Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Pept Res ; 65(6): 529-37, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15885112

RESUMEN

Protein farnesyltransferase (PFTase) catalyzes the attachment of a geranylazide (C10) or farnesylazide (C15) moiety from the corresponding prenyldiphosphates to a model peptide substrate, N-dansyl-Gly-Cys-Val-Ile-Ala-OH. The rates of incorporation for these two substrate analogs are comparable and approximately twofold lower than that using the natural substrate farnesyl diphosphate (FPP). Reaction of N-dansyl-Gly-Cys(S-farnesylazide)-Val-Ile-Ala-OH with 2-diphenylphosphanylbenzoic acid methyl ester then gives a stable alkoxy-imidate linked product. This result suggests future generations whereby azide groups introduced using this enzymatic approach are functionalized using a broad range of azide-reactive reagents. Thus, chemistry has been developed that could be used to achieve highly specific peptide and protein modification. The farnesylazide analog may be useful in certain biological studies, whereas the geranylazide group may be more useful for general protein modification and immobilization.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Azidas/química , Péptidos/síntesis química , Difosfatos/síntesis química
2.
Curr Opin Chem Biol ; 5(6): 696-704, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11738181

RESUMEN

Covalent modification methods allow an almost unlimited range of functionality to be introduced into proteins. In concert with genetic techniques, chemical strategies have had significant impact in the field of enzyme design. Major recent developments include introducing catalytic activity into inactive proteins, modifying the selectivity and/or reactivity of existing enzymes and designing novel enzyme-based biosensors. New chemical methods promise to further increase the range of functionality that can be incorporated into proteins. These results suggest that semi-synthetic methods will play a key role in the development of future biocatalysts.


Asunto(s)
Enzimas/síntesis química , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos , Técnicas Biosensibles , Diseño de Fármacos , Enzimas/química , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Especificidad por Sustrato
4.
Bioorg Med Chem ; 9(9): 2461-6, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11553487

RESUMEN

Artificial enzymes can be created by covalent attachment of a catalytic active group to a protein scaffold. Recently, we assembled an artificial transaminase by conjugation of intestinal fatty acid binding protein (IFABP) with a pyridoxamine derivative via a disulfide bond; the resulting construct catalyzed a transamination reaction 200-fold faster than free pyridoxamine. To identify the origin of this increased catalytic efficiency computer modeling was first used to identify two putative residues, Y14 and R126, that were in close proximity to the gamma-carboxylate group of the substrate, alpha-ketoglutartate. These positions were mutated to phenylalanine and methionine, respectively, and used to prepare semisynthetic transaminases by conjugation to pyridoxamine (Px) or an N-methylated derivative (MPx). Kinetic analysis of the resulting constructs showed that the R126M mutation reduced substrate affinity 3- to 6-fold while the additional Y14F mutation had a negligible effect. These results are consistent with a model for substrate recognition that involves an electrostatic interaction between the cationic guanidinium group of R126 and the anionic carboxylate from the substrate. Interestingly, one of the conjugates that contains an N-methylated pyridoxamine catalyzes a transamination reaction with a k(cat)' value of 1.1h(-1) which is the fastest value for k(cat) we have thus far obtained and is 34-fold greater than that for the free cofactor in the absence of the protein.


Asunto(s)
Proteínas Portadoras/química , Imitación Molecular , Proteínas de Neoplasias , Piridoxamina/química , Transaminasas/síntesis química , Proteínas Supresoras de Tumor , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Simulación por Computador , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Humanos , Intestinos/química , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Especificidad por Sustrato , Transaminasas/química
5.
J Am Chem Soc ; 123(19): 4373-81, 2001 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-11457220

RESUMEN

Protein prenylation, involving the alkylation of a specific C-terminal cysteine with a C(15) or C(20) isoprenoid unit, is an essential posttranslational modification required by most GTP-binding proteins for normal biological activity. Despite the ubiquitous nature of this modification and numerous efforts aimed at inhibiting prenylating enzymes for therapeutic purposes, the function of prenylation remains unclear. To explore the role the isoprenoid plays in mediating protein-protein recognition, we have synthesized a photoactivatable, isoprenoid-containing cysteine analogue (2) designed to act as a mimic of the C-terminus of prenylated proteins. Photolysis experiments with 2 and RhoGDI (GDI), a protein which interacts with prenylated Rho proteins, suggest that the GDI is in direct contact with the isoprenoid moiety. These results, obtained using purified GDI as well as Escherichia coli (E. coli) crude extract containing GDI, suggest that this analogue will be an effective and versatile tool for the investigation of putative isoprenoid binding sites in a variety of systems. Incorporation of this analogue into peptides or proteins should allow for even more specific interactions between the photoactivatable isoprenoid and any number of isoprenoid binding proteins.


Asunto(s)
Cisteína/análogos & derivados , Cisteína/síntesis química , Fosfatos de Poliisoprenilo/química , Unión Competitiva , Reactivos de Enlaces Cruzados , Escherichia coli/química , Indicadores y Reactivos , Marcaje Isotópico , Imitación Molecular , Fotoquímica , Fotólisis , Pruebas de Precipitina , Prenilación de Proteína , Espectrofotometría Ultravioleta , Radioisótopos de Azufre , Proteína de Unión al GTP cdc42/química
6.
J Org Chem ; 66(10): 3253-64, 2001 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-11348105

RESUMEN

Protein prenylation is a posttranslational lipid modification in which C(15) and C(20) isoprenoid units are linked to specific protein-derived cysteine residues through a thioether linkage. This process is catalyzed by a class of enzymes called prenyltransferases that are being intensively studied due to the finding that Ras protein is farnesylated coupled with the observation that mutant forms of Ras are implicated in a variety of human cancers. Inhibition of this posttranslational modification may serve as a possible cancer chemotherapy. Here, the syntheses of two new farnesyl diphosphate (FPP) analogues containing photoactive benzophenone groups are described. Each of these compounds was prepared in six steps from dimethylallyl alcohol. Substrate studies, inhibition kinetics, photoinactivation studies, and photolabeling experiments are also included; these experiments were performed with a number of protein prenyltransferases from different sources. A X-ray crystal structure of one of these analogues bound to rat farnesyltransferase illustrates that they are good substrate mimics. Of particular importance, these new analogues can be enzymatically incorporated into Ras-based peptide substrates allowing the preparation of molecules with photoactive isoprenoids that may serve as valuable probes for the study of prenylation function. Photoaffinity labeling of human protein geranylgeranyltransferase with (32)P-labeled forms of these analogues suggests that the C-10 locus of bound geranylgeranyl diphosphate (GGPP) is in close proximity to residues from the beta-subunit of this enzyme. These results clearly demonstrate the utility of these compounds as photoaffinity labeling analogues for the study of a variety of protein prenyltransferases and other enzymes that employ FPP or GGPP as their substrates.


Asunto(s)
Dimetilaliltranstransferasa/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/síntesis química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Benzofenonas , Cristalografía por Rayos X , Dimetilaliltranstransferasa/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Éteres , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Radioisótopos de Fósforo , Fotoquímica , Fosfatos de Poliisoprenilo/química , Fosfatos de Poliisoprenilo/farmacocinética , Ratas , Sesquiterpenos , Relación Estructura-Actividad , Levaduras
7.
Bioconjug Chem ; 12(3): 385-90, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11353536

RESUMEN

Artificial enzymes can be created by covalent conjugation of a catalytic active group to a protein scaffold. Here, two transamination catalysts were designed via computer modeling and assembled by chemically conjugating a pyridoxamine moiety within the large cavity of intestinal fatty acid binding protein. Each catalyst included a lysine residue, introduced via site-directed mutagenesis, that promotes catalysis by covalent interactions with the pyridoxamine group. Evidence for such interactions include the formation of a Schiff base with the pyridoxal form of the catalyst and a rate versus pH dependence that is bell shaped; both of these features are manifested in natural transaminases. The resulting constructs operate with high enantioselectivity (83-94% ee) and increase the rate of reaction as much as 4200-fold over the rate in the absence of the protein; this is a modest (12-fold) increase in catalytic efficiency (kcat/KM) compared to the conjugate lacking the lysine residue. Most importantly, these artificial aminotransferases are the first examples of designed bioconjugates capable of covalent catalysis, highlighting the potential of this chemogenetic approach.


Asunto(s)
Enzimas/síntesis química , Proteínas de Neoplasias , Proteínas Portadoras/química , Catálisis , Dominio Catalítico , Diseño de Fármacos , Enzimas/metabolismo , Proteínas de Unión a Ácidos Grasos , Concentración de Iones de Hidrógeno , Cinética , Lisina/química , Modelos Moleculares , Piridoxamina/química , Estereoisomerismo , Transaminasas/síntesis química , Transaminasas/metabolismo
8.
Biochemistry ; 40(13): 3920-30, 2001 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-11300771

RESUMEN

Protein geranylgeranyltransferase type I (PGGTase-I) catalyzes the nucleophilic substitution reaction between the C(20) geranylgeranyl diphosphate (GGPP) and a protein-derived thiol to form a thioether linkage. Here, we describe the stereochemical outcome, at the isoprenoid C1, of the reaction catalyzed by human PGGTase-I. To accomplish this, the pentapeptide N-dansyl-GCVLL was first enzymatically prenylated by human PGGTase-I with either (S)-[1-(2)H]farnesyl diphosphate or (S)-[1-(2)H]GGPP. The prenylated products were then degraded to dipeptides using carboxypeptidase Y. After HPLC purification, the prenylated dipeptide products were analyzed by (1)H NMR spectroscopy. The final spectra were compared with the spectra from the same product obtained via chemical synthesis to deduce the stereochemistry of the PGGTase-I-catalyzed reaction. This comparison showed that the reaction proceeds with inversion of configuration with no detectable (< 6%) racemization. These results are more consistent with an associative-type mechanism, but they cannot be used to rule out a dissociative mechanism involving a rigid, solvent-sequestered, tight ion pair.


Asunto(s)
Transferasas Alquil y Aril/química , Prenilación de Proteína , Transferasas Alquil y Aril/metabolismo , Catálisis , Compuestos de Dansilo/síntesis química , Deuterio/química , Dipéptidos/síntesis química , Diterpenos/química , Farnesol/análogos & derivados , Farnesol/química , Humanos , Resonancia Magnética Nuclear Biomolecular , Oligopéptidos/síntesis química , Estereoisomerismo , Especificidad por Sustrato
9.
Bioorg Med Chem Lett ; 10(18): 2091-5, 2000 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-10999478

RESUMEN

An N-methylated, cationic pyridoxamine conjugation reagent was synthesized and tethered via a disulfide bond to a cysteine residue inside the cavity of intestinal fatty acid binding protein. The conjugate was characterized and the kinetic parameters compared to its nonmethylated pyridoxamine analogue. Kinetic isotope effects were used for further mechanistic analysis. Taken together, these experiments suggest that a step distinct from deprotonation of the ketimine in the pyridoxamine to pyridoxal reaction is what limits the rate of the artificial transaminase IFABP-Px. However, the internal energetics of reactions catalyzed by the conjugate containing the N-methylated cofactor appear to be different suggesting that the MPx reagent will be useful in future experiments designed to alter the catalytic properties of semisynthetic transaminases.


Asunto(s)
Proteínas Portadoras/química , Proteínas de Neoplasias , Piridoxamina/química , Transaminasas/química , Proteínas Portadoras/metabolismo , Catálisis , Cationes , Proteínas de Unión a Ácidos Grasos , Cinética , Modelos Moleculares , Imitación Molecular , Piridoxamina/metabolismo , Transaminasas/metabolismo
10.
Bioorg Med Chem Lett ; 9(1): 79-84, 1999 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-9990461

RESUMEN

In an effort to construct catalysts with enzyme-like properties, we are employing a small, cavity-containing protein as a scaffold for the attachment of catalytic groups. In earlier work we demonstrated that a phenanthroline ligand could be introduced into the cavity of the protein ALBP and used to catalyze ester hydrolysis. To examine the effect of positioning the phenanthroline catalyst at different locations within the protein cavity, three new constructs--Phen60, Phen72 and Phen104--were prepared. Each new conjugate was characterized by UV/vis spectroscopy, fluorescence spectroscopy, guanidine hydrochloride denaturation, gel filtration chromatography, and CD spectroscopy to confirm the preparation of the desired construct. Analysis of reactions containing Ala-OiPr showed that Phen60 catalyzed ester hydrolysis with less selectivity than ALBP-Phen while Phen72 promoted this same reaction with higher selectivity. Reactions with Tyr-OMe were catalyzed with higher selectivity by Phen60 and more rapidly by Phen104. These results demonstrate that both the rates and selectivities of hydrolysis reactions catalyzed by these constructs are dependent on the precise site of attachment of the metal ligand within the protein cavity.


Asunto(s)
Proteínas Portadoras/genética , Enzimas/química , Metaloproteínas/química , Metaloproteínas/metabolismo , Metales/metabolismo , Proteína P2 de Mielina/genética , Proteínas de Neoplasias , Proteínas Portadoras/química , Dicroismo Circular , Enzimas/genética , Enzimas/metabolismo , Proteínas de Unión a Ácidos Grasos , Hidrólisis , Metaloproteínas/genética , Metales/química , Mutagénesis Sitio-Dirigida , Proteína P2 de Mielina/química , Fenantrolinas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometría Ultravioleta
11.
Curr Opin Struct Biol ; 8(4): 459-65, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9729737

RESUMEN

The combination of site-directed mutagenesis and chemical modification has resulted in the preparation of protein conjugates with new and useful properties. Proteins modified with metal-chelating groups are proving useful for mapping tertiary and quaternary interactions using the technique of affinity cleavage. The attachment of cofactors, including pyridoxal and pyridoxamine, has resulted in the preparation of semisynthetic transaminases that display enzyme-like properties, including enantioselectivity, substrate specificity and reaction-rate acceleration.


Asunto(s)
Dominio Catalítico , Enzimas/síntesis química , Ingeniería de Proteínas , Proteínas/síntesis química , Catálisis , Coenzimas/síntesis química , Coenzimas/química , Diseño Asistido por Computadora , Enzimas/química , Mutagénesis Sitio-Dirigida , Proteínas/química , Especificidad por Sustrato
12.
Protein Eng ; 11(4): 253-61, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9680187

RESUMEN

Adipocyte lipid-binding protein (ALBP) is a small (14.5 kDa) 10-stranded beta-barrel protein found in mammalian fat cells. The crystal structures of various holo-forms of ALBP have been solved and show the fatty acid ligand bound in a large (approximately 400 A3) cavity isolated from bulk solvent. Examination of the cavity suggests that it would be a good site for the creation of an artificial catalyst, as numerous well defined crystal structures of ALBP are available and past studies have shown the conformation to be reasonably tolerant to modification and mutagenesis. Previous work has shown ALBP to be a good protein scaffold for exploring enantio- and stereoselective reactions; two constructs, ALBP attached to either a pyridoxamine or a phenanthroline group at C117, have been chemically characterized. Both modified proteins have been crystallized and their structures solved and refined. The X-ray models have been used to examine the origin of the chiral selectivity seen in the products. It is apparent that these covalent adducts reduce the internal cavity volume, sterically limiting substrate interactions with the reactive groups, as well as solvent access to potential intermediates in the reaction pathway.


Asunto(s)
Adipocitos/química , Proteínas Portadoras/química , Proteína P2 de Mielina/química , Proteínas de Neoplasias , Proteínas Portadoras/metabolismo , Catálisis , Cristalografía por Rayos X , Proteínas de Unión a Ácidos Grasos , Ácidos Grasos/metabolismo , Datos de Secuencia Molecular , Proteína P2 de Mielina/metabolismo , Fenantrolinas/química , Conformación Proteica , Piridoxamina/química , Proteínas Recombinantes/química
13.
Bioorg Med Chem Lett ; 8(7): 875-80, 1998 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-9871558

RESUMEN

Fatty acid binding proteins are a class of small 15 kDa proteins with a simple architecture that forms a large solvent sequestered cavity. In previous work, we demonstrated that reductive amination reactions could be performed in this cavity by covalent attachment of a pyridoxamine cofactor and that the rate, enantioselectivity and substrate specificity of these reactions could be altered by site directed mutagenesis. Herein, we show that the chemistry performed by these conjugates can be extended to include catalytic transamination and describe the effects of added metal ions on reaction rate and enantioselectivity. We conclude that metal ions can be used to increase the rate of reactions catalyzed by semisynthetic transaminases; however, the addition of metal ions can also retard the reaction rate. Furthermore, it appears that the presence of metal ions almost always results in an erosion of reaction enantioselectivity. This limits their utility as a practical means of increasing reaction rate. The results reported here, for four independent systems, should be considered in future designs of artificial transaminases.


Asunto(s)
Proteínas Portadoras/metabolismo , Cobre/farmacología , Proteína P2 de Mielina/metabolismo , Proteínas de Neoplasias , Níquel/farmacología , Transaminasas/química , Transaminasas/metabolismo , Zinc/farmacología , Proteínas Portadoras/síntesis química , Proteínas Portadoras/química , Catálisis , Cationes Bivalentes/farmacología , Escherichia coli , Proteínas de Unión a Ácidos Grasos , Cinética , Peso Molecular , Mutagénesis Sitio-Dirigida , Proteína P2 de Mielina/síntesis química , Proteína P2 de Mielina/química , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Transaminasas/síntesis química
14.
Chem Biol ; 4(10): 731-7, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9375251

RESUMEN

BACKGROUND: Bioactive molecules that are covalently immobilized in patterns on surfaces have previously been used to control or study cell behavior such as adhesion, spreading, movement or differentiation. Photoimmobilization techniques can be used, however, to control not only the spatial pattern of molecular immobilization, termed the micropattern, but also the surface density of the molecules--a characteristic that has not been previously exploited. RESULTS: Oligopeptides containing the bioactive Arg-Gly-Asp cell-adhesion sequence were immobilized upon self-assembled monolayers of an oligo(ethylene glycol) alkanethiolate in patterns that were visualized and quantified by autoradiography. The amount and pattern of immobilized peptide were controlled by manipulating the exposure of the sample to a UV lamp or a laser beam. Patterns of peptides, including a density gradient, were used to control the location and number of adherent cells and also the cell shape. CONCLUSIONS: A photoimmobilization technique for decorating surfaces with micropatterns that consist of variable densities of bioactive molecules is described. The efficacy of the patterns for controlling cell adhesion and shape has been demonstrated. This technique is useful for the study of cell behavior on micropatterns.


Asunto(s)
Alcanos/metabolismo , Biopolímeros/metabolismo , Glicol de Etileno/metabolismo , Etiquetas de Fotoafinidad/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Células 3T3 , Animales , Adhesión Celular , Línea Celular , Tamaño de la Célula , Humanos , Rayos Láser , Hígado/citología , Ratones , Células PC12 , Péptidos/metabolismo , Ratas , Propiedades de Superficie
15.
Bioconjug Chem ; 8(5): 658-63, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9327128

RESUMEN

Gradients of biological molecules on a microscale have been postulated to elicit cellular responses, such as migration. However, it has been difficult to prepare such gradients for experimental testing. A means for producing such gradients has been developed using a heterobifunctional photolinking agent with laser light activation. The photolinking agent synthesized includes an N-hydroxysuccinimide group and a photoreactive benzophenone (BP) separated by a tetraethylene glycol (TEG) spacer. The presence of the tetraethylene glycol spacer renders the photolinker hydrophilic, a desirable trait for conjugation in aqueous solutions. The linker was then conjugated to R-phycoerythrin (R-PE), a fluorescent protein. The resulting photolinker-R-phycoerythrin conjugate (BP-TEG-PE) was then immobilized onto a polystyrene surface by laser irradiation on a motorized stage. By varying exposure time of the sample to the beam, the amount of BP-TEG-PE immobilized on the surface was changed over an order of magnitude over a distance of 250 microns. This method can be applied to prepare gradients of proteins that elicit biological responses, such as extracellular matrix proteins or growth factors, and to study the biological effects of such gradients.


Asunto(s)
Proteínas/química , Alanina/química , Benzofenonas/química , Reactivos de Enlaces Cruzados , Glicoles de Etileno/química , Indicadores y Reactivos , Rayos Láser , Fotoquímica , Ficoeritrina/química , Poliestirenos , Succinimidas/química , Propiedades de Superficie
16.
Biochem Biophys Res Commun ; 235(2): 377-82, 1997 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-9199201

RESUMEN

Farnesyl protein transferase (FPTase) catalyzes the covalent attachment of a farnesyl (C15) group from farnesyl pyrophosphate (FPP) to a specific cysteine residue of Ras and several other proteins. In this report, photoactive farnesyl and geranylgeranyl pyrophosphate analogs 2-diazo-3,3,3-trifluoropropionyloxy-geranyl pyrophosphate (DATFP-GPP) and 2-diazo-3,3,3-trifluoropropionyloxy-farnesyl pyrophosphate (DATFP-FPP) were used to study the active site of Saccharomyces cerevisiae FPTase. Both analogs are substrates for the enzyme, and upon irradiation, DATFP-GPP inhibits FPTase activity in a time-dependent manner. Photoinactivation by DATFP-GPP is prevented by the presence of the natural substrate FPP. Photolysis of radiolabeled DATFP-GPP results in preferential labeling of the beta subunit of FPTase, suggesting that this subunit is involved in recognition of FPP. Of particular importance, DATFP-GPP and DATFP-FPP were used to enzymatically transfer the photoactive isoprenoid moieties to peptides and to Ras; such molecules should be useful for identifying cellular components which specifically recognize farnesylated Ras and other prenylated proteins.


Asunto(s)
Marcadores de Afinidad/metabolismo , Transferasas Alquil y Aril , Prenilación de Proteína , Saccharomyces cerevisiae/enzimología , Transferasas/metabolismo , Proteínas ras/metabolismo , Marcadores de Afinidad/síntesis química , Sitios de Unión , Compuestos de Dansilo/síntesis química , Compuestos de Dansilo/metabolismo , Compuestos de Diazonio/síntesis química , Compuestos de Diazonio/metabolismo , Compuestos de Diazonio/farmacología , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Luz , Estructura Molecular , Fotólisis , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos , Transferasas/antagonistas & inhibidores , Transferasas/química
17.
Proc Natl Acad Sci U S A ; 90(4): 1179-83, 1993 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8433980

RESUMEN

Cooperativity in oligonucleotide-directed sequence-specific recognition of DNA by triple helix formation can be enhanced by the addition of discrete dimerization domains. The equilibrium association constants for cooperative binding of oligonucleotides that dimerize by Watson-Crick hydrogen bonds and occupy adjacent sites on double helical DNA by triple helix formation have been measured by quantitative affinity cleavage titration. For two oligonucleotides that bind unique neighboring 11-bp and 15-bp sites on double helical DNA, and dimerize by formation of an 8-bp Watson-Crick mini-helix, the free energy of binding is -8.0 and -9.7 kcal.mol-1, respectively, and the cooperative energy of interaction is -2.3 kcal.mol-1 (1 kcal = 4.18 kJ). The energetics of this artificial nucleic acid cooperative intermolecular assembly can mimic naturally occurring cooperative protein-DNA systems, such as the phage lambda repressor.


Asunto(s)
ADN Bacteriano/química , ADN/química , Conformación de Ácido Nucleico , Oligodesoxirribonucleótidos/química , Secuencia de Bases , Sitios de Unión , ADN Bacteriano/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Indicadores y Reactivos , Sustancias Macromoleculares , Matemática , Modelos Moleculares , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/síntesis química , Plásmidos
18.
Nature ; 352(6331): 168-72, 1991 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-2067577

RESUMEN

Several hundred million tons of toxic mercurials are dispersed in the biosphere. Microbes can detoxify organo-mercurials and mercury salts through sequential action of two enzymes, organomercury lyase and mercuric ion reductase (MerA). The latter, a homodimer with homology to the FAD-dependent disulphide oxidoreductases, catalyses the reaction NADPH + Hg(II)----NADP+ + H+ + Hg(0), one of the very rare enzymic reactions with metal substrates. Human glutathione reductase serves as a reference molecule for FAD-dependent disulphide reductases and between its primary structure and that of MerA from Tn501 (Pseudomonas), Tn21 (Shigella), p1258 (Staphylococcus) and Bacillus, 25-30% of the residues have been conserved. All MerAs have a C-terminal extension about 15 residues long but have very varied N termini. Although the enzyme from Streptomyces lividans has no addition, from Pseudomonas aeruginosa Tn501 and Bacillus sp. strain RC607 it has one and two copies respectively of a domain of 80-85 residues, highly homologous to MerP, the periplasmic component of proteins encoded by the mer operon. These domains can be proteolytically cleaved off without changing the catalytic efficiency. We report here the crystal structure of MerA from the Gram-positive bacterium Bacillus sp. strain RC607. Analysis of its complexes with nicotinamide dinucleotide substrates and the inhibitor Cd(II) reveals how limited structural changes enable an enzyme to accept as substrate what used to be a dangerous inhibitor. Knowledge of the mode of mercury ligation is a prerequisite for understanding this unique detoxification mechanism.


Asunto(s)
Bacillus/enzimología , Oxidorreductasas/química , Sitios de Unión , Cadmio/farmacología , Cristalización , Disulfuros/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Glutatión Reductasa/química , Glutatión Reductasa/metabolismo , Humanos , Sustancias Macromoleculares , Mercurio/metabolismo , NADP/metabolismo , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/metabolismo , Conformación Proteica
19.
Biochemistry ; 29(11): 2703-13, 1990 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-2189495

RESUMEN

Mercuric reductase catalyzes the two-electron reduction of Hg(II) to Hg(0) using NADPH as the reductant; this reaction constitutes the molecular basis for detoxification of Hg(II) by bacteria. The enzyme is an alpha 2 homodimer and possesses two pairs of cysteine residues, Cys135 and Cys140 (redox-active pair) and Cys558 and Cys559 (C-terminal pair), which are known to be essential for catalysis. In the present study, we have obtained evidence for an intersubunit active site, consisting of a redox-active cysteine pair from one subunit and a C-terminal pair from the adjacent subunit, by reconstituting catalytic activity both in vivo and in vitro starting with two inactive, mutant enzymes, Ala135Ala140Cys558Cys559 (AACC) and Cys135Cys140Ala558Ala559 (CCAA). Genetic complementation studies were used to show that coexpression of AACC and CCAA in the same cell yielded an HgR phenotype, some 10(4)-fold more resistant than cells expressing only one mutant. Purification and catalytic characterization of a similarly coexpressed protein mixture showed the mixture to have activity levels ca. 25% those of wild type; this is the same as that statistically anticipated for a CCAA-AACC heterodimeric/homodimeric mixture with only one functional active site per heterodimer. Actual physical evidence for the formation of active mutant heterodimers was obtained by chaotrope-induced subunit interchange of inactive pure CCAA and AACC homodimers in vitro followed by electrophoretic separation of heterodimers from homodimers. Taken together, these data provide compelling evidence that the active site in mercuric reductase resides at the subunit interface and contains cysteine residues originating from separate polypeptide chains.


Asunto(s)
Cisteína/genética , Eritrocitos/metabolismo , Escherichia coli/genética , Oxidorreductasas/genética , Secuencia de Bases , Sitios de Unión , Catálisis , Cisteína/metabolismo , ADN/genética , Prueba de Complementación Genética , Humanos , Mutación , Oxidación-Reducción , Oxidorreductasas/metabolismo , Regiones Promotoras Genéticas , Conformación Proteica
20.
Biochemistry ; 29(11): 2831-41, 1990 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-2189497

RESUMEN

A mutant form of mercuric reductase, which has three of its four catalytically essential cysteine residues replaced by alanines (ACAA: Ala135Cys140Ala558Ala559), has been constructed and used for mechanistic investigations. With disruption of the Hg(II) binding site, the mutant enzyme is devoid of Hg(II) reductase activity. However, it appears to fold properly since it binds FAD normally and exhibits very tight binding of pyridine nucleotides as is seen with the wild-type enzyme. This mutant enzyme allows quantitative accumulation of two species thought to function as intermediates in the catalytic sequence of the flavoprotein disulfide reductase family of enzymes. NADPH reduces the flavin in this mutant, and a stabilized E-FADH- form accumulates. The second intermediate is a flavin C(4a)-Cys140 thiol adduct, which is quantitatively accumulated by reaction of oxidized ACAA enzyme with NADP+. The conversion of the Cys135-Cys140 disulfide in wild-type enzyme to the monothiol Cys140 in ACAA and the elevated pKa of Cys140 (6.7 vs 5.0 in wild type) have permitted detection of these intermediates at low pH (5.0). The rates of formation of E-FADH- and the breakdown of the flavin C(4a)-thiol adduct have been measured and indicate that both intermediates are kinetically competent for both the reductive half-reaction and turnover by wild-type enzyme. These results validate the general proposal that electrons flow from NADPH to FADH- to C(4a)-thiol adduct to the FAD/dithiol form that accumulates as the EH2 form in the reductive half-reaction for this class of enzymes.


Asunto(s)
Flavinas/metabolismo , Oxidorreductasas/genética , Compuestos de Sulfhidrilo/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Escherichia coli/genética , Flavina-Adenina Dinucleótido/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Mutación , NAD , Oxidación-Reducción , Oxidorreductasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA