Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Arch Biochem Biophys ; 681: 108256, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31923393

RESUMO

Aldehyde dehydrogenases catalyze the NAD(P)+-dependent oxidation of aldehydes to their corresponding carboxylic acids. The three-dimensional structures of the human ALDH1A enzymes were recently obtained, while a complete kinetic characterization of them, under the same experimental conditions, is lacking. We show that the three enzymes, ALDH1A1, ALDH1A2 and ALDH1A3, have similar topologies, although with decreasing volumes in their substrate-binding pockets. The activity with aliphatic and retinoid aldehydes was characterized side-by-side, using an improved HPLC-based method for retinaldehyde. Hexanal was the most efficient substrate. ALDH1A1 displayed lower Km values with hexanal, trans-2-hexenal and citral, compared to ALDH1A2 and ALDH1A3. ALDH1A2 was the best enzyme for the lipid peroxidation product, 4-hydroxy-2-nonenal, in terms of kcat/Km. The catalytic efficiency towards all-trans and 9-cis-retinaldehyde was in general lower than for alkanals and alkenals. ALDH1A2 and ALDH1A3 showed higher catalytic efficiency for all-trans-retinaldehyde. The lower specificity of ALDH1A3 for 9-cis-retinaldehyde against the all-trans- isomer might be related to the smaller volume of its substrate-binding pocket. Magnesium inhibited ALDH1A1 and ALDH1A2, while it activated ALDH1A3, which is consistent with cofactor dissociation being the rate-limiting step for ALDH1A1 and ALDH1A2, and deacylation for ALDH1A3, with hexanal as a substrate. We mutated both ALDH1A1 (L114P) and ALDH1A2 (N475G, A476V, L477V, N478S) to mimic their counterpart substrate-binding pockets. ALDH1A1 specificity for citral was traced to residue 114 and to residues 458 to 461. Regarding retinaldehyde, the mutants did not show significant differences with their respective wild-type forms, suggesting that the mutated residues are not critical for retinoid specificity.


Assuntos
Família Aldeído Desidrogenase 1/metabolismo , Aldeído Oxirredutases/metabolismo , Células-Tronco Neoplásicas/metabolismo , Retinal Desidrogenase/metabolismo , Tretinoína/metabolismo , Humanos , Magnésio/metabolismo , Modelos Moleculares , Células-Tronco Neoplásicas/patologia , Retinaldeído/metabolismo , Especificidade por Substrato
2.
Chem Biol Interact ; 307: 186-194, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31028727

RESUMO

The aldo-keto reductase (AKR) superfamily comprises NAD(P)H-dependent enzymes that catalyze the reduction of a variety of carbonyl compounds. AKRs are classified in families and subfamilies. Humans exhibit three members of the AKR1B subfamily: AKR1B1 (aldose reductase, participates in diabetes complications), AKR1B10 (overexpressed in several cancer types), and the recently described AKR1B15. AKR1B10 and AKR1B15 share 92% sequence identity, as well as the capability of being active towards retinaldehyde. However, AKR1B10 and AKR1B15 exhibit strong differences in substrate specificity and inhibitor selectivity. Remarkably, their substrate-binding sites are the most divergent parts between them. Out of 27 residue substitutions, six are changes to Phe residues in AKR1B15. To investigate the participation of these structural changes, especially the Phe substitutions, in the functional features of each enzyme, we prepared two AKR1B10 mutants. The AKR1B10 m mutant carries a segment of six AKR1B15 residues (299-304, including three Phe residues) in the respective AKR1B10 region. An additional substitution (Val48Phe) was incorporated in the second mutant, AKR1B10mF48. This resulted in structures with smaller and more hydrophobic binding pockets, more similar to that of AKR1B15. In general, the AKR1B10 mutants mirrored well the specific functional features of AKR1B15, i.e., the different preferences towards the retinaldehyde isomers, the much higher activity with steroids and ketones, and the unique behavior with inhibitors. It can be concluded that the Phe residues of loop C (299-304) contouring the substrate-binding site, in addition to Phe at position 48, strongly contribute to a narrower and more hydrophobic site in AKR1B15, which would account for its functional uniqueness. In addition, we have investigated the AKR1B10 and AKR1B15 activity toward steroids. While AKR1B10 only exhibits residual activity, AKR1B15 is an efficient 17-ketosteroid reductase. Finally, the functional role of AKR1B15 in steroid and retinaldehyde metabolism is discussed.


Assuntos
Aldo-Ceto Redutases/metabolismo , Engenharia de Proteínas , Retinoides/metabolismo , Esteroides/metabolismo , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Isomerismo , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Retinaldeído/química , Retinaldeído/metabolismo , Retinoides/química , Alinhamento de Sequência , Esteroides/química , Especificidade por Substrato
3.
Eur J Med Chem ; 152: 160-174, 2018 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-29705708

RESUMO

Human aldose reductase (AKR1B1, AR) is a key enzyme of the polyol pathway, catalyzing the reduction of glucose to sorbitol at high glucose concentrations, as those found in diabetic condition. Indeed, AKR1B1 overexpression is related to diabetes secondary complications and, in some cases, with cancer. For many years, research has been focused on finding new AKR1B1 inhibitors (ARIs) to overcome these diseases. Despite the efforts, most of the new drug candidates failed because of their poor pharmacokinetic properties and/or unacceptable side effects. Here we report the synthesis of a series of 1-oxopyrimido[4,5-c]quinoline-2-acetic acid derivatives as novel ARIs. IC50 assays and X-ray crystallographic studies proved that these compounds are promising hits for further drug development, with high potency and selectivity against AKR1B1. Based on the determined X-ray structures with hit-to-lead compounds, we designed and synthesized a second series that yielded lead compound 68 (Kiappvs. AKR1B1 = 73 nM). These compounds are related to the previously reported 2-aminopyrimido[4,5-c]quinolin-1(2H)-ones, which exhibit antimitotic activity. Regardless of their similarity, the 2-amino compounds are unable to inhibit AKR1B1 while the 2-acetic acid derivatives are not cytotoxic against fibrosarcoma HT-1080 cells. Thus, the replacement of the amino group by an acetic acid moiety changes their biological activity, improving their potency as ARIs.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Quinolinas/farmacologia , Aldeído Redutase/metabolismo , Aldo-Ceto Redutases , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estrutura Molecular , Quinolinas/síntese química , Quinolinas/química , Relação Estrutura-Atividade
4.
Chem Biol Interact ; 276: 182-193, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28322781

RESUMO

Aldo-keto reductases (AKRs) are distributed in three families and multiple subfamilies in mammals. The mouse Akr1b3 gene is clearly orthologous to human AKR1B1, both coding for aldose reductase, and their gene products show similar tissue distribution, regulation by osmotic stress and kinetic properties. In contrast, no unambiguous orthologs of human AKR1B10 and AKR1B15.1 have been identified in rodents. Although two more AKRs, AKR1B7 and AKR1B8, have been identified and characterized in mouse, none of them seems to exhibit properties similar to the human AKRs. Recently, a novel mouse AKR gene, Akr1b16, was annotated and the respective gene product, AKR1B16 (sharing 83% and 80% amino acid sequence identity with AKR1B10 and AKR1B15.1, respectively), was expressed as insoluble and inactive protein in a bacterial expression system. Here we describe the expression and purification of a soluble and enzymatically active AKR1B16 from E. coli using three chaperone systems. A structural model of AKR1B16 allowed the estimation of its active-site pocket volume, which was much wider (402 Å3) than those of AKR1B10 (279 Å3) and AKR1B15.1 (60 Å3). AKR1B16 reduced aliphatic and aromatic carbonyl compounds, using NADPH as a cofactor, with moderate or low activity (highest kcat values around 5 min-1). The best substrate for the enzyme was pyridine-3-aldehyde. AKR1B16 showed poor inhibition with classical AKR inhibitors, tolrestat being the most potent. Kinetics and inhibition properties resemble those of rat AKR1B17 but differ from those of the human enzymes. In addition, AKR1B16 catalyzed the oxidation of 17ß-hydroxysteroids in a NADP+-dependent manner. These results, together with a phylogenetic analysis, suggest that mouse AKR1B16 is an ortholog of rat AKR1B17, but not of human AKR1B10 or AKR1B15.1. These human enzymes have no counterpart in the murine species, which is evidenced by forming a separate cluster in the phylogenetic tree and by their unique activity with retinaldehyde.


Assuntos
Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/metabolismo , Aldo-Ceto Redutases/química , Aldo-Ceto Redutases/metabolismo , Sequência de Aminoácidos , Animais , Biocatálise , Células HeLa , Humanos , Cinética , Camundongos , NADP/química , NADP/metabolismo , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/classificação , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/genética , Filogenia , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Retinaldeído/química , Retinaldeído/metabolismo , Alinhamento de Sequência , Espectrofotometria , Esteroides/química , Esteroides/metabolismo , Especificidade por Substrato
5.
Chem Biol Interact ; 276: 174-181, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28161411

RESUMO

UVI2008, a retinoic acid receptor (RAR) ß/γ agonist originated from C3 bromine addition to the parent RAR pan-agonist 4-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propenyl]benzoic acid (TTNPB), is also a selective inhibitor of aldo-keto reductase family member 1B10 (AKR1B10). Thus, it might become a lead drug for the design of compounds targeting both activities, as an AKR1B10 inhibitor and RAR agonist, which could constitute a novel therapeutic approach against cancer and skin-related diseases. Herein, the X-ray structure of the methylated Lys125Arg/Val301Leu AKR1B10 (i.e. AKME2MU) holoenzyme in complex with UVI2008 was determined at 1.5 Å resolution, providing an explanation for UVI2008 selectivity against AKR1B10 (IC50 = 6.1 µM) over the closely related aldose reductase (AR, IC50 = 70 µM). The carboxylic acid group of UVI2008 is located in the anion-binding pocket, at hydrogen-bond distance of catalytically important residues Tyr49 and His111. The inhibitor bromine atom can only fit in the wider active site of AKR1B10, mainly because of the native Trp112 side-chain orientation, not possible in AR. In AKR1B10, Trp112 native conformation, and thus UVI2008 binding, is facilitated through interaction with Gln114. IC50 analysis of the corresponding Thr113Gln mutant in AR confirmed this hypothesis. The elucidation of the binding mode of UVI2008 to AKR1B10, along with the previous studies on the retinoid specificity of AKR1B10 and on the stilbene retinoid scaffold conforming UVI2008, could indeed be used to foster the drug design of bifunctional antiproliferative compounds.


Assuntos
Aldeído Redutase/metabolismo , Benzoatos/química , Clorobenzoatos/metabolismo , Inibidores Enzimáticos/metabolismo , Retinoides/química , Tetra-Hidronaftalenos/metabolismo , Aldeído Redutase/antagonistas & inibidores , Aldo-Ceto Redutases , Benzoatos/metabolismo , Sítios de Ligação , Domínio Catalítico , Clorobenzoatos/química , Desenho de Fármacos , Inibidores Enzimáticos/química , Halogenação , Simulação de Acoplamento Molecular , Receptores do Ácido Retinoico/agonistas , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Retinoides/metabolismo , Relação Estrutura-Atividade , Tetra-Hidronaftalenos/química
6.
ChemMedChem ; 10(12): 1989-2003, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26549844

RESUMO

The human enzymes aldose reductase (AR) and AKR1B10 have been thoroughly explored in terms of their roles in diabetes, inflammatory disorders, and cancer. In this study we identified two new lead compounds, 2-(3-(4-chloro-3-nitrobenzyl)-2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)acetic acid (JF0048, 3) and 2-(2,4-dioxo-3-(2,3,4,5-tetrabromo-6-methoxybenzyl)-3,4-dihydropyrimidin-1(2H)-yl)acetic acid (JF0049, 4), which selectively target these enzymes. Although 3 and 4 share the 3-benzyluracil-1-acetic acid scaffold, they have different substituents in their aryl moieties. Inhibition studies along with thermodynamic and structural characterizations of both enzymes revealed that the chloronitrobenzyl moiety of compound 3 can open the AR specificity pocket but not that of the AKR1B10 cognate. In contrast, the larger atoms at the ortho and/or meta positions of compound 4 prevent the AR specificity pocket from opening due to steric hindrance and provide a tighter fit to the AKR1B10 inhibitor binding pocket, probably enhanced by the displacement of a disordered water molecule trapped in a hydrophobic subpocket, creating an enthalpic signature. Furthermore, this selectivity also occurs in the cell, which enables the development of a more efficient drug design strategy: compound 3 prevents sorbitol accumulation in human retinal ARPE-19 cells, whereas 4 stops proliferation in human lung cancer NCI-H460 cells.


Assuntos
Acetatos/química , Aldeído Redutase/antagonistas & inibidores , Inibidores Enzimáticos/química , Uracila/análogos & derivados , Acetatos/metabolismo , Acetatos/farmacologia , Aldeído Redutase/metabolismo , Aldo-Ceto Redutases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Ligação de Hidrogênio , Concentração Inibidora 50 , Cinética , Simulação de Dinâmica Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Termodinâmica , Uracila/química
7.
PLoS One ; 10(7): e0134506, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222439

RESUMO

Human aldo-keto reductase 1B15 (AKR1B15) is a newly discovered enzyme which shares 92% amino acid sequence identity with AKR1B10. While AKR1B10 is a well characterized enzyme with high retinaldehyde reductase activity, involved in the development of several cancer types, the enzymatic activity and physiological role of AKR1B15 are still poorly known. Here, the purified recombinant enzyme has been subjected to substrate specificity characterization, kinetic analysis and inhibitor screening, combined with structural modeling. AKR1B15 is active towards a variety of carbonyl substrates, including retinoids, with lower kcat and Km values than AKR1B10. In contrast to AKR1B10, which strongly prefers all-trans-retinaldehyde, AKR1B15 exhibits superior catalytic efficiency with 9-cis-retinaldehyde, the best substrate found for this enzyme. With ketone and dicarbonyl substrates, AKR1B15 also shows higher catalytic activity than AKR1B10. Several typical AKR inhibitors do not significantly affect AKR1B15 activity. Amino acid substitutions clustered in loops A and C result in a smaller, more hydrophobic and more rigid active site in AKR1B15 compared with the AKR1B10 pocket, consistent with distinct substrate specificity and narrower inhibitor selectivity for AKR1B15.


Assuntos
Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/metabolismo , Retinaldeído/metabolismo , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Aldeído Redutase/metabolismo , Aldo-Ceto Redutases , Sequência de Aminoácidos , Substituição de Aminoácidos , Domínio Catalítico/genética , Diterpenos , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Modelos Moleculares , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/antagonistas & inibidores , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/genética , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia Estrutural de Proteína , Especificidade por Substrato
8.
Chem Biol Interact ; 234: 105-13, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25619643

RESUMO

Prostaglandins (PGs) are lipid compounds derived from arachidonic acid by the action of cyclooxygenases, acting locally as messenger molecules in a wide variety of physiological processes, such as inflammation, cell survival, apoptosis, smooth muscle contraction, adipocyte differentiation, vasodilation and platelet aggregation inhibition. In the inactivating pathway of PGs, the first metabolic intermediates are 15-keto-PGs, which are further converted into 13,14-dihydro-15-keto-PGs by different enzymes having 15-keto-PG reductase activity. Three human PG reductases (PGR), zinc-independent members of the medium-chain dehydrogenase/reductase (MDR) superfamily, perform the first irreversible step of the degradation pathway. We have focused on the characterization of the recombinant human enzyme prostaglandin reductase 1 (PGR1), also known as leukotriene B4 dehydrogenase. Only a partial characterization of this enzyme, isolated from human placenta, had been previously reported. In the present work, we have developed a new HPLC-based method for the determination of the 15-keto-PG reductase activity. We have performed an extensive kinetic characterization of PGR1, which catalyzes the NADPH-dependent reduction of the α,ß-double bond of aliphatic and aromatic aldehydes and ketones, and 15-keto-PGs. PGR1 also shows low activity in the oxidation of leukotriene B4. The best substrates in terms of kcat/Km were 15-keto-PGE2, trans-3-nonen-2-one and trans-2-decenal. Molecular docking simulations, based on the three-dimensional structure of the human enzyme (PDB ID 2Y05), and site-directed mutagenesis studies were performed to pinpoint important structural determinants, highlighting the role of Arg56 and Tyr245 in 15-keto-PG binding. Finally, inhibition analysis was done using non-steroidal anti-inflammatory drugs (NSAIDs) as potential inhibitors.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidade por Substrato/genética , Aldeídos/metabolismo , Alcenos/metabolismo , Sequência de Aminoácidos , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Humanos , Cetonas/metabolismo , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/métodos , NADP/metabolismo , Oxirredutases/metabolismo , Alinhamento de Sequência
9.
Chem Biol Interact ; 234: 290-6, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25532697

RESUMO

Aldose reductase (AR, AKR1B1) and AKR1B10 are enzymes implicated in important pathologies (diabetes and cancer) and therefore they have been proposed as suitable targets for drug development. Sulindac is the metabolic precursor of the potent non-steroidal anti-inflammatory drug (NSAID) sulindac sulfide, which suppresses prostaglandin production by inhibition of cyclooxygenases (COX). In addition, sulindac has been found to be one of the NSAIDs with higher antitumoral activity, presumably through COX inhibition. However, sulindac anticancer activity could be partially mediated through COX-independent mechanisms, including the participation of AR and AKR1B10. Previously, it had been shown that sulindac and sulindac sulfone were good AR inhibitors and the structure of the ternary complex with NADP(+) and sulindac was described (PDB ID 3U2C). In this work, we determined the three-dimensional structure of AKR1B10 with sulindac and established structure-activity relationships (SAR) of sulindac and their derivatives with AR and AKR1B10. The difference in the IC50 values for sulindac between AR (0.36 µM) and AKR1B10 (2.7 µM) might be explained by the different positioning and stacking interaction given by Phe122/Phe123, and by the presence of two buried and ordered water molecules in AKR1B10 but not in AR. Moreover, SAR analysis shows that the substitution of the sulfinyl group is structurally allowed in sulindac derivatives. Hence, sulindac and its derivatives emerge as lead compounds for the design of more potent and selective AR and AKR1B10 inhibitors.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Sulindaco/farmacologia , Aldeído Redutase/metabolismo , Aldo-Ceto Redutases , Anti-Inflamatórios não Esteroides/farmacologia , Humanos , NADP/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas/metabolismo , Relação Estrutura-Atividade
10.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 3): 889-903, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24598757

RESUMO

Aldo-keto reductases (AKRs) are mostly monomeric enzymes which fold into a highly conserved (α/ß)8 barrel, while their substrate specificity and inhibitor selectivity are determined by interaction with residues located in three highly variable external loops. The closely related human enzymes aldose reductase (AR or AKR1B1) and AKR1B10 are of biomedical interest because of their involvement in secondary diabetic complications (AR) and in cancer, e.g. hepatocellular carcinoma and smoking-related lung cancer (AKR1B10). After characterization of the IC50 values of both AKRs with a series of polyhalogenated compounds, 2,2',3,3',5,5',6,6'-octafluoro-4,4'-biphenyldiol (JF0064) was identified as a lead inhibitor of both enzymes with a new scaffold (a 1,1'-biphenyl-4,4'-diol). An ultrahigh-resolution X-ray structure of the AR-NADP(+)-JF0064 complex has been determined at 0.85 Šresolution, allowing it to be observed that JF0064 interacts with the catalytic residue Tyr48 through a negatively charged hydroxyl group (i.e. the acidic phenol). The non-competitive inhibition pattern observed for JF0064 with both enzymes suggests that this acidic hydroxyl group is also present in the case of AKR1B10. Moreover, the combination of surface lysine methylation and the introduction of K125R and V301L mutations enabled the determination of the X-ray crystallographic structure of the corresponding AKR1B10-NADP(+)-JF0064 complex. Comparison of the two structures has unveiled some important hints for subsequent structure-based drug-design efforts.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/química , Desenho de Fármacos , Aldo-Ceto Redutases , Ácidos Carboxílicos/química , Cristalografia por Raios X , Estabilidade Enzimática/efeitos dos fármacos , Halogênios , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , NADP/química , Proteínas Recombinantes/química
11.
Chem Biol Interact ; 202(1-3): 195-203, 2013 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-23295224

RESUMO

The α-hydroxy ketones are used as building blocks for compounds of pharmaceutical interest (such as antidepressants, HIV-protease inhibitors and antitumorals). They can be obtained by the action of enzymes or whole cells on selected substrates, such as diketones. We have studied the enantiospecificities of several fungal (AKR3C1, AKR5F and AKR5G) and human (AKR1B1 and AKR1B10) aldo-keto reductases in the production of α-hydroxy ketones and diols from vicinal diketones. The reactions have been carried out with pure enzymes and with an NADPH-regenerating system consisting of glucose-6-phosphate and glucose-6-phosphate dehydrogenase. To ascertain the regio and stereoselectivity of the reduction reactions catalyzed by the AKRs, we have separated and characterized the reaction products by means of a gas chromatograph equipped with a chiral column and coupled to a mass spectrometer as a detector. According to the regioselectivity and stereoselectivity, the AKRs studied can be divided in two groups: one of them showed preference for the reduction of the proximal keto group, resulting in the S-enantiomer of the corresponding α-hydroxy ketones. The other group favored the reduction of the distal keto group and yielded the corresponding R-enantiomer. Three of the AKRs used (AKR1B1, AKR1B10 and AKR3C1) could produce 2,3-butanediol from acetoin. We have explored the structure/function relationships in the reactivity between several yeast and human AKRs and various diketones and acetoin. In addition, we have demonstrated the utility of these AKRs in the synthesis of selected α-hydroxy ketones and diols.


Assuntos
Oxirredutases do Álcool/metabolismo , Proteínas Fúngicas/metabolismo , Cetonas/metabolismo , Leveduras/enzimologia , Leveduras/metabolismo , Aldeído Redutase , Aldo-Ceto Redutases , Butileno Glicóis/metabolismo , Catálise , Glucose-6-Fosfato/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glucosídeos/metabolismo , Humanos , Cinética , NADP/metabolismo , Oxirredução , Pirimidinonas/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
12.
Chem Biol Interact ; 202(1-3): 186-94, 2013 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-23220004

RESUMO

Biological activity of natural retinoids requires the oxidation of retinol to retinoic acid (RA) and its binding to specific nuclear receptors in target tissues. The first step of this pathway, the reversible oxidoreduction of retinol to retinaldehyde, is essential to control RA levels. The enzymes of retinol oxidation are NAD-dependent dehydrogenases of the cytosolic medium-chain (MDR) and the membrane-bound short-chain (SDR) dehydrogenases/reductases. Retinaldehyde reduction can be performed by SDR and aldo-keto reductases (AKR), while its oxidation to RA is carried out by aldehyde dehydrogenases (ALDH). In contrast to SDR, AKR and ALDH are cytosolic. A common property of these enzymes is that they only use free retinoid, but not retinoid bound to cellular retinol binding protein (CRBP). The relative contribution of each enzyme type in retinoid metabolism is discussed in terms of the different subcellular localization, topology of membrane-bound enzymes, kinetic constants, binding affinity of CRBP for retinol and retinaldehyde, and partition of retinoid pools between membranes and cytoplasm. The development of selective inhibitors for AKR enzymes 1B1 and 1B10, of clinical relevance in diabetes and cancer, granted the investigation of some structure-activity relationships. Kinetics with the 4-methyl derivatives of retinaldehyde isomers was performed to identify structural features for substrate specificity. Hydrophilic derivatives were better substrates than the more hydrophobic compounds. We also explored the inhibitory properties of some synthetic retinoids, known for binding to retinoic acid receptors (RAR) and retinoid X receptors (RXR). Consistent with its substrate specificity towards retinaldehyde, AKR1B10 was more effectively inhibited by synthetic retinoids than AKR1B1. A RARß/γ agonist (UVI2008) inhibited AKR1B10 with the highest potency and selectivity, and docking simulations predicted that its carboxyl group binds to the anion-binding pocket.


Assuntos
Oxirredutases do Álcool/metabolismo , Retinaldeído/metabolismo , Retinoides/metabolismo , Aldeído Desidrogenase/metabolismo , Aldeído Redutase , Aldo-Ceto Redutases , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Citosol/enzimologia , Citosol/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Oxirredução , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/metabolismo , Proteínas Celulares de Ligação ao Retinol/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Tretinoína/metabolismo , Vitamina A/metabolismo
13.
Front Pharmacol ; 3: 58, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22529810

RESUMO

Several aldo-keto reductase (AKR) enzymes from subfamilies 1B and 1C show retinaldehyde reductase activity, having low K(m) and k(cat) values. Only AKR1B10 and 1B12, with all-trans-retinaldehyde, and AKR1C3, with 9-cis-retinaldehyde, display high catalytic efficiency. Major structural determinants for retinaldehyde isomer specificity are located in the external loops (A and C for AKR1B10, and B for AKR1C3), as assessed by site-directed mutagenesis and molecular dynamics. Cellular models have shown that AKR1B and 1C enzymes are well suited to work in vivo as retinaldehyde reductases and to regulate retinoic acid (RA) biosynthesis at hormone pre-receptor level. An additional physiological role for the retinaldehyde reductase activity of these enzymes, consistent with their tissue localization, is their participation in ß-carotene absorption. Retinaldehyde metabolism may be subjected to subcellular compartmentalization, based on enzyme localization. While retinaldehyde oxidation to RA takes place in the cytosol, reduction to retinol could take place in the cytosol by AKRs or in the membranes of endoplasmic reticulum by microsomal retinaldehyde reductases. Upregulation of some AKR1 enzymes in different cancer types may be linked to their induction by oxidative stress and to their participation in different signaling pathways related to cell proliferation. AKR1B10 and AKR1C3, through their retinaldehyde reductase activity, trigger a decrease in the RA biosynthesis flow, resulting in RA deprivation and consequently lower differentiation, with an increased cancer risk in target tissues. Rational design of selective AKR inhibitors could lead to development of novel drugs for cancer treatment as well as reduction of chemotherapeutic drug resistance.

14.
Biochem J ; 440(3): 335-44, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21851338

RESUMO

Human AKR (aldo-keto reductase) 1C proteins (AKR1C1-AKR1C4) exhibit relevant activity with steroids, regulating hormone signalling at the pre-receptor level. In the present study, investigate the activity of the four human AKR1C enzymes with retinol and retinaldehyde. All of the enzymes except AKR1C2 showed retinaldehyde reductase activity with low Km values (~1 µM). The kcat values were also low (0.18-0.6 min-1), except for AKR1C3 reduction of 9-cis-retinaldehyde whose kcat was remarkably higher (13 min-1). Structural modelling of the AKR1C complexes with 9-cis-retinaldehyde indicated a distinct conformation of Trp227, caused by changes in residue 226 that may contribute to the activity differences observed. This was partially supported by the kinetics of the AKR1C3 R226P mutant. Retinol/retinaldehyde conversion, combined with the use of the inhibitor flufenamic acid, indicated a relevant role for endogenous AKR1Cs in retinaldehyde reduction in MCF-7 breast cancer cells. Overexpression of AKR1C proteins depleted RA (retinoic acid) transactivation in HeLa cells treated with retinol. Thus AKR1Cs may decrease RA levels in vivo. Finally, by using lithocholic acid as an AKR1C3 inhibitor and UVI2024 as an RA receptor antagonist, we provide evidence that the pro-proliferative action of AKR1C3 in HL-60 cells involves the RA signalling pathway and that this is in part due to the retinaldehyde reductase activity of AKR1C3.


Assuntos
20-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , Hidroxiprostaglandina Desidrogenases/metabolismo , Retinaldeído/química , 20-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/genética , Membro C3 da Família 1 de alfa-Ceto Redutase , Substituição de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiesteroide Desidrogenases/química , Hidroxiesteroide Desidrogenases/metabolismo , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Oxirredutases/química , Oxirredutases/metabolismo , Ligação Proteica , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Retinaldeído/farmacologia , Retinaldeído/fisiologia , Especificidade por Substrato , Ativação Transcricional , Vitamina A/química , Vitamina A/farmacologia , Vitamina A/fisiologia
15.
Chem Biol Interact ; 191(1-3): 32-7, 2011 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-21276778

RESUMO

ζ-Crystallins are a Zn(2+)-lacking enzyme group with quinone reductase activity, which belongs to the medium-chain dehydrogenase/reductase superfamily. It has been recently observed that human ζ-crystallin is capable of reducing the α,ß-double bond of alkenals and alkenones. Here we report that this activity is also shared by the homologous Zta1p enzyme from Saccharomyces cerevisiae. While the two enzymes show similar substrate specificity, human ζ-crystallin exhibits higher activity with lipid peroxidation products and Zta1p is more active with cinnamaldehyde. The presence of Zta1p has an in vivo protective effect on yeast strains exposed to the toxic substrate 3-penten-2-one. Analysis of ZTA1 gene expression indicates an induction under different types of cellular stress, including ethanol and dimethylsulfoxide exposure and by reaching the stationary growth phase. The role of Zta1p in the yeast adaptation to some stress types and the general functional significance of ζ-crystallins are discussed.


Assuntos
NAD(P)H Desidrogenase (Quinona)/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , zeta-Cristalinas/metabolismo , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , NAD(P)H Desidrogenase (Quinona)/genética , Pentanonas/química , Pentanonas/toxicidade , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , zeta-Cristalinas/genética
16.
Cell Mol Life Sci ; 68(6): 1065-77, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20835842

RESUMO

Human ζ-crystallin is a Zn(2+)-lacking medium-chain dehydrogenase/reductase (MDR) included in the quinone oxidoreductase (QOR) family because of its activity with quinones. In the present work a novel enzymatic activity was characterized: the double bond α,ß-hydrogenation of medium-chain 2-alkenals and 3-alkenones. The enzyme is especially active with lipid peroxidation products such as 4-hydroxyhexenal, and a role in their detoxification is discussed. This specificity is novel in the QOR family, and it is similar to that described in the distantly related alkenal/one reductase family. Moreover, we report the X-ray structure of ζ-crystallin, which represents the first structure solved for a tetrameric Zn(2+)-lacking MDR, and which allowed the identification of the active-site lining residues. Docking simulations suggest a role for Tyr53 and Tyr59 in catalysis. The kinetics of Tyr53Phe and Tyr59Phe mutants support the implication of Tyr53 in binding/catalysis of alkenal/one substrates, while Tyr59 is involved in the recognition of 4-OH-alkenals.


Assuntos
Modelos Moleculares , zeta-Cristalinas/química , zeta-Cristalinas/metabolismo , Aldeídos/metabolismo , Catálise , Clonagem Molecular , Cristalografia por Raios X , Primers do DNA/genética , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Hidrogenação , Cinética , Estrutura Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Conformação Proteica , Especificidade por Substrato , zeta-Cristalinas/isolamento & purificação
17.
J Biol Chem ; 284(25): 17194-17205, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19349281

RESUMO

Tumor suppressor p53 regulates the expression of p53-induced genes (PIG) that trigger apoptosis. PIG3 or TP53I3 is the only known member of the medium chain dehydrogenase/reductase superfamily induced by p53 and is used as a proapoptotic marker. Although the participation of PIG3 in the apoptotic pathway is proven, the protein and its mechanism of action were never characterized. We analyzed human PIG3 enzymatic function and found NADPH-dependent reductase activity with ortho-quinones, which is consistent with the classification of PIG3 in the quinone oxidoreductase family. However, the activity is much lower than that of zeta-crystallin, a better known quinone oxidoreductase. In addition, we report the crystallographic structure of PIG3, which allowed the identification of substrate- and cofactor-binding sites, with residues fully conserved from bacteria to human. Tyr-59 in zeta-crystallin (Tyr-51 in PIG3) was suggested to participate in the catalysis of quinone reduction. However, kinetics of Tyr/Phe and Tyr/Ala mutants of both enzymes demonstrated that the active site Tyr is not catalytic but may participate in substrate binding, consistent with a mechanism based on propinquity effects. It has been proposed that PIG3 contribution to apoptosis would be through oxidative stress generation. We found that in vitro activity and in vivo overexpression of PIG3 accumulate reactive oxygen species. Accordingly, an inactive PIG3 mutant (S151V) did not produce reactive oxygen species in cells, indicating that enzymatically active protein is necessary for this function. This supports that PIG3 action is through oxidative stress produced by its enzymatic activity and provides essential knowledge for eventual control of apoptosis.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Sequência de Aminoácidos , Apoptose/fisiologia , Sítios de Ligação , Domínio Catalítico/genética , Cristalografia por Raios X , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , NADP/metabolismo , Filogenia , Estrutura Quaternária de Proteína , Proteínas Proto-Oncogênicas/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Proteína Supressora de Tumor p53/metabolismo , Tirosina/química
18.
Chem Biol Interact ; 178(1-3): 288-94, 2009 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-19007762

RESUMO

The medium-chain dehydrogenase/reductase (MDR) superfamily can be divided into Zn-containing and Zn-lacking proteins. Zn-containing MDRs are generally well-known enzymes, mostly acting as dehydrogenases. The non-Zn MDR are much less studied, and classified in several families of NADP(H)-dependent reductases, including quinone oxidoreductases (QOR). zeta-Crystallins are the best studied group of QOR, have a structural function in the lens of several mammals, exhibit ortho-quinone reductase activity, and bind to specific adenine-uracil-rich elements (ARE) in RNA. In the present work, we have further characterized human zeta-crystallin and Saccharomyces cerevisiae Zta1p, the only QOR in yeast. Subcellular localization using a fluorescent protein tag indicates that zeta-crystallin is distributed in the cytoplasm but not in nucleus. The protein may also be present in mitochondria. Zta1p localizes in both cytoplasm and nucleus. NADPH, but not NADH, competitively prevents binding of zeta-crystallin to RNA, suggesting that the cofactor-binding site is involved in RNA binding. Interference of NADPH on Zta1p binding to RNA is much lower, consistent with a weaker binding of NADPH to the yeast enzyme. Disruption of the yeast ZTA1 gene does not affect cell growth under standard conditions but makes yeast more sensitive to oxidative stress agents. Sequence alignments, phylogenetic tree analysis and kinetic properties reveal a close relationship between zeta-crystallin and Zta1p. Amino acid conservation, between the substrate-binding sites of the two proteins and that of an E. coli QOR, indicates that zeta-crystallins maintained their kinetic function throughout evolution. Quinones are toxic compounds and a relevant step in their detoxification is reduction to their corresponding hydroquinones. Many enzymes of several superfamilies can reduce quinones, including NAD(P)H:quinone oxidoreductase 1 (NQO1 or DT-diaphorase), aldo-keto reductases and short-chain dehydrogenases/reductases. In this context, the physiological role of zeta-crystallins is discussed.


Assuntos
Quinona Redutases/metabolismo , Saccharomyces cerevisiae/metabolismo , zeta-Cristalinas/metabolismo , Sequência de Bases , Western Blotting , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Filogenia , RNA/metabolismo , Frações Subcelulares/enzimologia , Transcrição Gênica , zeta-Cristalinas/química
19.
Biochem J ; 399(1): 101-9, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16787387

RESUMO

Retinoic acid biosynthesis in vertebrates occurs in two consecutive steps: the oxidation of retinol to retinaldehyde followed by the oxidation of retinaldehyde to retinoic acid. Enzymes of the MDR (medium-chain dehydrogenase/reductase), SDR (short-chain dehydrogenase/reductase) and AKR (aldo-keto reductase) superfamilies have been reported to catalyse the conversion between retinol and retinaldehyde. Estimation of the relative contribution of enzymes of each type was difficult since kinetics were performed with different methodologies, but SDRs would supposedly play a major role because of their low K(m) values, and because they were found to be active with retinol bound to CRBPI (cellular retinol binding protein type I). In the present study we employed detergent-free assays and HPLC-based methodology to characterize side-by-side the retinoid-converting activities of human MDR [ADH (alcohol dehydrogenase) 1B2 and ADH4), SDR (RoDH (retinol dehydrogenase)-4 and RDH11] and AKR (AKR1B1 and AKR1B10) enzymes. Our results demonstrate that none of the enzymes, including the SDR members, are active with CRBPI-bound retinoids, which questions the previously suggested role of CRBPI as a retinol supplier in the retinoic acid synthesis pathway. The members of all three superfamilies exhibit similar and low K(m) values for retinoids (0.12-1.1 microM), whilst they strongly differ in their kcat values, which range from 0.35 min(-1) for AKR1B1 to 302 min(-1) for ADH4. ADHs appear to be more effective retinol dehydrogenases than SDRs because of their higher kcat values, whereas RDH11 and AKR1B10 are efficient retinaldehyde reductases. Cell culture studies support a role for RoDH-4 as a retinol dehydrogenase and for AKR1B1 as a retinaldehyde reductase in vivo.


Assuntos
Acil-CoA Desidrogenase/metabolismo , Oxirredutases do Álcool/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Retinoides/metabolismo , Aldeído Redutase , Aldo-Ceto Redutases , Animais , Linhagem Celular , Regulação Enzimológica da Expressão Gênica , Humanos , Insetos
20.
Eur J Biochem ; 270(12): 2652-62, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787032

RESUMO

ADH1 and ADH4 are the major alcohol dehydrogenases (ADH) in ethanol and retinol oxidation. ADH activity and protein expression were investigated in rat gastrointestinal tissue homogenates by enzymatic and Western blot analyses. In addition, sections of adult rat gastrointestinal tract were examined by in situ hybridization and immunohistochemistry. ADH1 and ADH4 were detected along the whole tract, changing their localization and relative content as a function of the area studied. While ADH4 was more abundant in the upper (esophagus and stomach) and lower (colorectal) regions, ADH1 was predominant in the intestine but also present in stomach. Both enzymes were detected in mucosa but, in general, ADH4 was found in outer cell layers, lining the lumen, while ADH1 was detected in the inner cell layers. Of interest were the sharp discontinuities in the expression found in the pyloric region (ADH1) and the gastroduodenal junction (ADH4), reflecting functional changes. The precise localization of ADH in the gut reveals the cell types where active alcohol oxidation occurs during ethanol ingestion, providing a molecular basis for the gastrointestinal alcohol pathology. Localization of ADH, acting as retinol dehydrogenase/retinal reductase, also indicates sites of active retinoid metabolism in the gut, essential for mucosa function and vitamin A absorption.


Assuntos
Álcool Desidrogenase/genética , Sistema Digestório/enzimologia , Mucosa Intestinal/enzimologia , Álcool Desidrogenase/metabolismo , Animais , Esôfago/enzimologia , Íleo/enzimologia , Imuno-Histoquímica , Hibridização In Situ , Isoenzimas/genética , Isoenzimas/metabolismo , Jejuno/enzimologia , Ratos , Língua/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA