molecular , Transcripción Genética
8.
Biochemistry ; 39(45): 13625-32, 2000 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-11076500

RESUMEN

It is now appreciated that the relationships of proteins, particularly enzymes, within a protein superfamily can be understood not only in terms of their sequence similarities and three-dimensional structures but also by chemical threads that relate their functional attributes. The mechanistic ties among superfamily members can often be traced to a common transition state for the rate-limiting step of the reactions being catalyzed. This paper presents an analysis of a metalloenzyme superfamily, the members of which catalyze a very diverse set of reactions with unrelated transition states but a more general common mechanistic imperative. The vicinal oxygen chelate (VOC) superfamily is composed of structurally related proteins with paired beta alpha beta beta beta motifs that provide a metal coordination environment with two or three open or readily accessible coordination sites to promote direct electrophilic participation of the metal ion in catalysis. The known types of reactions that are catalyzed include isomerizations (glyoxalase I), epimerizations (methylmalonyl-CoA epimerase), oxidative cleavage of C-C bonds (extradiol dioxygenase), and nucleophilic substitutions (fosfomycin resistance proteins). The remarkable access to mechanism space that is provided by the VOC superfamily appears to derive from a simple, pseudosymmetric structural fold that maximizes the catalytic versatility of the metal center.


Asunto(s)
Acetiltransferasas/química , Dioxigenasas , Glutatión Transferasa/química , Lactoilglutatión Liasa/química , Metaloproteínas/química , Oxigenasas/química , Racemasas y Epimerasas/química , Homología de Secuencia de Aminoácido , Acetaldehído/química , Proteínas Bacterianas/química , Bleomicina/química , Quelantes/química , Farmacorresistencia Microbiana , Fosfomicina/química , Humanos , Oxígeno/química
9.
Biochemistry ; 39(40): 12336-44, 2000 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-11015213

RESUMEN

The conformational stabilities of two homodimeric class mu glutathione transferases (GSTM1-1 and GSTM2-2) were studied by urea- and guanidinium chloride-induced denaturation. Unfolding is reversible and structural changes were followed with far-ultraviolet circular dichroism, tryptophan fluorescence, enzyme activity, chemical cross-linking, and size-exclusion chromatography. Disruption of secondary structure occurs as a monophasic transition and is independent of protein concentration. Changes in tertiary structure occur as two transitions; the first is protein concentration dependent, while the second is weakly dependent (GSTM1-1) or independent (GSTM2-2). The second transition corresponds with the secondary structure transition. Loss in catalytic activity occurs as two transitions for GSTM1-1 and as one transition for GSTM2-2. These transitions are dependent upon protein concentration. The first deactivation transition coincides with the first tertiary structure transition. Dimer dissociation occurs prior to disruption of secondary structure. The data suggest that the equilibrium unfolding/refolding of the class mu glutathione transferases M1-1 and M2-2 proceed via a three-state process: N(2) <--> 2I <--> 2U. Although GSTM1-1 and GSTM2-2 are homologous (78% identity/94% homology), their N(2) tertiary structures are not identical. Dissociation of the GSTM1-1 dimer to structured monomers (I) occurs at lower denaturant concentrations than for GSTM2-2. The monomeric intermediate for GSTM1-1 is, however, more stable than the intermediate for GSTM2-2. The intermediates are catalytically inactive and display nativelike secondary structure. Guanidinium chloride-induced denaturation yields monomeric intermediates, which have a more loosely packed tertiary structure displaying enhanced solvent exposure of its tryptophans and enhanced ANS binding. The three-state model for the class mu enzymes is in contrast to the equilibrium two-state models previously proposed for representatives of classes alpha/pi/Sj26 GSTs. Class mu subunits appear to be intrinsically more stable than those of the other GST classes.


Asunto(s)
Glutatión Transferasa/química , Glutatión Transferasa/metabolismo , Pliegue de Proteína , Naftalenosulfonatos de Anilina/metabolismo , Animales , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Reactivos de Enlaces Cruzados/química , Dimerización , Estabilidad de Enzimas , Glutaral/química , Guanidina , Calor , Isoenzimas/química , Isoenzimas/metabolismo , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Ratas , Relación Estructura-Actividad , Urea
10.
Biochem J ; 347 Pt 1: 193-7, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10727418

RESUMEN

We have shown previously that the solvent-induced equilibrium unfolding mechanism of class Sigma glutathione S-transferase (GST) is strongly affected by ionic strength [Stevens, Hornby, Armstrong and Dirr (1998) Biochemistry 37, 15534-15541]. The protein is dimeric and has a hydrophilic subunit interface. Here we show that ionic strength alone has significant effects on the conformation of the protein, in particular at the active site. With the use of NaCl at up to 2 M under equilibrium conditions, the protein lost 60% of its catalytic activity and the single tryptophan residue per subunit became partly exposed. The effect was independent of protein concentration, eliminating the dissociation of the dimer as a possibility for the conformational changes. This was confirmed by size-exclusion HPLC. There was no significant change in the secondary structure of the protein according to far-UV CD data. Manual-mixing and stopped-flow kinetics experiments showed a slow single-exponential salt-induced change in protein fluorescence. For equilibrium and kinetics experiments, the addition of an active-site ligand (S-hexylglutathione) completely protected the protein from the ionic-strength-induced conformational changes. This suggests that the change occurs at or near the active site. Possible structural reasons for these novel effects are proposed, such as the flexibility of the alpha-helix 2 region as well as the hydrophilic subunit interface, highlighting the importance of electrostatic interactions in maintaining the structure of the active site of this GST.


Asunto(s)
Glutatión Transferasa/química , Triptófano , Sitios de Unión , Dimerización , Glutatión Transferasa/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Sustancias Macromoleculares , Modelos Moleculares , Concentración Osmolar , Conformación Proteica/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Cloruro de Sodio/farmacología , Electricidad Estática
11.
Drug Metab Rev ; 32(3-4): 327-38, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11139132

RESUMEN

New structural and mechanistic data on epoxide hydrolases provide additional insight into the details of how these enzymes function. The data reveal that, in addition to the catalytic triad located in the core domain that carries out the hydrolytic reaction, there are two tyrosine residues, located in the cap domain, which assist in the initial alkylation event.


Asunto(s)
Epóxido Hidrolasas/química , Epóxido Hidrolasas/metabolismo , Unión Competitiva , Catálisis , Compuestos Epoxi/metabolismo , Predicción , Humanos , Hidrólisis , Cinética , Modelos Moleculares , Conformación Proteica
12.
Biochemistry ; 38(23): 7462-9, 1999 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-10360943

RESUMEN

The fosfomycin resistance protein FosA is a member of a distinct superfamily of metalloenzymes containing glyoxalase I, extradiol dioxygenases, and methylmalonyl-CoA epimerase. The dimeric enzyme, with the aid of a single mononuclear Mn2+ site in each subunit, catalyzes the addition of glutathione (GSH) to the oxirane ring of the antibiotic, rendering it inactive. Sequence alignments suggest that the metal binding site of FosA is composed of three residues: H7, H67, and E113. The single mutants H7A, H67A, and E113A as well as the more conservative mutants H7Q, H67Q, and E113Q exhibit marked decreases in the ability to bind Mn2+ and, in most instances, decreases in catalytic efficiency and the ability to confer resistance to the antibiotic. The enzyme also requires the monovalent cation K+ for optimal activity. The K+ ion activates the enzyme 100-fold with an activation constant of 6 mM, well below the physiologic concentration of K+ in E. coli. K+ can be replaced by other monovalent cations of similar ionic radii. Several lines of evidence suggest that the K+ ion interacts directly with the active site. Interaction of the enzyme with K+ is found to be dependent on the presence of the substrate fosfomycin. Moreover, the E113Q mutant exhibits a kcat which is 40% that of wild-type in the absence of K+. This mutant is not activated by monovalent cations. The behavior of the E113Q mutant is consistent with the proposition that the K+ ion helps balance the charge at the metal center, further lowering the activation barrier for addition of the anionic nucleophile. The fully activated, native enzyme provides a rate acceleration of >10(15) with respect to the spontaneous addition of GSH to the oxirane.


Asunto(s)
Proteínas Bacterianas , Fosfomicina/química , Glutatión Transferasa/química , Glutatión Transferasa/metabolismo , Metaloproteínas/química , Metaloproteínas/metabolismo , Sitios de Unión/genética , Catálisis , Cationes Bivalentes/química , Cationes Monovalentes/química , Farmacorresistencia Microbiana , Espectroscopía de Resonancia por Spin del Electrón , Activación Enzimática/genética , Escherichia coli/enzimología , Escherichia coli/genética , Fosfomicina/metabolismo , Glutatión Transferasa/genética , Cinética , Ligandos , Metaloproteínas/genética , Mutagénesis Sitio-Dirigida , Plásmidos/síntesis química , Plásmidos/genética , Potasio/química , Alineación de Secuencia , Especificidad por Sustrato/genética
13.
14.
Curr Opin Chem Biol ; 2(5): 618-23, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9818188

RESUMEN

Several significant advances in the understanding of the catalytic mechanisms, structures and evolution of glutathione transferases have occurred in the past year. These advances include new mechanistic information concerning the canonical soluble enzymes, the finding that the fosfomycin-specific enzyme, FosA, is a metalloglutathione transferase and a higher resolution projection structure of the microsomal enzyme.


Asunto(s)
Proteínas Bacterianas , Evolución Biológica , Catálisis , Glutatión Transferasa/química , Glutatión/metabolismo , Estimulantes del Sistema Nervioso Central/química , Farmacorresistencia Microbiana/genética , Fosfomicina/química , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Hidroquinonas/química , Metaloproteínas/química , Oxidación-Reducción , Tirosina/análogos & derivados , Tirosina/química
15.
Biochemistry ; 37(44): 15534-41, 1998 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-9799517

RESUMEN

Solvent-induced equilibrium unfolding of a homodimeric class sigma glutathione transferase (GSTS1-1, EC 2.5.1.18) was characterized by tryptophan fluorescence, anisotropy, enzyme activity, 8-anilino-1-naphthalenesulfonate (ANS) binding, and circular dichroism. Urea induces a triphasic unfolding transition with evidence for two well-populated thermodynamically stable intermediate states of GSTS1-1. The first unfolding transition is protein concentration independent and involves a change in the subunit tertiary structure yielding a partially active dimeric intermediate (i.e., N2 left and right arrow I2). This is followed by a protein concentration dependent step in which I2 dissociates into compact inactive monomers (M) displaying enhanced hydrophobicity. The third unfolding transition, which is protein concentration independent, involves the complete unfolding of the monomeric state. Increasing NaCl concentrations destabilize N2 and appear to shift the equilibrium toward I2 whereas the stability of the monomeric intermediate M is enhanced. The binding of substrate or product analogue (i.e., glutathione or S-hexylglutathione) to the protein's active site stabilizes the native dimeric state (N2), causing the first two unfolding transitions to shift toward higher urea concentrations. The stability of M was not affected. The data implicate a region at/near the active site in domain I (most likely alpha-helix 2) as being highly unstable/flexible which undergoes local unfolding, resulting initially in I2 formation followed by a disruption in quaternary structure to a monomeric intermediate. The unfolding/refolding pathway is compared with those observed for other cytosolic GSTs and discussed in light of the different structural features at the subunit interfaces, as well as the evolutionary selection of this GST as a lens crystallin.


Asunto(s)
Glutatión Transferasa/química , Conformación Proteica , Pliegue de Proteína , Animales , Cromatografía Líquida de Alta Presión , Decapodiformes , Dimerización , Estabilidad de Enzimas/efectos de los fármacos , Colorantes Fluorescentes , Glutatión/análogos & derivados , Glutatión/farmacología , Glutatión Transferasa/metabolismo , Calor , Familia de Multigenes , Conformación Proteica/efectos de los fármacos , Desnaturalización Proteica , Cloruro de Sodio , Espectrometría de Fluorescencia , Urea
16.
J Mol Biol ; 281(2): 323-39, 1998 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-9698551

RESUMEN

The structure of the tetradeca-(3-fluorotyrosyl) M1-1 GSH transferase (3-FTyr GSH transferase), a protein in which tyrosine residues are globally substituted by 3-fluorotyrosines has been determined at 2.2 A resolution. This variant was produced to study the effect on the enzymatic mechanism and the structure was undertaken to assess how the presence of the 3-fluorotyrosyl residue influences the protein conformation and hence its function. Although fluorinated amino acid residues have frequently been used in biochemical and NMR investigations of proteins, no structure of a protein that has been globally substituted with a fluorinated amino acid has previously been reported. Thus, this structure represents the first crystal structure of such a protein containing a library of 14 (28 crystallographically distinct) microenvironments from which the nature of the interactions of fluorine atoms with the rest of the protein can be evaluated. Numerous conformational changes are observed in the protein structure as a result of substitution of 3-fluorotyrosine for tyrosine. The results of the comparison of the crystal structure of the fluorinated protein with the native enzyme reveal that conformational changes are observed for most of the 3-fluorotyrosines. The largest differences are seen for residues where the fluorine, the OH, or both are directly involved in interactions with other regions of the protein or with a symmetry-related molecule. The fluorine atoms of the 3-fluorotyrosine interact primarily through hydrogen bonds with other residues and water molecules. In several cases, the conformation of a 3-fluorotyrosine is different in one of the monomers of the enzyme from that observed in the other, including different hydrogen-bonding patterns. Altered conformations can be related to differences in the crystal packing interactions of the two monomers in the asymmetric unit. The fluorine atom on the active-site Tyr6 is located near the S atom of the thioether product (9R,10R)-9-(S-glutathionyl)-10-hydroxy-9,10-dihydrophenanthrene and creates a different pattern of interactions between 3-fluorotyrosine 6 and the S atom. Studies of these interactions help explain why 3-FTyr GSH transferase exhibits spectral and kinetic properties distinct from the native GSH transferase.


Asunto(s)
Cristalografía por Rayos X/métodos , Glutatión Transferasa/química , Conformación Proteica , Tirosina/análogos & derivados , Animales , Sitios de Unión , Dimerización , Enlace de Hidrógeno , Modelos Moleculares , Ratas , Tirosina/química , Agua/química , Xenobióticos/química
17.
Chem Biol Interact ; 111-112: 41-50, 1998 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-9679542

RESUMEN

A number of glutathione (GSH) transferases are now known in prokaryotes and eukaryotes. The enzymes appear to be primarily involved in the metabolism of foreign compounds. At least six distinct classes of soluble GSH transferases have been identified in eukaryotes and named alpha, mu, pi, sigma, theta and kappa. Sequences and the known three-dimensional structures of the soluble enzymes suggest that these proteins share a common ancestry, though the precise details of their evolution remain obscure. A second distinct family of GSH transferases are the microsomal or membrane-bound enzymes that include leukotriene C4 synthase. A third family is represented by a bacterial GSH transferase (FosA) responsible for conferring resistance to the antibiotic fosfomycin, reported some years ago by Suarez and co-workers (Arca et al., Antimicrob. Agents Chemother. 34 (1990) 1552-1556). The enzyme is quite specific for fosfomycin, which contains a very stable epoxide moiety. Evidence is presented that FosA is a metalloprotein related to iron- and manganese-dependent dioxygenases and to glyoxalase I. These enzymes are members of a previously unrecognized group of enzymes; the vicinal oxygen chelate superfamily. The mechanistic imperative driving the evolution of FosA and its relatives, which are enzymes catalyzing quite diverse chemical reactions, is proposed to be the electrophilic assistance provided by the metal through chelation of a substrate or intermediate.


Asunto(s)
Proteínas Bacterianas , Evolución Molecular , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Animales , Sitios de Unión , Cationes Bivalentes/farmacología , Quelantes/metabolismo , Activación Enzimática/efectos de los fármacos , Glutatión Transferasa/clasificación , Humanos , Metaloproteínas/clasificación , Metaloproteínas/genética , Metaloproteínas/metabolismo , Metales/metabolismo , Oxígeno/metabolismo
18.
Biochemistry ; 37(18): 6286-94, 1998 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-9572843

RESUMEN

The catalytic characteristics and structure of the M1-1 isoenzyme of rat glutathione (GSH) transferase in which all four tryptophan residues in each monomer are replaced with 5-fluorotryptophan are described. The fluorine-for-hydrogen substitution does not change the interaction of the enzyme with GSH even though two tryptophan residues (Trp7 and Trp45) are involved in direct hydrogen-bonding interactions with the substrate. The rate constants for association and dissociation of the peptide, measured by stopped-flow spectrometry, remain unchanged by the unnatural amino acid. The 5-FTrp-substituted enzyme exhibits a kcat of 73 s-1 as compared to 18 s-1 for the native enzyme toward 1-chloro-2,4-dinitrobenzene. That the increase in the turnover number is due to an enhanced rate of product release in the mutant is confirmed by the kinetics of the approach to equilibrium for binding of the product. The crystal structure of the 5-FTrp-containing enzyme was solved at a resolution of 2.0 A by difference Fourier techniques. The structure reveals local conformational changes in the structural elements that define the approach to the active site which are attributed to steric interactions of the fluorine atoms associated with 5-FTrp146 and 5-FTrp214 in domain II. These changes appear to result in the enhanced rate of product release. This structure represents the first of a protein substituted with 5-fluorotryptophan.


Asunto(s)
Glutatión Transferasa/metabolismo , Isoenzimas/metabolismo , Triptófano/análogos & derivados , Animales , Catálisis , Cristalografía por Rayos X , Glutatión/metabolismo , Glutatión Transferasa/química , Enlace de Hidrógeno , Isoenzimas/química , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Triptófano/química , Triptófano/metabolismo
19.
Biochemistry ; 37(9): 2897-904, 1998 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-9485441

RESUMEN

Microsomal epoxide hydrolase (MEH) catalyzes the addition of water to epoxides in a two-step reaction involving initial attack of an active site carboxylate on the oxirane to give an ester intermediate followed by hydrolysis of the ester. An efficient bacterial expression system for the enzyme from rat that facilitates the production of native and mutant enzymes for mechanistic analysis is described. Pre-steady-state kinetics of the native enzyme toward glycidyl-4-nitrobenzoates, 1, indicate the rate-limiting step in the reaction is hydrolysis of the alkyl-enzyme intermediate. The enzyme is enantioselective, turning over (2R)-1 about 10-fold more efficiently than (2S)-1, and regiospecific toward both substrates with exclusive attack at the least hindered oxirane carbon. Facile isomerization of the monoglyceride product is observed and complicates the regiochemical analysis. The D226E and D226N mutants of the protein are catalytically inactive, behavior that is consistent with the role of D226 as the active-site nucleophile as suggested by sequence alignments with other alpha/beta-hydrolase fold enzymes. The D226N mutant undergoes hydrolytic autoactivation with a half-life of 9.3 days at 37 degreesC, suggesting that the mutant is still capable of catalyzing the hydrolytic half-reaction (in this instance an amidase reaction) and confirming that D226 is in the active site. The indoylyl side chain of W227, which is in or near the active site, is not required for efficient alkylation of the enzyme or for hydrolysis of the intermediate. However, the W227F mutant does exhibit altered stereoselectivity toward (2R)-1, (2S)-1, and phenanthrene-9,10-oxide, suggesting that modifications at this position might be used to manipulate the stereo- and regioselectivity of the enzyme.


Asunto(s)
Epóxido Hidrolasas/metabolismo , Alquilación , Animales , Catálisis , Epóxido Hidrolasas/genética , Escherichia coli , Cinética , Espectroscopía de Resonancia Magnética , Modelos Químicos , Mutagénesis Sitio-Dirigida , Ratas , Proteínas Recombinantes/metabolismo , Estereoisomerismo
20.
Biochemistry ; 37(9): 2905-11, 1998 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-9485442

RESUMEN

Microsomal epoxide hydrolase (MEH) is a member of the alpha/beta-hydrolase fold family of enzymes, each of which has a catalytic triad consisting of a nucleophile involved in the formation of a covalent intermediate and a general base and charge relay carboxylate that catalyze the hydrolysis of the intermediate. The rate-limiting step in the catalytic mechanism of MEH is hydrolysis of the ester intermediate. An efficient bacterial expression system for a C-terminal hexahistidine tagged version of the native enzyme, which facilitates the isolation of mutant enzymes in which residues involved in the hydrolytic half-reaction have been altered, is described. The H431S mutant of this enzyme is efficiently alkylated by substrate to form the ester intermediate but is unable to hydrolyze the ester to complete the catalytic cycle, a fact that confirms that H431 acts as the base in the hydrolytic half-reaction. The charge relay carboxylate, which is not apparent in paired sequence alignments with other alpha/beta-hydrolase fold enzymes, is thought to be located between residues 340 and 405. A mutagenic survey of all eight Asp and Glu residues in this region reveals that only two (E376 and E404) influence the catalytic mechanism. Steady-state and pre-steady-state kinetic analyses of these residues suggest that both E404 and E376 may serve the charge relay function in the hydrolysis half-reaction. Finally, the tryptophan residue (W150), which resides in the oxyanion hole sequence HGWP, is demonstrated to contribute to the large change in intrinsic protein fluorescence observed when the enzyme is alkylated.


Asunto(s)
Epóxido Hidrolasas/metabolismo , Animales , Catálisis , Epóxido Hidrolasas/genética , Escherichia coli , Hidrólisis , Cinética , Modelos Químicos , Mutagénesis Sitio-Dirigida , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...