Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 510(7505): 422-426, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24814345

RESUMEN

2-Oxoglutarate (2OG)-dependent oxygenases have important roles in the regulation of gene expression via demethylation of N-methylated chromatin components and in the hydroxylation of transcription factors and splicing factor proteins. Recently, 2OG-dependent oxygenases that catalyse hydroxylation of transfer RNA and ribosomal proteins have been shown to be important in translation relating to cellular growth, TH17-cell differentiation and translational accuracy. The finding that ribosomal oxygenases (ROXs) occur in organisms ranging from prokaryotes to humans raises questions as to their structural and evolutionary relationships. In Escherichia coli, YcfD catalyses arginine hydroxylation in the ribosomal protein L16; in humans, MYC-induced nuclear antigen (MINA53; also known as MINA) and nucleolar protein 66 (NO66) catalyse histidine hydroxylation in the ribosomal proteins RPL27A and RPL8, respectively. The functional assignments of ROXs open therapeutic possibilities via either ROX inhibition or targeting of differentially modified ribosomes. Despite differences in the residue and protein selectivities of prokaryotic and eukaryotic ROXs, comparison of the crystal structures of E. coli YcfD and Rhodothermus marinus YcfD with those of human MINA53 and NO66 reveals highly conserved folds and novel dimerization modes defining a new structural subfamily of 2OG-dependent oxygenases. ROX structures with and without their substrates support their functional assignments as hydroxylases but not demethylases, and reveal how the subfamily has evolved to catalyse the hydroxylation of different residue side chains of ribosomal proteins. Comparison of ROX crystal structures with those of other JmjC-domain-containing hydroxylases, including the hypoxia-inducible factor asparaginyl hydroxylase FIH and histone N(ε)-methyl lysine demethylases, identifies branch points in 2OG-dependent oxygenase evolution and distinguishes between JmjC-containing hydroxylases and demethylases catalysing modifications of translational and transcriptional machinery. The structures reveal that new protein hydroxylation activities can evolve by changing the coordination position from which the iron-bound substrate-oxidizing species reacts. This coordination flexibility has probably contributed to the evolution of the wide range of reactions catalysed by oxygenases.


Asunto(s)
Eucariontes/enzimología , Modelos Moleculares , Oxigenasas/química , Células Procariotas/enzimología , Ribosomas/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Eucariontes/clasificación , Humanos , Oxigenasas/metabolismo , Filogenia , Células Procariotas/clasificación , Pliegue de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
2.
Proc Natl Acad Sci U S A ; 108(19): 7745-50, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21508329

RESUMEN

Endoplasmatic reticulum aminopeptidase 1 (ERAP1) is a multifunctional enzyme involved in trimming of peptides to an optimal length for presentation by major histocompatibility complex (MHC) class I molecules. Polymorphisms in ERAP1 have been associated with chronic inflammatory diseases, including ankylosing spondylitis (AS) and psoriasis, and subsequent in vitro enzyme studies suggest distinct catalytic properties of ERAP1 variants. To understand structure-activity relationships of this enzyme we determined crystal structures in open and closed states of human ERAP1, which provide the first snapshots along a catalytic path. ERAP1 is a zinc-metallopeptidase with typical H-E-X-X-H-(X)(18)-E zinc binding and G-A-M-E-N motifs characteristic for members of the gluzincin protease family. The structures reveal extensive domain movements, including an active site closure as well as three different open conformations, thus providing insights into the catalytic cycle. A K(528)R mutant strongly associated with AS in GWAS studies shows significantly altered peptide processing characteristics, which are possibly related to impaired interdomain interactions.


Asunto(s)
Aminopeptidasas/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Presentación de Antígeno , Dominio Catalítico/genética , Cristalografía por Rayos X , Antígeno HLA-B27/metabolismo , Humanos , Antígenos de Histocompatibilidad Menor , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Polimorfismo de Nucleótido Simple , Conformación Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espondilitis Anquilosante/enzimología , Espondilitis Anquilosante/genética
3.
J Biol Chem ; 287(3): 2119-29, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123821

RESUMEN

Biosynthesis of UDP-glucuronic acid by UDP-glucose 6-dehydrogenase (UGDH) occurs through the four-electron oxidation of the UDP-glucose C6 primary alcohol in two NAD(+)-dependent steps. The catalytic reaction of UGDH is thought to involve a Cys nucleophile that promotes formation of a thiohemiacetal enzyme intermediate in the course of the first oxidation step. The thiohemiacetal undergoes further oxidation into a thioester, and hydrolysis of the thioester completes the catalytic cycle. Herein we present crystallographic and kinetic evidence for the human form of UGDH that clarifies participation of covalent catalysis in the enzymatic mechanism. Substitution of the putative catalytic base for water attack on the thioester (Glu(161)) by an incompetent analog (Gln(161)) gave a UGDH variant (E161Q) in which the hydrolysis step had become completely rate-limiting so that a thioester enzyme intermediate accumulated at steady state. By crystallizing E161Q in the presence of 5 mm UDP-glucose and 2 mm NAD(+), we succeeded in trapping a thiohemiacetal enzyme intermediate and determined its structure at 2.3 Å resolution. Cys(276) was covalently modified in the structure, establishing its role as catalytic nucleophile of the reaction. The thiohemiacetal reactive C6 was in a position suitable to become further oxidized by hydride transfer to NAD(+). The proposed catalytic mechanism of human UGDH involves Lys(220) as general base for UDP-glucose alcohol oxidation and for oxyanion stabilization during formation and breakdown of the thiohemiacetal and thioester enzyme intermediates. Water coordinated to Asp(280) deprotonates Cys(276) to function as an aldehyde trap and also provides oxyanion stabilization. Glu(161) is the Brønsted base catalytically promoting the thioester hydrolysis.


Asunto(s)
NAD/química , Uridina Difosfato Glucosa Deshidrogenasa/química , Uridina Difosfato Glucosa/química , Sustitución de Aminoácidos , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Cinética , Mutación Missense , NAD/genética , NAD/metabolismo , Oxidación-Reducción , Relación Estructura-Actividad , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/genética , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo
4.
J Biol Chem ; 287(37): 31349-58, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22810237

RESUMEN

UDP-xylose synthase (UXS) catalyzes decarboxylation of UDP-D-glucuronic acid to UDP-xylose. In mammals, UDP-xylose serves to initiate glycosaminoglycan synthesis on the protein core of extracellular matrix proteoglycans. Lack of UXS activity leads to a defective extracellular matrix, resulting in strong interference with cell signaling pathways. We present comprehensive structural and mechanistic characterization of the human form of UXS. The 1.26-Å crystal structure of the enzyme bound with NAD(+) and UDP reveals a homodimeric short-chain dehydrogenase/reductase (SDR), belonging to the NDP-sugar epimerases/dehydratases subclass. We show that enzymatic reaction proceeds in three chemical steps via UDP-4-keto-D-glucuronic acid and UDP-4-keto-pentose intermediates. Molecular dynamics simulations reveal that the D-glucuronyl ring accommodated by UXS features a marked (4)C(1) chair to B(O,3) boat distortion that facilitates catalysis in two different ways. It promotes oxidation at C(4) (step 1) by aligning the enzymatic base Tyr(147) with the reactive substrate hydroxyl and it brings the carboxylate group at C(5) into an almost fully axial position, ideal for decarboxylation of UDP-4-keto-D-glucuronic acid in the second chemical step. The protonated side chain of Tyr(147) stabilizes the enolate of decarboxylated C(4) keto species ((2)H(1) half-chair) that is then protonated from the Si face at C(5), involving water coordinated by Glu(120). Arg(277), which is positioned by a salt-link interaction with Glu(120), closes up the catalytic site and prevents release of the UDP-4-keto-pentose and NADH intermediates. Hydrogenation of the C(4) keto group by NADH, assisted by Tyr(147) as catalytic proton donor, yields UDP-xylose adopting the relaxed (4)C(1) chair conformation (step 3).


Asunto(s)
Carboxiliasas/química , Uridina Difosfato Ácido Glucurónico/química , Carboxiliasas/metabolismo , Catálisis , Cristalografía por Rayos X , Humanos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad
5.
Nature ; 448(7149): 87-91, 2007 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-17589501

RESUMEN

Post-translational histone modification has a fundamental role in chromatin biology and is proposed to constitute a 'histone code' in epigenetic regulation. Differential methylation of histone H3 and H4 lysyl residues regulates processes including heterochromatin formation, X-chromosome inactivation, genome imprinting, DNA repair and transcriptional regulation. The discovery of lysyl demethylases using flavin (amine oxidases) or Fe(II) and 2-oxoglutarate as cofactors (2OG oxygenases) has changed the view of methylation as a stable epigenetic marker. However, little is known about how the demethylases are selective for particular lysyl-containing sequences in specific methylation states, a key to understanding their functions. Here we reveal how human JMJD2A (jumonji domain containing 2A), which is selective towards tri- and dimethylated histone H3 lysyl residues 9 and 36 (H3K9me3/me2 and H3K36me3/me2), discriminates between methylation states and achieves sequence selectivity for H3K9. We report structures of JMJD2A-Ni(II)-Zn(II) inhibitor complexes bound to tri-, di- and monomethyl forms of H3K9 and the trimethyl form of H3K36. The structures reveal a lysyl-binding pocket in which substrates are bound in distinct bent conformations involving the Zn-binding site. We propose a mechanism for achieving methylation state selectivity involving the orientation of the substrate methyl groups towards a ferryl intermediate. The results suggest distinct recognition mechanisms in different demethylase subfamilies and provide a starting point to develop chemical tools for drug discovery and to study and dissect the complexity of reversible histone methylation and its role in chromatin biology.


Asunto(s)
Proteínas de Unión al ADN/química , Histonas/metabolismo , Oxidorreductasas N-Desmetilantes/química , Factores de Transcripción/química , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Unión al ADN/metabolismo , Histona Demetilasas con Dominio de Jumonji , Modelos Moleculares , Oxidorreductasas N-Desmetilantes/metabolismo , Conformación Proteica , Proteínas Recombinantes , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Relación Estructura-Actividad , Especificidad por Sustrato , Factores de Transcripción/metabolismo
6.
J Biol Chem ; 286(27): 23877-87, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21502315

RESUMEN

Elevated production of the matrix glycosaminoglycan hyaluronan is strongly implicated in epithelial tumor progression. Inhibition of synthesis of the hyaluronan precursor UDP-glucuronic acid (UDP-GlcUA) therefore presents an emerging target for cancer therapy. Human UDP-glucose 6-dehydrogenase (hUGDH) catalyzes, in two NAD(+)-dependent steps without release of intermediate aldehyde, the biosynthetic oxidation of UDP-glucose (UDP-Glc) to UDP-GlcUA. Here, we present a structural characterization of the hUGDH reaction coordinate using crystal structures of the apoenzyme and ternary complexes of the enzyme bound with UDP-Glc/NADH and UDP-GlcUA/NAD(+). The quaternary structure of hUGDH is a disc-shaped trimer of homodimers whose subunits consist of two discrete α/ß domains with the active site located in the interdomain cleft. Ternary complex formation is accompanied by rigid-body and restrained movement of the N-terminal NAD(+) binding domain, sequestering substrate and coenzyme in their reactive positions through interdomain closure. By alternating between conformations in and out of the active site during domain motion, Tyr(14), Glu(161), and Glu(165) participate in control of coenzyme binding and release during 2-fold oxidation. The proposed mechanism of hUGDH involves formation and breakdown of thiohemiacetal and thioester intermediates whereby Cys(276) functions as the catalytic nucleophile. Stopped-flow kinetic data capture the essential deprotonation of Cys(276) in the course of the first oxidation step, allowing the thiolate side chain to act as a trap of the incipient aldehyde. Because thiohemiacetal intermediate accumulates at steady state under physiological reaction conditions, hUGDH inhibition might best explore ligand binding to the NAD(+) binding domain.


Asunto(s)
Coenzimas/química , NAD/química , Uridina Difosfato Glucosa Deshidrogenasa/química , Catálisis , Coenzimas/metabolismo , Humanos , NAD/metabolismo , Oxidación-Reducción , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo
7.
BMC Struct Biol ; 12: 14, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22720794

RESUMEN

BACKGROUND: Aspartyl aminopeptidase (DNPEP), with specificity towards an acidic amino acid at the N-terminus, is the only mammalian member among the poorly understood M18 peptidases. DNPEP has implicated roles in protein and peptide metabolism, as well as the renin-angiotensin system in blood pressure regulation. Despite previous enzyme and substrate characterization, structural details of DNPEP regarding ligand recognition and catalytic mechanism remain to be delineated. RESULTS: The crystal structure of human DNPEP complexed with zinc and a substrate analogue aspartate-ß-hydroxamate reveals a dodecameric machinery built by domain-swapped dimers, in agreement with electron microscopy data. A structural comparison with bacterial homologues identifies unifying catalytic features among the poorly understood M18 enzymes. The bound ligands in the active site also reveal the coordination mode of the binuclear zinc centre and a substrate specificity pocket for acidic amino acids. CONCLUSIONS: The DNPEP structure provides a molecular framework to understand its catalysis that is mediated by active site loop swapping, a mechanism likely adopted in other M18 and M42 metallopeptidases that form dodecameric complexes as a self-compartmentalization strategy. Small differences in the substrate binding pocket such as shape and positive charges, the latter conferred by a basic lysine residue, further provide the key to distinguishing substrate preference. Together, the structural knowledge will aid in the development of enzyme-/family-specific aminopeptidase inhibitors.


Asunto(s)
Biocatálisis , Glutamil Aminopeptidasa/química , Familia de Multigenes , Secuencia de Aminoácidos , Bacterias/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Metales , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína , Electricidad Estática , Especificidad por Sustrato
8.
Biochem J ; 433(2): 303-11, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21029046

RESUMEN

Human GLRX5 (glutaredoxin 5) is an evolutionarily conserved thiol-disulfide oxidoreductase that has a direct role in the maintenance of normal cytosolic and mitochondrial iron homoeostasis, and its expression affects haem biosynthesis and erythropoiesis. We have crystallized the human GLRX5 bound to two [2Fe-2S] clusters and four GSH molecules. The crystal structure revealed a tetrameric organization with the [2Fe-2S] clusters buried in the interior and shielded from the solvent by the conserved ß1-α2 loop, Phe69 and the GSH molecules. Each [2Fe-2S] cluster is ligated by the N-terminal activesite cysteine (Cys67) thiols contributed by two protomers and two cysteine thiols from two GSH. The two subunits co-ordinating the cluster are in a more extended conformation compared with iron-sulfur-bound human GLRX2, and the intersubunit interactions are more extensive and involve conserved residues among monothiol GLRXs. Gel-filtration chromatography and analytical ultracentrifugation support a tetrameric organization of holo-GLRX5, whereas the apoprotein is monomeric. MS analyses revealed glutathionylation of the cysteine residues in the absence of the [2Fe-2S] cluster, which would protect them from further oxidation and possibly facilitate cluster transfer/acceptance. Apo-GLRX5 reduced glutathione mixed disulfides with a rate 100 times lower than did GLRX2 and was active as a glutathione-dependent electron donor for mammalian ribonucleotide reductase.


Asunto(s)
Glutarredoxinas/química , Proteínas Hierro-Azufre/química , Multimerización de Proteína , Cristalografía por Rayos X , Disulfuros/química , Disulfuros/metabolismo , Glutarredoxinas/metabolismo , Humanos , Proteínas Hierro-Azufre/metabolismo , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Estructura Cuaternaria de Proteína
9.
J Biol Chem ; 285(24): 18452-63, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20207735

RESUMEN

Mammalian ALDH7A1 is homologous to plant ALDH7B1, an enzyme that protects against various forms of stress, such as salinity, dehydration, and osmotic stress. It is known that mutations in the human ALDH7A1 gene cause pyridoxine-dependent and folic acid-responsive seizures. Herein, we show for the first time that human ALDH7A1 protects against hyperosmotic stress by generating osmolytes and metabolizing toxic aldehydes. Human ALDH7A1 expression in Chinese hamster ovary cells attenuated osmotic stress-induced apoptosis caused by increased extracellular concentrations of sucrose or sodium chloride. Purified recombinant ALDH7A1 efficiently metabolized a number of aldehyde substrates, including the osmolyte precursor, betaine aldehyde, lipid peroxidation-derived aldehydes, and the intermediate lysine degradation product, alpha-aminoadipic semialdehyde. The crystal structure for ALDH7A1 supports the enzyme's substrate specificities. Tissue distribution studies in mice showed the highest expression of ALDH7A1 protein in liver, kidney, and brain, followed by pancreas and testes. ALDH7A1 protein was found in the cytosol, nucleus, and mitochondria, making it unique among the aldehyde dehydrogenase enzymes. Analysis of human and mouse cDNA sequences revealed mitochondrial and cytosolic transcripts that are differentially expressed in a tissue-specific manner in mice. In conclusion, ALDH7A1 is a novel aldehyde dehydrogenase expressed in multiple subcellular compartments that protects against hyperosmotic stress by generating osmolytes and metabolizing toxic aldehydes.


Asunto(s)
Aldehído Deshidrogenasa/fisiología , Ósmosis , Proteínas/fisiología , Aldehído Deshidrogenasa/metabolismo , Aldehídos/química , Animales , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X/métodos , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Ratones , Proteínas/metabolismo , Distribución Tisular
10.
Biochem Biophys Res Commun ; 408(4): 553-8, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21530488

RESUMEN

Phytanoyl-CoA hydroxylase (PAHX) catalyzes an important step in the metabolism of the fatty acid side chain of chlorophyll. PHYHD1 exists in three isoforms and is the closest human homologue of PAHX. We show that like PAHX, the PHYHD1A but likely not the PHYHD1B/C isoforms, is a functional Fe(II) and 2-oxoglutarate (2OG) dependent oxygenase. Crystallographic and biochemical analyses reveal that PHYHD1A has the double-stranded ß-helix fold and Fe(II) and cosubstrate binding residues characteristic of the 2-oxoglutarate dependent oxygenases and catalyzes the conversion of 2-oxoglutarate to succinate and CO(2) in an iron-dependent manner. However, PHYHD1A did not couple 2OG turnover to the hydroxylation of acyl-coenzyme A derivatives that are substrates for PAHX, implying that it is not directly involved in phytanoyl coenzyme-A metabolism.


Asunto(s)
Oxigenasas de Función Mixta/química , Oxigenasas/química , Cristalografía por Rayos X , Humanos , Hierro/química , Oxigenasas de Función Mixta/genética , Oxigenasas/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína
11.
Arch Biochem Biophys ; 513(1): 19-26, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21689631

RESUMEN

Fungal methionine synthase, Met6p, transfers a methyl group from 5-methyl-tetrahydrofolate to homocysteine to generate methionine. The enzyme is essential to fungal growth and is a potential anti-fungal drug design target. We have characterized the enzyme from the pathogen Candida albicans but were unable to crystallize it in native form. We converted Lys103, Lys104, and Glu107 all to Tyr (Met6pY), Thr (Met6pT) and Ala (Met6pA). All variants showed wild-type kinetic activity and formed useful crystals, each with unique crystal packing. In each case the mutated residues participated in beneficial crystal contacts. We have solved the three structures at 2.0-2.8Å resolution and analyzed crystal packing, active-site residues, and similarity to other known methionine synthase structures. C. albicans Met6p has a two domain structure with each of the domains having a (ßα)(8)-barrel fold. The barrels are arranged face-to-face and the active site is located in a cleft between the two domains. Met6p utilizes a zinc ion for catalysis that is bound in the C-terminal domain and ligated by four conserved residues: His657, Cys659, Glu679 and Cys739.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/química , Candida albicans/enzimología , Proteínas Fúngicas/química , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Sustitución de Aminoácidos , Candida albicans/genética , Catálisis , Cristalografía por Rayos X , Proteínas Fúngicas/genética , Mutación Missense , Estructura Terciaria de Proteína , Relación Estructura-Actividad
12.
J Inherit Metab Dis ; 34(3): 671-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21445611

RESUMEN

Fumarate hydratase catalyzes the stereospecific hydration across the olefinic double bond in fumarate leading to L-malate. The enzyme is expressed in mitochondrial and cytosolic compartments, and participates in the Krebs cycle in mitochondria, as well as in regulation of cytosolic fumarate levels. Fumarate hydratase deficiency is an autosomal recessive trait presenting as metabolic disorder with severe encephalopathy, seizures and poor neurological outcome. Heterozygous mutations are associated with a predisposition to cutaneous and uterine leiomyomas and to renal cancer. The crystal structure of human fumarate hydratase shows that mutations can be grouped into two distinct classes either affecting structural integrity of the core enzyme architecture, or are localized around the enzyme active site. An interactive version of this manuscript (which may contain additional mutations appended after acceptance of this manuscript) may be found on the SSIEM website at: http://www.ssiem.org/resources/structures/FH .


Asunto(s)
Fumarato Hidratasa/química , Fumarato Hidratasa/genética , Errores Innatos del Metabolismo/etiología , Dominio Catalítico/genética , Cristalografía por Rayos X , Fumarato Hidratasa/deficiencia , Humanos , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/etiología , Enfermedades Mitocondriales/genética , Modelos Moleculares , Proteínas Mutantes/química , Mutación/fisiología , Conformación Proteica , Pliegue de Proteína , Relación Estructura-Actividad
13.
Biochem Soc Trans ; 38(5): 1378-85, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20863317

RESUMEN

Biosynthesis of the glycosaminoglycan precursor UDP-α-D-glucuronic acid occurs through a 2-fold oxidation of UDP-α-D-glucose that is catalysed by UGDH (UDP-α-D-glucose 6-dehydrogenase). Structure-function relationships for UGDH and proposals for the enzymatic reaction mechanism are reviewed in the present paper, and structure-based sequence comparison is used for subclassification of UGDH family members. The eukaryotic group of enzymes (UGDH-II) utilize an extended C-terminal domain for the formation of complex homohexameric assemblies. The comparably simpler oligomerization behaviour of the prokaryotic group of enzymes (UGDH-I), in which dimeric forms prevail, is traced back to the lack of relevant intersubunit contacts and trimmings within the C-terminal region. The active site of UGDH contains a highly conserved cysteine residue, which plays a key role in covalent catalysis. Elevated glycosaminoglycan formation is implicated in a variety of human diseases, including the progression of tumours. The inhibition of synthesis of UDP-α-D-glucuronic acid using UGDH antagonists might therefore be a useful strategy for therapy.


Asunto(s)
Uridina Difosfato Glucosa Deshidrogenasa/química , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo , Animales , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ácido Hialurónico/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/antagonistas & inhibidores , Uridina Difosfato Glucosa Deshidrogenasa/genética , Uridina Difosfato Ácido Glucurónico/metabolismo
14.
Mol Cell Endocrinol ; 301(1-2): 199-204, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19013211

RESUMEN

Two members of the human aldo-keto reductase (AKR) superfamily participate in the biosynthesis of bile acids by catalyzing the NADP(H) dependent reduction of 3-keto groups (AKR1C4) and Delta4 double bonds (AKR1D1) of oxysterol precursors. Structure determination of human AKR1C4 and homology modelling of AKR1D1 followed by docking experiments were used to explore active site geometries. Substrate docking resulted in ligand poses satisfying catalytic constraints, and indicates a critical role for Trp227/230 in positioning the substrate in a catalytically competent orientation. Based on the evidence gathered from our docking experiments and experimental structures, this tryptophan residue emerges as a major determinant governing substrate specificity of a subset of enzymes belonging to the AKR1 subfamily.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Ligandos , Modelos Moleculares , Homología Estructural de Proteína , Relación Estructura-Actividad , Triptófano/metabolismo
15.
ACS Catal ; 9(4): 2962-2968, 2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30984471

RESUMEN

Biosynthesis of 6-deoxy sugars, including l-fucose, involves a mechanistically complex, enzymatic 4,6-dehydration of hexose nucleotide precursors as the first committed step. Here, we determined pre- and postcatalytic complex structures of the human GDP-mannose 4,6-dehydratase at atomic resolution. These structures together with results of molecular dynamics simulation and biochemical characterization of wildtype and mutant enzymes reveal elusive mechanistic details of water elimination from GDP-mannose C5″ and C6″, coupled to NADP-mediated hydride transfer from C4″ to C6″. We show that concerted acid-base catalysis from only two active-site groups, Tyr179 and Glu157, promotes a syn 1,4-elimination from an enol (not an enolate) intermediate. We also show that the overall multistep catalytic reaction involves the fewest position changes of enzyme and substrate groups and that it proceeds under conserved exploitation of the basic (minimal) catalytic machinery of short-chain dehydrogenase/reductases.

16.
J Med Chem ; 51(7): 2187-95, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18327899

RESUMEN

The nitrogen-containing bisphosphonates (N-BPs) are the main drugs currently used to treat diseases characterized by excessive bone resorption. The major molecular target of N-BPs is farnesylpyrophosphate synthase. N-BPs inhibit the enzyme by a mechanism that involves time dependent isomerization of the enzyme. We investigated features of N-BPs that confer maximal slow and tight-binding by quantifying the initial and final K(i)s and calculating the isomerization constant K(isom) for many N-BPs. Disruption of the phosphonate-carbon-phosphonate backbone resulted in loss of potency and reduced K(isom). The lack of a hydroxyl group on the geminal carbon also reduced K(isom). The position of the nitrogen in the side chain was crucial to both K(i) and K(isom). A correlation of K(isom) and also final K(i) with previously published in vivo potency reveals that the isomerization constant ( R = -0.77, p < 0.0001) and the final inhibition of FPPS by N-BPs ( R = 0.74, p < 0.0001) are closely linked to antiresorptive efficacy.


Asunto(s)
Difosfonatos/farmacología , Inhibidores Enzimáticos/farmacología , Geraniltranstransferasa/antagonistas & inhibidores , Nitrógeno/química , Sitios de Unión , Difosfonatos/química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad , Factores de Tiempo
17.
J Med Chem ; 51(1): 1-3, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18062662

RESUMEN

Quinone oxidoreductase 2 (NQO2) binds the prodrug tretazicar (also known as CB1954, 5-(aziridin-1-yl)-2,4-dinitrobenzamide), which exhibits a profound antitumor effect in human cancers when administered together with caricotamide. X-ray structure determination allowed for two possible orientations of the ligand. Here we describe a new NMR method, SALMON (solvent accessibility, ligand binding, and mapping of ligand orientation by NMR spectroscopy), based on waterLOGSY to determine the orientation of a ligand bound to a protein by mapping its solvent accessibility, which was used to unambiguously determine the orientation of CB1954 in NQO2.


Asunto(s)
Antineoplásicos/química , Aziridinas/química , Modelos Moleculares , NAD(P)H Deshidrogenasa (Quinona)/química , Profármacos/química , Agua/química , Sitios de Unión , Ligandos , Espectroscopía de Resonancia Magnética , Unión Proteica , Solventes
18.
Chem Biol ; 14(11): 1243-53, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18022563

RESUMEN

Mammals utilize a single phosphopantetheinyl transferase for the posttranslational modification of at least three different apoproteins: the carrier protein components of cytosolic and mitochondrial fatty acid synthases and the aminoadipate semialdehyde reductase involved in lysine degradation. We determined the crystal structure of the human phosphopantetheinyl transferase, a eukaryotic phosphopantetheinyl transferase characterized, complexed with CoA and Mg(2+), and in ternary complex with CoA and ACP. The involvement of key residues in ligand binding and catalysis was confirmed by mutagenesis and kinetic analysis. Human phosphopantetheinyl transferase exhibits an alpha/beta fold and 2-fold pseudosymmetry similar to the Sfp phosphopantetheinyl transferase from Bacillus subtilis. Although the bound ACP exhibits a typical four-helix structure, its binding is unusual in that it is facilitated predominantly by hydrophobic interactions. A detailed mechanism is proposed describing the substrate binding and catalytic process.


Asunto(s)
Ácido Graso Sintasas/metabolismo , Secuencia de Aminoácidos , Catálisis , Coenzima A/metabolismo , Cristalografía por Rayos X , Ácido Graso Sintasas/química , Ácido Graso Sintasas/genética , Humanos , Cinética , Magnesio/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
19.
Biochem J ; 402(3): 419-27, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17067289

RESUMEN

To this day, a significant proportion of the human genome remains devoid of functional characterization. In this study, we present evidence that the previously functionally uncharacterized product of the human DHRS10 gene is endowed with 17beta-HSD (17beta-hydroxysteroid dehydrogenase) activity. 17beta-HSD enzymes are primarily involved in the metabolism of steroids at the C-17 position and also of other substrates such as fatty acids, prostaglandins and xenobiotics. In vitro, DHRS10 converts NAD+ into NADH in the presence of oestradiol, testosterone and 5-androstene-3beta,17beta-diol. Furthermore, the product of oestradiol oxidation, oestrone, was identified in intact cells transfected with a construct plasmid encoding the DHRS10 protein. In situ fluorescence hybridization studies have revealed the cytoplasmic localization of DHRS10. Along with tissue expression data, this suggests a role for DHRS10 in the local inactivation of steroids in the central nervous system and placenta. The crystal structure of the DHRS10 apoenzyme exhibits secondary structure of the SDR (short-chain dehydrogenase/reductase) family: a Rossmann-fold with variable loops surrounding the active site. It also reveals a broad and deep active site cleft into which NAD+ and oestradiol can be docked in a catalytically competent orientation.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/química , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Citosol/enzimología , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/aislamiento & purificación , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Expresión Génica , Humanos , Cinética , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , NAD/metabolismo , Especificidad de Órganos , Oxidación-Reducción , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Homología Estructural de Proteína
20.
Mol Cell Endocrinol ; 265-266: 71-6, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17234335

RESUMEN

Hydroxysteroid dehydrogenases catalyze the NAD(P)(H)-dependent oxidoreduction of hydroxyl and oxo-functions at distinct positions of steroid hormones. This reversible reaction constitutes an important pre-receptor control mechanism for nuclear receptor ligands of the androgen, estrogen and glucocorticoid classes, since the conversion "switches" between receptor ligands and their inactive metabolites. The major reversible activities found in mammals acting on steroid hormones comprise 3alpha-, 11beta- and 17beta-hydroxysteroid dehydrogenases, and for each group several distinct isozymes have been described. The enzymes differ in their expression pattern, nucleotide cofactor preference, steroid substrate specificity and subcellular localization, and thus constitute a complex system ensuring cell-specific adaptation and regulation of steroid hormone levels. Several isoforms constitute promising drug targets, of particular importance in cancer, metabolic diseases, neurodegeneration and immunity.


Asunto(s)
Hormonas/metabolismo , Hidroxiesteroide Deshidrogenasas/química , Hidroxiesteroide Deshidrogenasas/metabolismo , Esteroides/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA