Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Genet ; 19(3): 254-6, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9662397

RESUMEN

The breast cancer specific tumour suppressor protein, BRCA1 (refs 1,2), activates transcription when linked with a DNA-binding domain and is a component of the RNA polymerase II (Pol II) holoenzyme. We show here that RNA helicase A (RHA) protein links BRCA1 to the holoenzyme complex. The region of BRCA1 which interacts with RHA and, thus, the holoenzyme complex, corresponds to subregions of the BRCT domain of BRCA1 (ref. 9). This interaction was shown to occur in yeast nuclei, and expression in human cells of a truncated RHA molecule which retains binding to BRCA1 inhibited transcriptional activation mediated by the BRCA1 carboxy terminus. These data are the first to identify a specific protein interaction with the BRCA1 C-terminal domain and are consistent with the model that BRCA1 functions as a transcriptional coactivator.


Asunto(s)
Proteína BRCA1/metabolismo , Coenzimas/metabolismo , ARN Nucleotidiltransferasas/metabolismo , ARN Polimerasa II/metabolismo , Animales , Proteína BRCA1/genética , Células HeLa , Humanos , Mutagénesis , ARN Helicasas , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
2.
Mol Endocrinol ; 11(13): 1971-84, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9415401

RESUMEN

In androgen target tissues, 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) may regulate occupancy of the androgen receptor (AR) by catalyzing the interconversion of 5alpha-dihydrotestosterone (5alpha-DHT) (a potent androgen) and 3alpha-androstanediol (a weak androgen). In this study, a 3alpha-HSD cDNA (1170 bp) was isolated from a human prostate cDNA library. The human prostatic 3alpha-HSD cDNA encodes a 323-amino acid protein with 69.9%, 84.1%, 99.4%, and 87.9% sequence identity to rat liver 3alpha-HSD and human type 1, type 2, and type 3 3alpha-HSDs, respectively, and is a member of the aldo-keto reductase superfamily. The close homology with human type 2 3alpha-HSD suggests that it is either identical to this enzyme or a structural allele. Surprisingly, when the recombinant protein was expressed and purified from Escherichia coli, the enzyme did not oxidize androsterone when measured spectrophotometrically, an activity previously assigned to recombinant type 2 3alpha-HSD using this assay. Complete kinetic characterization of the purified protein using spectrophotometric, fluorometric, and radiometric assays showed that the catalytic efficiency favored 3alpha-androstanediol oxidation over 5alpha-DHT reduction. Using [14C]-5alpha-DHT as substrate, TLC analysis confirmed that the reaction product was [14C]-3alpha-androstanediol. However, in the reverse reaction, [3H]-3alpha-androstanediol was oxidized first to [3H]-androsterone and then to [3H]-androstanedione, revealing that the expressed protein possessed both 3alpha- and 17beta-HSD activities. The 17beta-HSD activity accounted for the higher catalytic efficiency observed with 3alpha-androstanediol. These findings indicate that, in the prostate, type 2 3alpha-HSD does not interconvert 5alpha-DHT and 3alpha-androstanediol but inactivates 5alpha-DHT through its 3-ketosteroid reductase activity. Levels of 3alpha-HSD mRNA were measured in primary cultures of human prostatic cells and were higher in epithelial cells than stromal cells. In addition, elevated levels of 3alpha-HSD mRNA were observed in epithelial cells derived from benign prostatic hyperplasia and prostate carcinoma tissues. Expression of 3alpha-HSD was not prostate specific, since high levels of mRNA were also found in liver, small intestine, colon, lung, and kidney. This study is the first complete characterization of recombinant type 2 3alpha-HSD demonstrating dual activity and cellular distribution in the human prostate.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/genética , Próstata/enzimología , Proteínas Recombinantes/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/química , Secuencia de Aminoácidos , Secuencia de Bases , Northern Blotting , Células Cultivadas , Clonación Molecular , ADN Complementario/aislamiento & purificación , Activación Enzimática , Humanos , Cinética , Masculino , Datos de Secuencia Molecular , Complejos Multienzimáticos/biosíntesis , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Especificidad de Órganos/genética , Próstata/metabolismo
3.
Steroids ; 62(1): 101-11, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9029723

RESUMEN

Mammalian 3 alpha-hydroxysteroid dehydrogenases (3 alpha-HSDs) inactivate circulating steroid hormones, and in target tissues regulate the occupancy of steroid hormone receptors. Molecular cloning indicates that 3 alpha-HSDs are members of the aldo-keto reductase (AKR) superfamily and display high sequence identity (> 60%). Of these, the most extensively characterized is rat liver 3 alpha-HSD. X-ray crystal structures of the apoenzyme and the E.NADP+ complex have been determined and serve as structural templates for other 3 alpha-HSDs. These structures reveal that rat liver 3 alpha-HSD adopts an (alpha/beta)8-barrel protein fold. NAD(P)(H) lies perpendicular to the barrel axis in an extended conformation, with the nicotinamide ring at the core of the barrel, and the adenine ring at the periphery of the structure. The nicotinamide ring is stabilized by interaction with Y216, S166, D167, and Q190, so that the A-face points into the vacant active site. The 4-pro-(R) hydrogen transferred in the oxidoreduction of steroids is in close proximity to a catalytic tetrad that consists of D50, Y55, K84, and H117. A water molecule is within hydrogen bond distance of H117 and Y55, and its position may mimic the position of the carbonyl of a 3-ketosteroid substrate. The catalytic tetrad is conserved in members of the AKR superfamily and resides at the base of an apolar cleft implicated in binding steroid hormone. The apolar cleft consists of a side of apolar residues (L54, W86, F128, and F129), and opposing this side is a flexible loop that contains W227. These constraints suggest that the alpha-face of the steroid would orient itself along that side of the cleft containing W86. Site-directed mutagenesis of the catalytic tetrad indicates that Y55 and K84 are essential for catalysis. Y55S and Y55F mutants are catalytically inactive, but still form binary (E.NADPH) and ternary (E.NADH.Testosterone) complexes; by contrast K84R and K84M mutants are catalytically inactive, but do not bind steroid hormone. The reliance on a Tyr/Lys pair is reminiscent of catalytic mechanisms proposed for other AKR members as well as for HSDs that belong to the short-chain dehydrogenase/reductase (SDR) family, in which Tyr is the general acid, with its pKa being lowered by Lys. Superimposition of the nicotinamide rings in the structures of 3 alpha-HSD (an AKR) and 3 alpha, 20 beta-HSD (an SDR) show that the Tyr/Lys pairs are positionally conserved, suggesting convergent evolution across protein families to a common mechanism for HSD catalysis. W86Y and W227Y mutants bind testosterone to the E.NADH complex, with effective increases in Kd of 8- and 20-fold. These data provide the first evidence that the side of the apolar cleft containing W86 and the opposing flexible loop containing W227 are parts of the steroid-binding site. Detailed mutagenesis studies of the apolar cleft and elucidation of a ternary complex structure will ultimately provide details of the determinants that govern steroid hormone recognition. These determinants could provide a rational basis for structure-based inhibitor design.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/química , 3-Hidroxiesteroide Deshidrogenasas/fisiología , Oxidorreductasas de Alcohol/química , Hígado/enzimología , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica) , Oxidorreductasas de Alcohol/fisiología , Aldehído Reductasa , Aldo-Ceto Reductasas , Secuencia de Aminoácidos , Animales , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Ratas , Homología de Secuencia de Aminoácido , Esteroides/metabolismo
4.
Steroids ; 61(9): 508-23, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8883217

RESUMEN

Mammalian 3 alpha-hydroxysteroid dehydrogenases (3 alpha-HSDs) regulate steroid hormone levels. For example, hepatic 3 alpha-HSDs inactivate circulating androgens, progestins, and glucocorticoids. In target tissues they regulate access of steroid hormones to steroid hormone receptors. For example, in the prostate 3 alpha-HSD acts as a molecular switch and controls the amount of 5 alpha-dihydrotestosterone that can bind to the androgen receptor, while in the brain 3 alpha-HSD can regulate the amount of tetrahydrosteroids that can alter GABAa receptor function. Molecular cloning indicates that these mammalian 3 alpha-HSDs belong to the aldo-keto reductase superfamily and that they are highly homologous proteins. Using the three-dimensional structure of rat liver 3 alpha-HSD as a template for site-directed mutagenesis, details regarding structure function relationships, including catalysis and cofactor and steroid hormone recognition have been elucidated. These details may be relevant to all mammalian 3 alpha-HSDs.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/química , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica) , Secuencia de Aminoácidos , Animales , Encéfalo/enzimología , Clonación Molecular , Cristalografía por Rayos X , Predicción , Humanos , Cinética , Hígado/enzimología , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Próstata/enzimología , Conformación Proteica , Conejos , Ratas , Homología de Secuencia de Aminoácido , Esteroides/metabolismo , Especificidad por Sustrato
6.
Biochemistry ; 37(31): 11003-11, 1998 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-9692994

RESUMEN

Aldo-keto reductases (AKR) are monomeric oxidoreductases that retain a conserved catalytic tetrad (Tyr, Lys, Asp, and His) at their active sites in which the Tyr acts as a general acid-base catalyst. In rat liver 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD, AKR1C9), a well-characterized AKR, the catalytic tyrosine is Tyr 55. This enzyme displays a high catalytic efficiency for a common AKR substrate 9,10-phenanthrenequinone (9,10-PQ). Surprisingly, Y55F and Y55S mutants of 3alpha-HSD reduced 9,10-PQ with high kcat values. This is the first report whereby the invariant catalytic tyrosine of an AKR has been mutated with retention of kcat values similar to wild-type enzyme. The Y55F and Y55S mutants displayed narrow substrate specificity and reduced select aromatic quinones and alpha-dicarbonyls. kcat versus pH profiles for steroid oxidoreduction catalyzed by wild-type 3alpha-HSD exhibited a single ionizable group with a pK= 7.0-7.5, which has been assigned to Tyr 55. This group was not evident in the kcat versus pH profiles for 9, 10-PQ reduction catalyzed by either wild-type or the Tyr 55 mutant enzymes, indicating that the protonation state of Tyr 55 is unimportant for 9,10-PQ turnover. Instead, wild-type and the active-site mutants Y55F, Y55S, H117A, D50N, K84R, and K84M showed the presence of a new titratable group with a pKb = 8.3-9.9. Thus, the group being titrated is not part of the tetrad. All the mutants decreased kcat/Km considerably more than they decreased kcat. Thus, the K84R mutant demonstrated a 30-fold decrease in the pH-independent value of kcat but 2200-fold decrease in the pH-independent value of kcat/Km. This suggests that all the tetrad residues influence quinone binding and that Lys 84 plays a dominant role in maintaining proper substrate orientation. Using wild-type enzyme, the energy of activation (Ea) for 9,10-PQ reduction was approximately 11 kcal/mol less than steroid oxidoreduction. The Ea for 9,10-PQ reduction was unchanged in the Tyr 55 mutants, suggesting that the reaction proceeds through the same low-energy barrier in the wild-type enzyme and these mutants. The retention of quinone reductase activity in this AKR in the absence of Tyr 55 with kcat versus pH rate profiles and activation energies identical to wild-type enzyme suggests that quinone reduction occurs via a mechanism that differs from 3-ketosteroid reduction. In this mechanism, the electron donor (NADPH) and acceptor (o-quinone) are bound in close proximity, which permits hydride transfer without formal protonation of the acceptor carbonyl by Tyr 55. This represents a rare example where one enzyme can catalyze the same chemical reaction (carbonyl reduction) by either acid catalysis or by a propinquity effect and where these two mechanisms can be discriminated by site-directed mutagenesis.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Mutagénesis Sitio-Dirigida , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , NADP/metabolismo , Tirosina/genética , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Oxidorreductasas de Alcohol/genética , Aldehído Reductasa , Aldo-Ceto Reductasas , Animales , Catálisis , Transferencia de Energía/genética , Activación Enzimática/genética , Concentración de Iones de Hidrógeno , Mutágenos/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , Oxidación-Reducción , Fenantrenos/metabolismo , Ratas , Especificidad por Sustrato/genética
7.
J Biol Chem ; 271(47): 30190-8, 1996 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-8939970

RESUMEN

Rat liver 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD), a member of the aldoketoreductase superfamily, inactivates circulating steroid hormones using NAD(P)H as cofactor. Despite determination of the 3alpha-HSD.NADP+ binary complex structure, the functional elements that dictate the binding of steroids remain unclear (Bennett, M.J., Schlegel, B.P., Jez, J.M., Penning, T.M., and Lewis, M. (1996) Biochemistry 35, 10702-10711). Two tryptophans (Trp86 and Trp227) near the active site may have roles in substrate binding, and their fluorescence may be quenched upon binding of NADPH. Trp86 is located within an apolar cleft, while Trp227 is found on an opposing loop near the active site. A third tryptophan, Trp148, is on the periphery of the structure. To investigate the roles of these tryptophans in protein fluorescence and ligand binding, we generated three mutant enzymes (W86Y, W148Y, and W227Y) by site-directed mutagenesis. Spectroscopic measurements on these proteins showed that Trp148 contributed the most to the enzyme fluorescence spectra, with Trp227 adding the least. Trp86 was identified as the tryptophan quenched by bound NADPH through an energy transfer mechanism. The W86Y mutant altered binding of cofactor (a 3-fold increase in Kd for NADPH) and steroid (a 7-fold increase in Kd for testosterone). This mutation also dramatically decreased the catalytic efficiency observed with one-, two-, and three-ring substrates and decreased the binding affinity for nonsteroidal anti-inflammatory drugs but had little effect on the binding of aldose reductase inhibitors. Interestingly, mutation of Trp227 significantly impaired steroid binding (a 22-fold increase in Kd for testosterone), but did not alter binding of cofactor, smaller substrates, or inhibitors. Kinetically, the W148Y mutant was similar to wild-type enzyme. Our results demonstrate that Trp86 and the apolar cleft is part of the substrate binding pocket. In addition, we propose a role for Trp227 and its associated loop in binding steroids, but not small substrates or inhibitors, most likely through interaction with the C- and D-rings of the steroid. This work provides the first evidence that tryptophans on opposite sides of the apolar cleft are part of the steroid binding pocket and suggests how the enzyme may discriminate between nonsteroidal anti-inflammatory drugs and aldose reductase inhibitors like zopolrestat. A model of how androstanedione binds in the apolar cleft is developed. These data provide further evidence that loop structures in members of the aldoketoreductase superfamily are critical determinants of ligand binding.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Hígado/enzimología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Oxidorreductasas , Triptófano/química , 3-Hidroxiesteroide Deshidrogenasas/química , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica) , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Animales , Inhibidores Enzimáticos/farmacología , Fluorescencia , Cinética , Ligandos , Mutagénesis Sitio-Dirigida , NADP/metabolismo , Ratas , Especificidad por Sustrato , Testosterona/metabolismo
8.
Biochemistry ; 37(10): 3538-48, 1998 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-9521675

RESUMEN

Rat liver 3 alpha-hydroxysteroid dehydrogenase (3 alpha-HSD, E.C. 1.1.1.213, AKR1C9) is a member of the aldo-keto reductase (AKR) superfamily which inactivates circulating steroid hormones. We have proposed a catalytic mechanism in which Tyr 55 acts as a general acid with its pK value being lowered by a hydrogen bond with Lys 84 which is salt-linked to Asp 50. To test this mechanism, residues at the active site were mutated and the mutant enzymes (Y55F, Y55S, K84M, K84R, D50N, D50E, and H117A) were purified to homogeneity from an Escherichia coli expression system. Spectrophotometric assays showed that mutations of Tyr 55 and Lys 84 gave enzymes that were apparently inactive for steroid oxidation and reduction. All mutants appeared inactive for steroid reduction. The catalytic efficiencies for steroid oxidation were reduced 4-10-fold for the Asp 50 mutants and 300-fold for the H117A mutant. Fluorescence titration with NADPH demonstrated that each mutant bound cofactor unimpeded. Equilibrium dialysis indicated that the competitive inhibitor testosterone formed E.NADH.testosterone complexes only with the Y55F, Y55S, and D50N mutants with Kd values 10-fold greater than those for wild-type. Therefore the loss of steroid oxidoreductase activity observed for the Tyr 55 mutants cannot be attributed simply to an inability to bind steroid. Using a highly sensitive radiometric assay in which the conversion of [14C]-5 alpha-dihydrotestosterone (DHT) to [14C]-3 alpha-androstanediol (3 alpha-Diol) was measured, the rate enhancement (kcat/knoncat) for the reaction was estimated to be 2.6 x 10(9). Using this assay, all mutants formed steroid product with decreases in an overall rate enhancement of 10(1)-10(4). It was found that Tyr 55 made the single largest contribution to rate enhancement. This is the first instance where point mutations in the conserved catalytic tetrad of an AKR yielded enzymes which were still catalytically active. This enabled the construction of pH vs kcat profiles for the reduction of [14C]-5 alpha-DHT catalyzed by the tetrad mutants. These profiles revealed that the titratable group assigned to the general acid (pK = 6.50 +/- 0.42) was eliminated in the Y55F and H117A mutants. The pH-independent value of kcat was decreased in the H117A and Y55F mutants, by 2 and 4 log units, respectively. pH vs kcat(app) profiles for the oxidation of [3H]-3 alpha-Diol showed that the same titratable group (pK = 7.50 +/- 0.30) was eliminated in both the Y55F and K84M mutants but was retained in the H117A mutant. Since only the Y55F mutant eliminated the titratable group in both the reduction and oxidation directions it is assigned as the catalytic general acid/base. The differential effects of His 117 and Lys 84 on the ionization of Tyr 55 are explained by a "push-pull" mechanism in which His 117 facilitates proton donation and Lys 84 facilitates proton removal by Tyr 55.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/química , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica) , Oxidorreductasas de Alcohol/química , Aldehído Reductasa , Aldo-Ceto Reductasas , Animales , Secuencia de Bases , Sitios de Unión/genética , Cartilla de ADN/genética , Escherichia coli/genética , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Cinética , Modelos Químicos , Mutagénesis Sitio-Dirigida , NADP/metabolismo , Oxidación-Reducción , Mutación Puntual , Protones , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esteroides/metabolismo
9.
Biochemistry ; 35(33): 10702-11, 1996 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-8718859

RESUMEN

Rat liver 3 alpha-hydroxysteroid/dihydrodiol dehydrogenase (3 alpha-HSD) inactivates circulating steroid hormones and is involved in polycyclic aromatic hydrocarbon (PAH) carcinogenesis. It is the only HSD of known structure in the aldo-keto reductase (AKR) superfamily and may provide a paradigm for other mammalian HSDs in this family. The structure of the 3 alpha-HSD.NADP+ binary complex has been determined at 2.7 A resolution and refined to a crystallographic R-factor of 23.4% with good geometry. The model is similar to other binary complexes in the AKR superfamily in that NADP+ binds at the C-terminal end of an alpha/beta barrel. However, it is unique in that NADP+ is bound in two alternate conformations, probably because of the lack of a salt-linked "safety belt" over the pyrophosphate bridge. The structure supports a previously proposed catalytic mechanism for carbonyl reduction in which Tyr 55 is the general acid, and its effective pKa is lowered by the adjacent Lys 84. We present evidence that the structurally distinct short-chain dehydrogenase/reductase (SDR) superfamily may have convergently evolved a similar catalytic mechanism. Insight into substrate binding is offered by a crystal packing contact in which a neighboring molecule inserts a tryptophan residue (Trp 227) into an apolar cleft in 3 alpha-HSD. This cleft is proximal to the bound NADP+ cofactor and contains a surface of apolar residues (Leu 54, Trp 86, Leu 122, Phe 128, Phe 129, Leu 137, Phe 139), making it a likely candidate for the substrate-binding site. Thus, in forming this crystal contact, Trp 227 may mimic a portion of a bound steroid. In addition, we propose that a water molecule in the active site indicates the position of the hydroxyl oxygen in a 3 alpha-hydroxysteroid substrate. Knowledge of the position of this water molecule, combined with the stereochemistry of hydride transfer, suggests that the alpha face of a bound steroid will be oriented toward the side of the apolar cleft containing Trp 86.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/química , NADP/química , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Hígado/enzimología , Masculino , Modelos Moleculares , Estructura Molecular , NADP/metabolismo , Ratas , Ratas Sprague-Dawley , Esteroides/metabolismo
10.
Biochem J ; 326 ( Pt 3): 625-36, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9307009

RESUMEN

The aldo-keto reductases metabolize a wide range of substrates and are potential drug targets. This protein superfamily includes aldose reductases, aldehyde reductases, hydroxysteroid dehydrogenases and dihydrodiol dehydrogenases. By combining multiple sequence alignments with known three-dimensional structures and the results of site-directed mutagenesis studies, we have developed a structure/function analysis of this superfamily. Our studies suggest that the (alpha/beta)8-barrel fold provides a common scaffold for an NAD(P)(H)-dependent catalytic activity, with substrate specificity determined by variation of loops on the C-terminal side of the barrel. All the aldo-keto reductases are dependent on nicotinamide cofactors for catalysis and retain a similar cofactor binding site, even among proteins with less than 30% amino acid sequence identity. Likewise, the aldo-keto reductase active site is highly conserved. However, our alignments indicate that variation ofa single residue in the active site may alter the reaction mechanism from carbonyl oxidoreduction to carbon-carbon double-bond reduction, as in the 3-oxo-5beta-steroid 4-dehydrogenases (Delta4-3-ketosteroid 5beta-reductases) of the superfamily. Comparison of the proposed substrate binding pocket suggests residues 54 and 118, near the active site, as possible discriminators between sugar and steroid substrates. In addition, sequence alignment and subsequent homology modelling of mouse liver 17beta-hydroxysteroid dehydrogenase and rat ovary 20alpha-hydroxysteroid dehydrogenase indicate that three loops on the C-terminal side of the barrel play potential roles in determining the positional and stereo-specificity of the hydroxysteroid dehydrogenases. Finally, we propose that the aldo-keto reductase superfamily may represent an example of divergent evolution from an ancestral multifunctional oxidoreductase and an example of convergent evolution to the same active-site constellation as the short-chain dehydrogenase/reductase superfamily.


Asunto(s)
Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Evolución Molecular , Aldehído Reductasa , Aldo-Ceto Reductasas , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Ratas , Alineación de Secuencia , Análisis de Secuencia , Relación Estructura-Actividad
11.
Proc Natl Acad Sci U S A ; 97(7): 3148-53, 2000 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-10725406

RESUMEN

The functions of most of the 12 subunits of the RNA polymerase II (Pol II) enzyme are unknown. In this study, we demonstrate that two of the subunits, hRPB2 and hRPB10alpha, mediate the regulated stimulation of transcription. We find that the transcriptional coactivator BRCA1 interacts directly with the core Pol II complex in vitro. We tested whether single subunits from Pol II would compete with the intact Pol II complex to inhibit transcription stimulated by BRCA1. Excess purified Pol II subunits hRPB2 or hRPB10alpha blocked BRCA1- and VP16-dependent transcriptional activation in vitro with minimal effect on basal transcription. No other Pol II subunits tested inhibited activated transcription in these assays. Furthermore, hRPB10alpha, but not hRPB2, blocked Sp1-dependent activation.


Asunto(s)
Proteína BRCA1/metabolismo , ARN Polimerasa II/metabolismo , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Clonación Molecular , Humanos , Datos de Secuencia Molecular , Unión Proteica , ARN Polimerasa II/genética , ARN Polimerasa II/fisiología , Proteínas Recombinantes/metabolismo , Factor de Transcripción Sp1/metabolismo , Transactivadores/metabolismo
12.
Nucleic Acids Res ; 26(3): 847-53, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9443979

RESUMEN

The RNA polymerase II (Pol II) holoenzyme in yeast is an essential transcriptional regulatory complex which has been defined by genetic and biochemical approaches. The mammalian counterpart to this complex, however, is less well defined. Experiments herein demonstrate that, along with Pol II and SRB proteins, proteins associated with transcriptional regulation as cofactors are associated with the Pol II holoenzyme. Earlier experiments have demonstrated that the breast cancer-associated tumor suppressor BRCA1 and the CREB binding protein (CBP) were associated with the holoenzyme complex. The protein related to CBP, the E1A-associated p300 protein, is shown in these experiments to be associated with the holoenzyme complex as well as the BRG1 subunit of the chromatin remodeling SWI/SNF complex. Importantly, the Pol II holoenzyme complex does not contain some factors previously reported as stoichiometric components of the holoenzyme complex, most notably the proteins which function in repair of damaged DNA, such as PCNA, RFC and RPA. The presence of the p300 coactivator and the chromatin-modifying BRG1 protein support a role for the Pol II holoenzyme as a key target for regulation by enhancer binding proteins.


Asunto(s)
Coenzimas/química , Proteínas Nucleares/análisis , ARN Polimerasa II/química , Transactivadores , Factores de Transcripción/análisis , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína de Unión a CREB , Cromatografía de Afinidad/métodos , ADN/análisis , ADN Helicasas , Reparación del ADN , Proteína p300 Asociada a E1A , Células HeLa , Humanos , Complejo Mediador , Proteínas Nucleares/metabolismo , Mutación Puntual , ARN/análisis , ARN Polimerasa II/aislamiento & purificación , Proteínas Recombinantes de Fusión , Factores de Transcripción/metabolismo
13.
Biochemistry ; 33(34): 10367-74, 1994 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-8068673

RESUMEN

Rat liver 3 alpha-hydroxysteroid dehydrogenase (3 alpha-HSD, EC 1.1.1.50) inactivates circulating androgens, progestins, and glucocorticoids. 3 alpha-HSD is a member of the aldo-keto reductase superfamily, and the X-ray structure of the apoenzyme shows the presence of an (alpha/beta)8 barrel [Hoog, S. S., Pawlowski, J. E., Alzari, P. M., Penning, T. M., & Lewis, M. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 2517-2521]. As yet, a three-dimensional structure of the ternary complex E.NADPH.steroid is unavailable. To identify regions of the enzyme involved in steroid hormone recognition, we have employed mechanism-based inactivators and site-directed mutagenesis. (3 RS)-1,10-Seco-5 alpha-estr-1-yne-3,17 beta-diol (1) and (17 RS)- 17-hydroxy-14,15-secoandrost-4-en-15-yn-3-one (3) are secosteroids which contain latent Michael acceptors (alpha,beta-unsaturated alcohols) at opposite ends of the steroid nucleus (at the C-3 and C-17 positions, respectively). It was found that compounds 1 and 3 inactivated 3 alpha-HSD only in the presence of NAD+. The requirement for cofactor implies that 1 and 3 are oxidized to the corresponding alpha,beta-unsaturated ketones for inactivation to occur. Chemically prepared 17 beta-hydroxy-1,10-seco-5 alpha-estr-1-yn-3-one (2) and 14,15-secoandrost-4-en-15-yne-3,17-dione (4), the presumed products of 1 and 3 oxidation, behaved as stoichiometric inactivators of 3 alpha-HSD. In the presence and absence of NAD+, 2 and 4 inactivated > 50% of the enzyme in 10 s or less.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica) , Animales , Sitios de Unión , Técnicas In Vitro , Cinética , Hígado/enzimología , Masculino , Sondas Moleculares , Estructura Molecular , Mutagénesis Sitio-Dirigida , Ratas , Ratas Sprague-Dawley , Secoesteroides/química , Secoesteroides/farmacología , Especificidad por Sustrato
14.
J Biol Chem ; 273(43): 27786-93, 1998 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-9774387

RESUMEN

A human protein (RUVBL1), consisting of 456 amino acids (50 kDa) and highly homologous to RuvB, was identified by using the 14-kDa subunit of replication protein A (hsRPA3) as bait in a yeast two-hybrid system. RuvB is a bacterial protein involved in genetic recombination that bears structural similarity to subunits of the RF-C clamp loader family of proteins. Fluorescence in situ hybridization analysis demonstrated that the RUVBL1 gene is located at 3q21, a region with frequent rearrangements in different types of leukemia and solid tumors. RUVBL1 co-immunoprecipitated with at least three other unidentified cellular proteins and was detected in the RNA polymerase II holoenzyme complex purified over multiple chromatographic steps. In addition, two yeast homologs, scRUVBL1 and scRUVBL2 with 70 and 42% identity to RUVBL1, respectively, were revealed by screening the complete Saccharomyces cerevisiae genome sequence. Yeast with a null mutation in scRUVBL1 was nonviable. Thus RUVBL1 is an eukaryotic member of the RuvB/clamp loader family of structurally related proteins from bacteria and eukaryotes that is essential for viability of yeast.


Asunto(s)
Proteínas Portadoras/metabolismo , ADN Helicasas , Genes Fúngicos , ARN Polimerasa II/metabolismo , Saccharomyces cerevisiae/genética , ATPasas Asociadas con Actividades Celulares Diversas , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Proteínas Portadoras/aislamiento & purificación , Ciclo Celular , Cromosomas Humanos Par 3/genética , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Células Eucariotas , Genes Esenciales , Genoma Fúngico , Humanos , Datos de Secuencia Molecular , Unión Proteica , ARN Polimerasa II/aislamiento & purificación , Proteína de Replicación A , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA