Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Chem Biol Interact ; 202(1-3): 153-8, 2013 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-23298867

RESUMEN

The aldo-keto reductase superfamily contains 173 proteins which are present in all phyla. Examination of the human and mouse genomes has identified that in some instances a single AKR gene can give rise to alternatively spliced mRNA variants which in some cases can give rise to more than one protein isoform. This is currently well documented in the AKR6A subfamily which contains the ß-subunits of the voltage-gated potassium ion channels. With the emergence of second generation sequencing it is likely that the occurrence of transcript variants and protein isoforms from a single AKR gene may become common place. To deal with this issue we recommend that the Ensembl data-base nomenclature be used to annotate the transcript variants from a single AKR gene. However, since multiple transcript variants could give rise to either the same or multiple protein isoforms from the same AKR gene we also propose to expand the nomenclature of the AKR protein superfamily, so that when a protein isoform is shown to be expressed and is functional it would be assigned the standard AKR name followed by a "period or full-stop" and a number for that unique isoform. Numbers will be assigned chronologically and linked to the respective transcripts annotated in Ensembl e.g. AKR6A5.1 (Kvß2.1) (AKR6A5-001, -006 and -201), followed by AKR6A5.2 (Kvß2.2) (AKR6A5-002,-202). This nomenclature is expandable and it enables multiple protein isoforms to be assigned to their respective transcripts when they arise from the same AKR gene or for a single protein isoform to be assigned to multiple transcripts when the transcripts encode the same AKR protein.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Aldehído Reductasa , Aldo-Ceto Reductasas , Empalme Alternativo , Animales , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Isoformas de Proteínas , ARN Mensajero/genética , Regiones no Traducidas
2.
J Biol Chem ; 287(14): 11213-21, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334702

RESUMEN

Perakine reductase (PR) catalyzes the NADPH-dependent reduction of the aldehyde perakine to yield the alcohol raucaffrinoline in the biosynthetic pathway of ajmaline in Rauvolfia, a key step in indole alkaloid biosynthesis. Sequence alignment shows that PR is the founder of the new AKR13D subfamily and is designated AKR13D1. The x-ray structure of methylated His(6)-PR was solved to 2.31 Å. However, the active site of PR was blocked by the connected parts of the neighbor symmetric molecule in the crystal. To break the interactions and obtain the enzyme-ligand complexes, the A213W mutant was generated. The atomic structure of His(6)-PR-A213W complex with NADPH was determined at 1.77 Å. Overall, PR folds in an unusual α(8)/ß(6) barrel that has not been observed in any other AKR protein to date. NADPH binds in an extended pocket, but the nicotinamide riboside moiety is disordered. Upon NADPH binding, dramatic conformational changes and movements were observed: two additional ß-strands in the C terminus become ordered to form one α-helix, and a movement of up to 24 Å occurs. This conformational change creates a large space that allows the binding of substrates of variable size for PR and enhances the enzyme activity; as a result cooperative kinetics are observed as NADPH is varied. As the founding member of the new AKR13D subfamily, PR also provides a structural template and model of cofactor binding for the AKR13 family.


Asunto(s)
Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , NADP/metabolismo , Cristalografía por Rayos X , Evolución Molecular , Metilación , Modelos Moleculares , NADP/farmacología , Unión Proteica , Conformación Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Rauwolfia/enzimología , Alineación de Secuencia
3.
J Steroid Biochem Mol Biol ; 130(1-2): 7-15, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22265960

RESUMEN

Type 5 17ß-hydroxysteroid dehydrogenase (AKR1C3) is the major enzyme in the prostate that reduces 4-androstene-3,17-dione (Δ(4)-Adione) to the androgen receptor (AR) ligand testosterone. AKR1C3 is upregulated in prostate cancer (PCa) and castrate resistant prostate cancer (CRPC) that develops after androgen deprivation therapy. PCa and CRPC often depend on intratumoral androgen biosynthesis and upregulation of AKR1C3 could contribute to intracellular synthesis of AR ligands and stimulation of proliferation through AR signaling. To test this hypothesis, we developed an LNCaP prostate cancer cell line overexpressing AKR1C3 (LNCaP-AKR1C3) and compared its metabolic and proliferative responses to Δ(4)-Adione treatment with that of the parental, AKR1C3 negative LNCaP cells. In LNCaP and LNCaP-AKR1C3 cells, metabolism proceeded via 5α-reduction to form 5α-androstane-3,17-dione and then (epi)androsterone-3-glucuronide. LNCaP-AKR1C3 cells made significantly higher amounts of testosterone-17ß-glucuronide. When 5α-reductase was inhibited by finasteride, the production of testosterone-17ß-glucuronide was further elevated in LNCaP-AKR1C3 cells. When AKR1C3 activity was inhibited with indomethacin the production of testosterone-17ß-glucuronide was significantly decreased. Δ(4)-Adione treatment stimulated cell proliferation in both cell lines. Finasteride inhibited LNCaP cell proliferation, consistent with 5α-androstane-3,17-dione acting as the major metabolite that stimulates growth by binding to the mutated AR. However, LNCaP-AKR1C3 cells were resistant to the growth inhibitory properties of finasteride, consistent with the diversion of Δ(4)-Adione metabolism from 5α-reduced androgens to increased formation of testosterone. Indomethacin did not result in differences in Δ(4)-Adione induced proliferation since this treatment led to the same metabolic profile in LNCaP and LNCaP-AKR1C3 cells. We conclude that AKR1C3 overexpression diverts androgen metabolism to testosterone that results in proliferation in androgen sensitive prostate cancer. This effect is seen despite high levels of uridine glucuronosyl transferases suggesting that AKR1C3 activity can surmount the effects of this elimination pathway. Treatment options in prostate cancer that target 5α-reductase where AKR1C3 co-exists may be less effective due to the diversion of Δ(4)-Adione to testosterone.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Inhibidores de 5-alfa-Reductasa/farmacología , Andrógenos/metabolismo , Finasterida/farmacología , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Testosterona/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/genética , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Masculino , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo
4.
Mol Cell Endocrinol ; 349(2): 202-13, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22061621

RESUMEN

Hydroxysteroid dehydrogenases (HSDs) are involved in metabolism and pre-receptor regulation of steroid hormones. While 17beta-HSDs and 11beta-HSDs are extensively studied in mammals, only few orthologs are characterized in fish. We discovered a novel zebrafish HSD candidate closely related to 17beta-HSD types 3 and 12, which has orthologs in other species. The enzyme catalyzes the conversion of cortisone to 20beta-hydroxycortisone identified by LC-MS/MS. We named the new enzyme 20beta-HSD type 2. All 20beta-HSD type 2 orthologs localize in the endoplasmic reticulum. Zebrafish 20beta-HSD type 2 is expressed during embryonic development showing the same expression pattern as 11beta-HSD type 2 known to oxidize cortisol to cortisone. In adult tissues 20beta-HSD type 2 shows a ubiquitous expression pattern with some minor sex-specific differences. In contrast to other enzymes metabolizing C21-steroids and being mostly involved in reproduction we propose that novel type 2 20beta-HSDs in teleost fish are important enzymes in cortisol catabolism.


Asunto(s)
Cortisona Reductasa/metabolismo , Proteínas de Peces/metabolismo , Hidrocortisona/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Cortisona/metabolismo , Cortisona Reductasa/genética , Retículo Endoplásmico/metabolismo , Femenino , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica , Estudios de Asociación Genética , Células HEK293 , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido , Transfección , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
6.
Chem Biol Interact ; 191(1-3): 250-4, 2011 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-21185810

RESUMEN

The stereospecific 5ß-reduction of Δ(4)-3-ketosterols is very difficult to achieve chemically and introduces a 90° bend between ring A and B of the planar steroid. In mammals, the reaction is catalyzed by steroid 5ß-reductase, a member of the aldo-keto reductase (AKR) family. The human enzyme, AKR1D1, plays an essential role in bile-acid biosynthesis since the 5ß-configuration is required for the emulsifying properties of bile. Deficient 5ß-reductase activity can lead to cholestasis and neo-natal liver failure and is often lethal if it remains untreated. In five patients with 5ß-reductase deficiency, sequencing revealed individual, non-synonymous point mutations in the AKR1D1 gene: L106F, P133R, G223E, P198L and R261C. However, mapping these mutations to the AKR1D1 crystal structure failed to reveal any obvious involvement in substrate or cofactor binding or catalytic mechanism, and it remained unclear whether these mutations could be causal for the observed disease. We analyzed the positions of the reported mutations and found that they reside in highly conserved portions of AKR1D1 and hypothesized that they would likely lead to changes in protein folding, and hence enzyme activity. Attempts to purify the mutant enzymes for further characterization by over-expression in Escherichia coli yielded sufficient amounts of only one mutant (P133R). This enzyme exhibited reduced K(m) and k(cat) values with the bile acid intermediate Δ(4)-cholesten-7α-ol-3-one as substrate reminiscent of uncompetitive inhibition. In addition, P133R displayed no change in cofactor affinity but was more thermolabile as judged by CD-spectroscopy. When all AKR1D1 mutants were expressed in HEK 293 cells, protein expression levels and enzyme activity were dramatically reduced. Furthermore, cycloheximide treatment revealed decreased stability of several of the mutants compared to wild type. Our data show, that all five mutations identified in patients with functional bile acid deficiency strongly affected AKR1D1 enzyme functionality and therefore may be causal for this disease.


Asunto(s)
Enfermedad/genética , Oxidorreductasas/genética , Mutación Puntual , Animales , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/deficiencia , Secuencia Conservada , Células HEK293 , Humanos , Oxidorreductasas/química , Oxidorreductasas/metabolismo
7.
J Biol Chem ; 285(32): 24529-37, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20522910

RESUMEN

Bile acid deficiency is a serious syndrome in newborns that can result in death if untreated. 5beta-Reductase deficiency is one form of bile acid deficiency and is characterized by dramatically decreased levels of physiologically active 5beta-reduced bile acids. AKR1D1 (aldo-keto reductase 1D1) is the only known human enzyme that stereo-specifically reduces the Delta(4) double bond in 3-keto steroids and sterols to yield the 5beta-hydrogenated product. Analysis of the AKR1D1 gene in five patients with 5beta-reductase deficiency revealed five different mutations resulting in an amino acid substitution in the protein. To investigate a causal role for these observed point mutations in AKR1D1 in 5beta-reductase deficiency, we characterized their effect on enzymatic properties. Attempts to purify mutant enzymes by overexpression in Escherichia coli only yielded sufficient amounts of the P133R mutant for further characterization. This enzyme displayed a highly reduced K(m) and V(max) reminiscent of uncompetitive kinetics with 4-cholesten-7alpha-ol-3-one as substrate. In addition, this mutant displayed no change in cofactor affinity but was more thermolabile in the absence of NADPH as judged by CD spectroscopy. All mutants were compared following expression in HEK 293 cells. Although these enzymes were equally expressed based on mRNA levels, protein expression and functional activity were dramatically reduced. Cycloheximide treatment also revealed that several of the expressed mutants were less stable. Our findings show that the reported mutations in AKR1D1 in patients with 5beta-reductase lead to significantly decreased levels of active enzyme and could be causal in the development of bile acid deficiency syndrome.


Asunto(s)
Ácidos y Sales Biliares/deficiencia , Oxidorreductasas/genética , Línea Celular , Dicroismo Circular , Cicloheximida/farmacología , Vectores Genéticos , Humanos , Cinética , Modelos Moleculares , Conformación Molecular , Mutación , NADP/química , Oxidorreductasas/fisiología , Mutación Puntual , Inhibidores de la Síntesis de la Proteína/farmacología , Espectrometría de Fluorescencia/métodos
8.
J Mol Biol ; 399(2): 255-67, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20382160

RESUMEN

Steroids and retinoids are signaling molecules that control a variety of physiological processes. 17beta-Hydroxysteroid dehydrogenase type 1 (17beta-HSD1) catalyzes the reduction of estrone to estradiol, supplying biologically active estrogen-regulating sex-specific differentiation. Photoreceptor-associated retinol dehydrogenase (prRDH) is evolutionarily closely related to 17beta-HSD1 but reduces all-trans retinal to all-trans retinol, contributing to rhodopsin regeneration in the visual cycle. Sequence alignment revealed a new enzyme-specific conserved amino acid close to the active site: methionine (position 144 in human enzyme) in prRDH and glycine (position 145) in 17beta-HSD1. We investigated the role of this residue in substrate discrimination in human and zebrafish enzymes. Both recombinant enzymes were expressed in HEK 293 cells followed by normalization of expression by semiquantitative Western blots. Changing of the prRDH-specific methionine to glycine resulted in a gain of function: the mutants now catalyzed the reduction of estrone and all-trans retinal. Human and zebrafish wild-type 17beta-HSD1s efficiently catalyzed the reduction of all-trans retinal to its alcohol. Exchange of glycine for methionine increased the catalytic activity of 17beta-HSD1 toward all-trans retinal in zebrafish but not in the human enzyme, in which the opposite effect was observed. Molecular modeling showed that the zebrafish 17beta-HSD1 substrate-binding pocket is similar to that of prRDH and methionine insertion benefits all-trans retinal reduction. In contrast, in human 17beta-HSD1, the insertion of the bulky methionine causes a disruption of substrate-binding site. We demonstrate for the first time the role of a single amino acid in the evolution of these functionally diverse enzymes and suggest new physiological functions for 17beta-HSD1 in retinoid metabolism. This has implications for the validation of inhibitors of 17beta-HSD1 developed for cancer treatment.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Retinoides/metabolismo , Esteroides/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/genética , Oxidorreductasas de Alcohol/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Western Blotting , Dominio Catalítico , Línea Celular , Expresión Génica , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/metabolismo , Alineación de Secuencia , Especificidad por Sustrato , Pez Cebra
9.
Hum Genomics ; 3(4): 362-70, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19706366

RESUMEN

Aldo-keto reductases (AKRs) are phase I metabolising enzymes that catalyse the reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H)-dependent reduction of carbonyl groups to yield primary and secondary alcohols on a wide range of substrates, including aliphatic and aromatic aldehydes and ketones, ketoprostaglandins, ketosteroids and xenobiotics. In so doing they functionalise the carbonyl group for conjugation (phase II enzyme reactions). Although functionally diverse, AKRs form a protein superfamily based on their high sequence identity and common protein fold, the (alpha/beta) 8 -barrel structure. Well over 150 AKR enzymes, from diverse organisms, have been annotated so far and given systematic names according to a nomenclature that is based on multiple protein sequence alignment and degree of identity. Annotation of non-vertebrate AKRs at the National Center for Biotechnology Information or Vertebrate Genome Annotation (vega) database does not often include the systematic nomenclature name, so the most comprehensive overview of all annotated AKRs is found on the AKR website (http://www.med.upenn.edu/akr/). This site also hosts links to more detailed and specialised information (eg on crystal structures, gene expression and single nucleotide polymorphisms [SNPs]). The protein-based AKR nomenclature allows unambiguous identification of a given enzyme but does not reflect the wealth of genomic and transcriptomic variation that exists in the various databases. In this context, identification of putative new AKRs and their distinction from pseudogenes are challenging. This review provides a short summary of the characteristic features of AKR biochemistry and structure that have been reviewed in great detail elsewhere, and focuses mainly on nomenclature and database entries of human AKRs that so far have not been subject to systematic annotation. Recent developments in the annotation of SNP and transcript variance in AKRs are also summarised.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Genómica , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , Aldehído Reductasa , Aldo-Ceto Reductasas , Bases de Datos Genéticas , Conformación Proteica , Especificidad por Sustrato
10.
Chem Biol Interact ; 178(1-3): 197-205, 2009 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-19026618

RESUMEN

Hydroxysteroid dehydrogenase like 1 protein (HSDL1) is an uncharacterized member of short-chain dehydrogenase/reductase (SDR) protein family. In search for functional assignment of both human and zebrafish HSDL1 we characterized the subcellular localization as well as the tissue distribution and performed a screen for putative substrates of HSDL1 enzymes. Surprisingly, human HSDL1 shows exchange of an amino acid in the active center (Sx(12)FSxxK instead of Sx(12)YSxxK) that is considered critical for catalysis. Native human HSDL1 expressed in cells did not show enzymatic activity with any of the substrates tested. Expression of the point mutation F218Y HSDL1 though, resulted in the detection of weak dehydrogenase activity towards steroid and retinoid substrates. The role of this inactivating mutation is uncertain but was found to be conserved in many other vertebrate species, including zebrafish. Identification of protein interaction partners by yeast two-hybrid system suggests that despite the potential lack of enzymatic activity HSDL1 might retain regulatory functions in the cell.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Dominio Catalítico , Secuencia Conservada , Cartilla de ADN , Humanos , Hidroxiesteroide Deshidrogenasas/química , Hidroxiesteroide Deshidrogenasas/genética , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Biosíntesis de Proteínas , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Especificidad por Sustrato , Técnicas del Sistema de Dos Híbridos
11.
Chem Biol Interact ; 178(1-3): 94-8, 2009 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-19027726

RESUMEN

Short-chain dehydrogenases/reductases (SDR) constitute one of the largest enzyme superfamilies with presently over 46,000 members. In phylogenetic comparisons, members of this superfamily show early divergence where the majority have only low pairwise sequence identity, although sharing common structural properties. The SDR enzymes are present in virtually all genomes investigated, and in humans over 70 SDR genes have been identified. In humans, these enzymes are involved in the metabolism of a large variety of compounds, including steroid hormones, prostaglandins, retinoids, lipids and xenobiotics. It is now clear that SDRs represent one of the oldest protein families and contribute to essential functions and interactions of all forms of life. As this field continues to grow rapidly, a systematic nomenclature is essential for future annotation and reference purposes. A functional subdivision of the SDR superfamily into at least 200 SDR families based upon hidden Markov models forms a suitable foundation for such a nomenclature system, which we present in this paper using human SDRs as examples.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Terminología como Asunto , Internet , Cadenas de Markov
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...