Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Med Chem ; 247: 115013, 2023 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-36566714

RESUMO

Aldo-keto reductase 1C3 (AKR1C3) is overexpressed in multiple hormone related cancers, such as breast and prostate cancer, and is correlated with tumor development and aggressiveness. As a phase I biotransformation enzyme, AKR1C3 catalyzes the metabolic processes that lead to resistance to anthracyclines, the "gold standard" for breast cancer treatment. Novel approaches to restore the chemotherapy sensitivity of breast cancer are urgently required. Herein, we developed a new class of AKR1C3 inhibitors that demonstrated potent inhibitory activity and exquisite selectivity for closely related isoforms. The best derivative 27 (S19-1035) exhibits an IC50 value of 3.04 nM for AKR1C3 and >3289-fold selectivity over other isoforms. We determined the co-crystal structures of AKR1C3 with three of the inhibitors, providing a solid foundation for further structure-based drug optimization. Co-administration of these AKR1C3 inhibitors significantly reversed the doxorubicin (DOX) resistance in a resistant breast cancer cell line. Therefore, the novel AKR1C3 specific inhibitors developed in this work may serve as effective adjuvants to overcome DOX resistance in breast cancer treatment.


Assuntos
Neoplasias da Mama , Masculino , Humanos , Neoplasias da Mama/tratamento farmacológico , Preparações Farmacêuticas , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Antibióticos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química
2.
World J Gastroenterol ; 23(20): 3572-3580, 2017 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-28611510

RESUMO

The biosynthesis of prostaglandins and thromboxanes has been a focus of interest in the management of many liver diseases. Cyclooxygenases are the enzymes involved in the first step of the biosynthesis of these lipid mediators and selective inhibitors for these isoenzymes as well as pharmacological analogues of prostaglandins have been developed and are currently applied therapeutically. Here we discuss the implications of these enzymes in the onset of metabolic and lipid disorders in the liver and their potential role in the progression of the diseases towards fibrosis and hepatocellular carcinogenesis.


Assuntos
Carcinoma Hepatocelular/enzimologia , Ciclo-Oxigenase 2/metabolismo , Neoplasias Hepáticas/enzimologia , Animais , Carcinogênese , Catálise , Proliferação de Células , Progressão da Doença , Fibrose , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/química , Lipídeos/química , Fígado/metabolismo , Fígado/fisiopatologia , Cirrose Hepática/enzimologia , Camundongos , Hepatopatia Gordurosa não Alcoólica/enzimologia , Oxigênio/química , Transgenes , Resultado do Tratamento
3.
J Steroid Biochem Mol Biol ; 171: 270-280, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28457968

RESUMO

17beta-hydroxysteroid dehydrogenase type 5 (17ß-HSD5) is an important enzyme associated with sex steroid metabolism in hormone-dependent cancer. However, reports on its expression and its prognostic value in breast cancer are inconsistent. Here, we demonstrate the impact of 17ß-HSD5 expression modulation on the proteome of estrogen receptor-positive (ER+) breast cancer cells. RNA interference technique (siRNA) was used to knock down 17ß-HSD5 gene expression in the ER+ breast cancer cell line MCF-7 and the proteome of the 17ß-HSD5-knockdown cells was compared to that of MCF-7 cells using two-dimensional (2-D) gel electrophoresis followed by mass spectrometry analysis. Ingenuity pathway analysis (IPA) was additionally used to assess functional enrichment analyses of the proteomic dataset, including protein network and canonical pathways. Our proteomic analysis revealed only four differentially expressed protein spots (fold change > 2, p<0.05) between the two cell lines. The four spots were up-regulated in 17ß-HSD5-knockdown MCF-7 cells, and comprised 21 proteins involved in two networks and in functions that include apoptosis inhibition, regulation of cell growth and differentiation, signal transduction and tumor metastasis. Among the proteins are nucleoside diphosphate kinase A (NME1), 78kDa glucose-regulated protein (GRP78) and phosphoglycerate kinase 1 (PGK1). We also showed that expression of 17ß-HSD5 and that of the apoptosis inhibitor GRP78 are strongly but negatively correlated. Consistent with their opposite regulation, GRP78 knockdown decreased MCF-7 cell viability whereas 17ß-HSD5 knockdown or inhibition increased cell viability and proliferation. Besides, IPA analysis revealed that ubiquitination pathway is significantly affected by 17ß-HSD5 knockdown. Furthermore, IPA predicted the proto-oncogene c-Myc as an upstream regulator linked to the tumor-secreted protein PGK1. The latter is over-expressed in invasive ductal breast carcinoma as compared with normal breast tissue and its expression increased following 17ß-HSD5 knockdown. Our present results indicate a 17ß-HSD5 role in down-regulating breast cancer development. We thus propose that 17ß-HSD5 may not be a potent target for breast cancer treatment but its low expression could represent a poor prognosis factor.


Assuntos
3-Hidroxiesteroide Desidrogenases/metabolismo , Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/metabolismo , Hidroxiprostaglandina Desidrogenases/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoglicerato Quinase/metabolismo , 3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/genética , Membro C3 da Família 1 de alfa-Ceto Redutase , Neoplasias da Mama/patologia , Proliferação de Células , Sobrevivência Celular , Chaperona BiP do Retículo Endoplasmático , Ativação Enzimática , Feminino , Perfilação da Expressão Gênica , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Humanos , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/genética , Processamento de Imagem Assistida por Computador , Células MCF-7 , Nucleosídeo NM23 Difosfato Quinases/química , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fosfoglicerato Quinase/química , Fosfoglicerato Quinase/genética , Proteômica/métodos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Receptores de Estrogênio/metabolismo , Eletroforese em Gel Diferencial Bidimensional
4.
Biochem Pharmacol ; 116: 176-87, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27453434

RESUMO

The clinical stage anti-cancer agent PR-104 has potential utility as a cytotoxic prodrug for exogenous bacterial nitroreductases expressed from replicating vector platforms. However substrate selectivity is compromised due to metabolism by the human one- and two-electron oxidoreductases cytochrome P450 oxidoreductase (POR) and aldo-keto reductase 1C3 (AKR1C3). Using rational drug design we developed a novel mono-nitro analog of PR-104A that is essentially free of this off-target activity in vitro and in vivo. Unlike PR-104A, there was no biologically relevant cytotoxicity in cells engineered to express AKR1C3 or POR, under aerobic or anoxic conditions, respectively. We screened this inert prodrug analog, SN34507, against a type I bacterial nitroreductase library and identified E. coli NfsA as an efficient bioactivator using a DNA damage response assay and recombinant enzyme kinetics. Expression of E. coli NfsA in human colorectal cancer cells led to selective cytotoxicity to SN34507 that was associated with cell cycle arrest and generated a robust 'bystander effect' at tissue-like cell densities when only 3% of cells were NfsA positive. Anti-tumor activity of SN35539, the phosphate pre-prodrug of SN34507, was established in 'mixed' tumors harboring a minority of NfsA-positive cells and demonstrated marked tumor control following heterogeneous suicide gene expression. These experiments demonstrate that off-target metabolism of PR-104 can be avoided and identify the suicide gene/prodrug partnership of E. coli NfsA/SN35539 as a promising combination for development in armed vectors.


Assuntos
3-Hidroxiesteroide Desidrogenases/metabolismo , Antineoplásicos Alquilantes/uso terapêutico , Benzamidas/uso terapêutico , Carcinoma/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Desenho de Fármacos , Hidroxiprostaglandina Desidrogenases/metabolismo , Mesilatos/uso terapêutico , Modelos Moleculares , Organofosfonatos/uso terapêutico , Pró-Fármacos/uso terapêutico , 3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/genética , Ativação Metabólica/efeitos dos fármacos , Membro C3 da Família 1 de alfa-Ceto Redutase , Animais , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacologia , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacologia , Carcinoma/metabolismo , Carcinoma/patologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HCT116 , Humanos , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/genética , Mesilatos/química , Mesilatos/metabolismo , Mesilatos/farmacologia , Camundongos Nus , Simulação de Acoplamento Molecular , Nitrorredutases/genética , Nitrorredutases/metabolismo , Organofosfonatos/química , Organofosfonatos/metabolismo , Organofosfonatos/farmacologia , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Distribuição Aleatória , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Especificidade por Substrato , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 918-27, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849402

RESUMO

Type 5 17ß-hydroxysteroid dehydrogenase (17ß-HSD5) is an aldo-keto reductase expressed in the human prostate which catalyzes the conversion of androstenedione to testosterone. Testosterone is converted to 5α-dihydrotestosterone, which is present at high concentrations in patients with castration-resistant prostate cancer (CRPC). Inhibition of 17ß-HSD5 is therefore considered to be a promising therapy for treating CRPC. In the present study, crystal structures of complexes of 17ß-HSD5 with structurally diverse inhibitors derived from high-throughput screening were determined. In the structures of the complexes, various functional groups, including amide, nitro, pyrazole and hydroxyl groups, form hydrogen bonds to the catalytic residues His117 and Tyr55. In addition, major conformational changes of 17ß-HSD5 were observed following the binding of the structurally diverse inhibitors. These results demonstrate interactions between 17ß-HSD5 and inhibitors at the atomic level and enable structure-based drug design for anti-CRPC therapy.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Hidroxiprostaglandina Desidrogenases/química , Conformação Proteica/efeitos dos fármacos , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Cristalografia por Raios X , Humanos , Hidroxiprostaglandina Desidrogenases/metabolismo , Modelos Moleculares , NADP/metabolismo
6.
Bioorg Med Chem ; 22(3): 967-77, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24411201

RESUMO

Inhibitors of the aldo-keto reductase enzyme AKR1C3 are of interest as potential drugs for leukemia and hormone-related cancers. A series of non-carboxylate morpholino(phenylpiperazin-1-yl)methanones were prepared by palladium-catalysed coupling of substituted phenyl or pyridyl bromides with the known morpholino(piperazin-1-yl)methanone, and shown to be potent (IC50∼100nM) and very isoform-selective inhibitors of AKR1C3. Lipophilic electron-withdrawing substituents on the phenyl ring were positive for activity, as was an H-bond acceptor on the other terminal ring, and the ketone moiety (as a urea) was essential. These structure-activity relationships are consistent with an X-ray structure of a representative compound bound in the AKR1C3 active site, which showed H-bonding between the carbonyl oxygen of the drug and Tyr55 and His117 in the 'oxyanion hole' of the enzyme, with the piperazine bridging unit providing the correct twist to allow the terminal benzene ring to occupy the lipophilic pocket and align with Phe311.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Domínio Catalítico , Técnicas de Química Sintética , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Ligação de Hidrogênio , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Concentração Inibidora 50 , Modelos Moleculares , Estrutura Molecular , Morfolinas/química , Relação Estrutura-Atividade
7.
J Med Chem ; 56(6): 2429-46, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23432095

RESUMO

Castrate-resistant prostate cancer (CRPC) is a fatal, metastatic form of prostate cancer. CRPC is characterized by reactivation of the androgen axis due to changes in androgen receptor signaling and/or adaptive intratumoral androgen biosynthesis. AKR1C3 is upregulated in CRPC where it catalyzes the formation of potent androgens. This makes AKR1C3 a target for the treatment of CRPC. AKR1C3 inhibitors should not inhibit AKR1C1/AKR1C2, which inactivate 5α-dihydrotestosterone. Indomethacin, used to inhibit cyclooxygenase, also inhibits AKR1C3 and displays selectivity over AKR1C1/AKR1C2. Parallel synthetic strategies were used to generate libraries of indomethacin analogues, which exhibit reduced cyclooxygenase inhibitory activity but retain AKR1C3 inhibitory potency and selectivity. The lead compounds inhibited AKR1C3 with nanomolar potency, displayed >100-fold selectivity over AKR1C1/AKR1C2, and blocked testosterone formation in LNCaP-AKR1C3 cells. The AKR1C3·NADP(+)·2'-des-methyl-indomethacin crystal structure was determined, and it revealed a unique inhibitor binding mode. The compounds reported are promising agents for the development of therapeutics for CRPC.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Indometacina/análogos & derivados , Indometacina/farmacologia , Orquiectomia , Neoplasias da Próstata/enzimologia , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Domínio Catalítico , Genes Reporter/genética , Células HeLa , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Masculino , Modelos Moleculares , Metástase Neoplásica , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Receptores Androgênicos/genética , Especificidade por Substrato
8.
PLoS One ; 7(8): e43965, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22937138

RESUMO

Aldo-keto reductase 1C3 (AKR1C3) catalyses the NADPH dependent reduction of carbonyl groups in a number of important steroid and prostanoid molecules. The enzyme is also over-expressed in prostate and breast cancer and its expression is correlated with the aggressiveness of the disease. The steroid products of AKR1C3 catalysis are important in proliferative signalling of hormone-responsive cells, while the prostanoid products promote prostaglandin-dependent proliferative pathways. In these ways, AKR1C3 contributes to tumour development and maintenance, and suggest that inhibition of AKR1C3 activity is an attractive target for the development of new anti-cancer therapies. Non-steroidal anti-inflammatory drugs (NSAIDs) are one well-known class of compounds that inhibits AKR1C3, yet crystal structures have only been determined for this enzyme with flufenamic acid, indomethacin, and closely related analogues bound. While the flufenamic acid and indomethacin structures have been used to design novel inhibitors, they provide only limited coverage of the NSAIDs that inhibit AKR1C3 and that may be used for the development of new AKR1C3 targeted drugs. To understand how other NSAIDs bind to AKR1C3, we have determined ten crystal structures of AKR1C3 complexes that cover three different classes of NSAID, N-phenylanthranilic acids (meclofenamic acid, mefenamic acid), arylpropionic acids (flurbiprofen, ibuprofen, naproxen), and indomethacin analogues (indomethacin, sulindac, zomepirac). The N-phenylanthranilic and arylpropionic acids bind to common sites including the enzyme catalytic centre and a constitutive active site pocket, with the arylpropionic acids probing the constitutive pocket more effectively. By contrast, indomethacin and the indomethacin analogues sulindac and zomepirac, display three distinctly different binding modes that explain their relative inhibition of the AKR1C family members. This new data from ten crystal structures greatly broadens the base of structures available for future structure-guided drug discovery efforts.


Assuntos
3-Hidroxiesteroide Desidrogenases/química , Anti-Inflamatórios não Esteroides/química , Hidroxiprostaglandina Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Anti-Inflamatórios não Esteroides/metabolismo , Ácido Flufenâmico/química , Ácido Flufenâmico/metabolismo , Flurbiprofeno/química , Flurbiprofeno/metabolismo , Hidroxiprostaglandina Desidrogenases/metabolismo , Ibuprofeno/química , Ibuprofeno/metabolismo , Indometacina/química , Indometacina/metabolismo , Ácido Meclofenâmico/química , Ácido Meclofenâmico/metabolismo , Ácido Mefenâmico/química , Ácido Mefenâmico/metabolismo , Naproxeno/química , Naproxeno/metabolismo , Sulindaco/química , Sulindaco/metabolismo , Tolmetino/análogos & derivados , Tolmetino/química , Tolmetino/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-22505408

RESUMO

Aldo-keto reductase 1C3 (AKR1C3) is a human enzyme that catalyzes the NADPH-dependent reduction of steroids and prostaglandins. AKR1C3 overexpression is associated with the proliferation of hormone-dependent cancers, most notably breast and prostate cancers. Nonsteroidal anti-inflammatory drugs (NSAIDs) and their analogues are well characterized inhibitors of AKR1C3. Here, the X-ray crystal structure of 3-phenoxybenzoic acid in complex with AKR1C3 is presented. This structure provides useful information for the future development of new anticancer agents by structure-guided drug design.


Assuntos
3-Hidroxiesteroide Desidrogenases/química , Benzoatos/química , Inibidores Enzimáticos/química , Hidroxiprostaglandina Desidrogenases/química , Domínios e Motivos de Interação entre Proteínas , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Benzoatos/metabolismo , Domínio Catalítico , Inibidores Enzimáticos/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/metabolismo , Ligantes , Modelos Moleculares , Ligação Proteica
10.
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
11.
Biochem J ; 437(1): 53-61, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21521174

RESUMO

Active sex hormones such as testosterone and progesterone are metabolized to tetrahydrosteroids in the liver to terminate hormone action. One main metabolic pathway, the 5ß-pathway, involves 5ß-steroid reductase (AKR1D1, where AKR refers to the aldo-keto reductase superfamily), which catalyses the reduction of the 4-ene structure, and ketosteroid reductases (AKR1C1-AKR1C4), which catalyse the subsequent reduction of the 3-oxo group. The activities of the four human AKR1C enzymes on 5ß-dihydrotestosterone, 5ß-pregnane-3,20-dione and 20α-hydroxy-5ß-pregnan-3-one, the intermediate 5ß-dihydrosteroids on the 5ß-pathway of testosterone and progesterone metabolism, were investigated. Product characterization by liquid chromatography-MS revealed that the reduction of the 3-oxo group of the three steroids predominantly favoured the formation of the corresponding 3α-hydroxy steroids. The stereochemistry was explained by molecular docking. Kinetic properties of the enzymes identified AKR1C4 as the major enzyme responsible for the hepatic formation of 5ß-tetrahydrosteroid of testosterone, but indicated differential routes and roles of human AKR1C for the hepatic formation of 5ß-tetrahydrosteroids of progesterone. Comparison of the kinetics of the AKR1C1-AKR1C4-catalysed reactions with those of AKR1D1 suggested that the three intermediate 5ß-dihydrosteroids derived from testosterone and progesterone are unlikely to accumulate in liver, and that the identities and levels of 5ß-reduced metabolites formed in peripheral tissues will be governed by the local expression of AKR1D1 and AKR1C1-AKR1C3.


Assuntos
Oxirredutases/metabolismo , Progesterona/metabolismo , Testosterona/metabolismo , 20-Hidroxiesteroide Desidrogenases/química , 20-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Sítios de Ligação , Catálise , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Hidroxiesteroide Desidrogenases/química , Hidroxiesteroide Desidrogenases/metabolismo , Cetosteroides/metabolismo , Cinética , Oxirredução , Oxirredutases/química , Estereoisomerismo
12.
Chem Biol Interact ; 191(1-3): 227-33, 2011 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-21182831

RESUMO

The human aldo-keto reductases 1C1 and 1C3 (AKR1C1 and AKR1C3) are important 20-ketosteroid reductases in pre-receptor regulation of progesterone action. Both AKR1C1 and AKR1C3 convert progesterone to the less potent metabolite 20α-hydroxyprogesterone, although AKR1C1 has a higher catalytic efficiency than AKR1C3. Recently, we reported significant up-regulation of AKR1C1 and AKR1C3 in ovarian endometriosis, a complex estrogen-dependent disease. The typical characteristics of endometriosis are increased formation of estradiol, which stimulates proliferation of endometriotic tissue, and disturbed action of the protective progesterone. Although progestins have been used for treatment of endometriosis since the 1960s, their detailed mechanisms of action are still not completely understood. In the present study, we evaluated the potential inhibitory effects of progestins on the pre-receptor regulatory enzymes AKR1C1 and AKR1C3. We examined the following progestins as inhibitors of progesterone reduction catalyzed by recombinant AKR1C1 and AKR1C3: progesterone derivatives (dydrogesterone, its metabolite, 20α-hydroxydydrogesterone; and medroxyprogesterone acetate), 19-nortestosterone derivatives (desogestrel, norethinodrone and levonorgestrel), and the androgen danazol. Dydrogesterone, medroxyprogesterone acetate, 20α-hydroxydydrogesterone and norethinodrone inhibited AKR1C1 and AKR1C3 with K(i) values of 1.9 µM, 7.9 µM, 20.8 µM and 48.0 µM, and of 0.5 µM, 1.4 µM, 18.2 µM and 6.6 µM, respectively. Levonorgestrel and desogestrel preferentially inhibited AKR1C3 with K(i) values of 5.6µM and 39.1µM, respectively. Our data thus show that dydrogesterone, medroxyprogesterone acetate, 20α-hydroxydydrogesterone and norethinodrone inhibit AKR1C1 and AKR1C3 in vitro, although their physiological inhibitory effects still need to be evaluated further. Additionally, we investigated whether progestin dydrogesterone can be metabolized to its active 20α-hydroxymetabolite by AKR1C1 and AKR1C3. AKR1C1 converted dydrogesterone with a high catalytic efficiency while AKR1C3 was less active, which suggests that in vivo dydrogesterone is metabolized mainly by AKR1C1. Docking simulations of dydrogesterone into AKR1C1 and AKR1C3 also support these experimental data.


Assuntos
20-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Progestinas/farmacologia , 20-Hidroxiesteroide Desidrogenases/química , 20-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Biocatálise , Avaliação Pré-Clínica de Medicamentos , Didrogesterona/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Modelos Moleculares , Oxirredução/efeitos dos fármacos , Progestinas/química , Progestinas/metabolismo , Conformação Proteica
13.
J Steroid Biochem Mol Biol ; 125(1-2): 95-104, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21087665

RESUMO

There is considerable interest in the development of an inhibitor of aldo-keto reductase (AKR) 1C3 (type 5 17ß-hydroxysteroid dehydrogenase and prostaglandin F synthase) as a potential therapeutic for both hormone-dependent and hormone-independent cancers. AKR1C3 catalyzes the reduction of 4-androstene-3,17-dione to testosterone and estrone to 17ß-estradiol in target tissues, which will promote the proliferation of hormone dependent prostate and breast cancers, respectively. AKR1C3 also catalyzes the reduction of prostaglandin (PG) H(2) to PGF(2α) and PGD(2) to 9α,11ß-PGF(2), which will limit the formation of anti-proliferative prostaglandins, including 15-deoxy-Δ(12,14)-PGJ(2), and contribute to proliferative signaling. AKR1C3 is overexpressed in a wide variety of cancers, including breast and prostate cancer. An inhibitor of AKR1C3 should not inhibit the closely related isoforms AKR1C1 and AKR1C2, as they are involved in other key steroid hormone biotransformations in target tissues. Several structural leads have been explored as inhibitors of AKR1C3, including non-steroidal anti-inflammatory drugs, steroid hormone analogues, flavonoids, cyclopentanes, and benzodiazepines. Inspection of the available crystal structures of AKR1C3 with multiple ligands bound, along with the crystal structures of the other AKR1C isoforms, provides a structural basis for the rational design of isoform specific inhibitors of AKR1C3. We find that there are subpockets involved in ligand binding that are considerably different in AKR1C3 relative to the closely related AKR1C1 or AKR1C2 isoforms. These pockets can be used to further improve the binding affinity and selectivity of the currently available AKR1C3 inhibitors. Article from the special issue on Targeted Inhibitors.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Isoenzimas/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Estrutura Molecular , Neoplasias/enzimologia , Esteroides/química , Esteroides/metabolismo
14.
J Am Chem Soc ; 130(43): 14123-8, 2008 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-18826220

RESUMO

We here present an optical method for monitoring the activity of the inducible aldo-keto reductases AKR1C2 and AKR1C3 in living human cells. The induction of these enzymes is regulated by the antioxidant response element (ARE), as demonstrated in recent literature, which in turn is dependent on the transcription factor Nrf2. The activation of ARE leads to the transcription of a coalition of cytoprotective enzymes and thus represents an important target for the development of new therapies in the area of neurodegenerative diseases and cancer. Through the use of Coumberone, a metabolic fluorogenic probe, and isoform-selective inhibitors, the upregulation of cellular stress markers AKR1C2 and AKR1C3 can be quantitatively measured in the presence of ARE activator compounds, via either a fluorimetric assay or fluorescence microscopy imaging of intact cells. The method has both high sensitivity and broad dynamic range, as demonstrated by induction studies in three cell lines with dramatically different metabolic capabilities (transfected monkey kidney COS-1 cells, human neuroblastoma IMR-32 cells, and human liver HepG2 cells). We applied the new method to examine a number of neurotrophic natural products (spirotenuipesine A, spirotenuipesine B, scabronine G-methylester, and panaxytriol), and discovered that panaxytriol, an active component of red ginseng extracts, is a potent ARE inducer. The upregulation of AKR1C enzymes, induced by chemically homogeneous panaxytriol, was partially dependent on PKC and PI3K kinases as demonstrated by the application of selective inhibitors. This cellular mechanism may account for panaxytriol's neurotrophic, neuroprotective, and anticancer properties. The protective effects of ARE inducers against tumorgenesis and neurodegeneration fuel the growing interest in this area of research and the method described here will greatly enable these endeavors.


Assuntos
3-Hidroxiesteroide Desidrogenases/efeitos dos fármacos , Enedi-Inos/farmacologia , Álcoois Graxos/farmacologia , Corantes Fluorescentes/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Hidroxiprostaglandina Desidrogenases/efeitos dos fármacos , Hidroxiesteroide Desidrogenases/efeitos dos fármacos , Panax/química , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Animais , Antineoplásicos/farmacologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Citoproteção , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Enedi-Inos/química , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Álcoois Graxos/química , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Hidroxiesteroide Desidrogenases/química , Hidroxiesteroide Desidrogenases/metabolismo , Indometacina/farmacologia , Microscopia de Fluorescência/métodos , Modelos Biológicos , Estrutura Molecular , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Sensibilidade e Especificidade , Regulação para Cima/efeitos dos fármacos , Ácido Ursodesoxicólico/farmacologia
15.
Nat Genet ; 40(6): 789-93, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18500342

RESUMO

Digital clubbing, recognized by Hippocrates in the fifth century BC, is the outward hallmark of pulmonary hypertrophic osteoarthropathy, a clinical constellation that develops secondary to various acquired diseases, especially intrathoracic neoplasm. The pathogenesis of clubbing and hypertrophic osteoarthropathy has hitherto been poorly understood, but a clinically indistinguishable primary (idiopathic) form of hypertrophic osteoarthropathy (PHO) is recognized. This familial disorder can cause diagnostic confusion, as well as significant disability. By autozygosity methods, we mapped PHO to chromosome 4q33-q34 and identified mutations in HPGD, encoding 15-hydroxyprostaglandin dehydrogenase, the main enzyme of prostaglandin degradation. Homozygous individuals develop PHO secondary to chronically elevated prostaglandin E(2) levels. Heterozygous relatives also show milder biochemical and clinical manifestations. These findings not only suggest therapies for PHO, but also imply that clubbing secondary to other pathologies may be prostaglandin mediated. Testing for HPGD mutations and biochemical testing for HPGD deficiency in patients with unexplained clubbing might help to obviate extensive searches for occult pathology.


Assuntos
Cromossomos Humanos Par 4/genética , Mutação da Fase de Leitura/genética , Hidroxiprostaglandina Desidrogenases/genética , Osteoartropatia Hipertrófica Primária/etiologia , Adolescente , Adulto , Sequência de Aminoácidos , Criança , Consanguinidade , Dinoprostona/urina , Feminino , Genoma Humano , Heterozigoto , Homozigoto , Humanos , Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Osteoartropatia Hipertrófica Primária/enzimologia , Osteoartropatia Hipertrófica Primária/patologia , Linhagem , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
16.
Curr Pharm Des ; 12(8): 955-62, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16533162

RESUMO

NAD(+)-linked 15-hydroxyprostaglandin dehydrogenase (15-PGDH) catalyzes the oxidation of 15(S)-hydroxyl group of prostaglandins and lipoxins resulting in the formation of 15-keto metabolites which exhibit greatly reduced biological activities. Therefore, this enzyme has been considered the key enzyme responsible for the inactivation of prostaglandins and lipoxins. Both the cDNA and the genomic DNA of the 15-PGDH gene have been cloned. Structural characterization, transcriptional regulation and biological functions of this enzyme have been investigated. Molecular modeling corroborated with site-directed mutagenesis has identified key residues and domains involved in coenzyme and substrate binding. Catalytic mechanism has been proposed. Studies on the regulation of enzyme expression and activity by physiological and pharmacological agents have begun to uncover its significant roles in cancer, inflammation and reproduction. Apparently, 15-PGDH works with cyclooxygenase-2 to control the cellular levels of prostaglandins. Their reciprocal regulation within the same cells appears to determine the fate of the cells. Because of its ability to inactivate both prostaglandins and lipoxins of two opposite biological activities, the roles of 15-PGDH in cancer and inflammation are particularly intriguing and challenging. Future investigations in these areas are warranted.


Assuntos
Hidroxiprostaglandina Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/metabolismo , Prostaglandinas/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Domínio Catalítico/genética , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiprostaglandina Desidrogenases/genética , Lipoxinas/metabolismo , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Camundongos , Camundongos Knockout , Modelos Animais , Mutagênese Sítio-Dirigida , Conformação Proteica , Especificidade por Substrato , Neoplasias da Bexiga Urinária/enzimologia , Neoplasias da Bexiga Urinária/genética
17.
Cancer Res ; 64(5): 1802-10, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14996743

RESUMO

It is becoming increasingly well established that nonsteroidal anti-inflammatory drugs (NSAID) protect against tumors of the gastrointestinal tract and that they may also protect against a variety of other tumors. These activities have been widely attributed to the inhibition of cylooxygenases (COX) and, in particular, COX-2. However, several observations have indicated that other targets may be involved. Besides targeting COX, certain NSAID also inhibit enzymes belonging to the aldo-keto reductase (AKR) family, including AKR1C3. We have demonstrated previously that overexpression of AKR1C3 acts to suppress cell differentiation and promote proliferation in myeloid cells. However, this enzyme has a broad tissue distribution and therefore represents a novel candidate for the target of the COX-independent antineoplastic actions of NSAID. Here we report on the X-ray crystal structures of AKR1C3 complexed with the NSAID indomethacin (1.8 A resolution) or flufenamic acid (1.7 A resolution). One molecule of indomethacin is bound in the active site, whereas flufenamic acid binds to both the active site and the beta-hairpin loop, at the opposite end of the central beta-barrel. Two other crystal structures (1.20 and 2.1 A resolution) show acetate bound in the active site occupying the proposed oxyanion hole. The data underline AKR1C3 as a COX-independent target for NSAID and will provide a structural basis for the future development of new cancer therapies with reduced COX-dependent side effects.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Ácido Flufenâmico/metabolismo , Hidroxiprostaglandina Desidrogenases/química , Indometacina/metabolismo , Sequência de Aminoácidos , Anti-Inflamatórios não Esteroides/farmacologia , Sítios de Ligação , Cristalização , Inibidores de Ciclo-Oxigenase/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Hidroxiprostaglandina Desidrogenases/metabolismo , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas Recombinantes/química
18.
Biochim Biophys Acta ; 1633(2): 96-105, 2003 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-12880869

RESUMO

Distinct functional coupling between cyclooxygenases (COXs) and specific terminal prostanoid synthases leads to phase-specific production of particular prostaglandins (PGs). In this study, we examined the coupling between COX isozymes and PGF synthase (PGFS). Co-transfection of COXs with PGFS-I belonging to the aldo-keto reductase family into HEK293 cells resulted in increased production of PGF(2alpha) only when a high concentration of exogenous arachidonic acid (AA) was supplied. However, this enzyme failed to produce PGF(2alpha) from endogenous AA, even though significant increase in PGF(2alpha) production occurred in cells transfected with COX-2 alone. This poor COX/PGFS-I coupling was likely to arise from their distinct subcellular localization. Measurement of PGF(2alpha)-synthetic enzyme activity in homogenates of several cells revealed another type of PGFS activity that was membrane-bound, glutathione (GSH)-activated, and stimulus-inducible. In vivo, membrane-bound PGFS activity was elevated in the lung of lipopolysaccharide-treated mice. Taken together, our results suggest the presence of a novel, membrane-associated form of PGFS that is stimulus-inducible and is likely to be preferentially coupled with COX-2.


Assuntos
Dinoprosta/biossíntese , Hidroxiprostaglandina Desidrogenases/química , Isoenzimas/química , Prostaglandina-Endoperóxido Sintases/química , Animais , Ácido Araquidônico/farmacologia , Linhagem Celular , Membrana Celular/enzimologia , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Glutationa/farmacologia , Glutationa Transferase/análise , Humanos , Hidroxiprostaglandina Desidrogenases/biossíntese , Hidroxiprostaglandina Desidrogenases/metabolismo , Oxirredutases Intramoleculares/análise , Isoenzimas/genética , Lipopolissacarídeos/administração & dosagem , Pulmão/enzimologia , Macrófagos Peritoneais/enzimologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/genética , Transfecção , Células Tumorais Cultivadas
20.
Biol Reprod ; 62(1): 125-31, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10611076

RESUMO

During reproductive processes, prostaglandin (PG) E(2) (PGE(2)) and PGF(2alpha) play important roles in which they often exert opposite effects. At the time of recognition of pregnancy in vivo, PGF(2alpha) is recognized as the luteolytic factor in ruminants and in most species including human, whereas PGE(2) may exert a luteoprotective action. We have previously demonstrated that recombinant interferon-tau (rIFN-tau), the embryonic signal responsible for recognition of pregnancy in ruminants, stimulated in vitro the production of PGE(2) and prostaglandin-endoperoxide synthase 2 (Ptgs2; also called cyclooxygenase-2) gene expression in both epithelial and stromal endometrial cells. Since PGE(2) is the major prostaglandin produced by stromal cells, the effect on Ptgs2 could explain the increase in PGE(2) output. At high concentrations, however, recombinant ovine (ro) IFN-tau acts on epithelial cells by changing the primary PG produced from PGF(2alpha) to PGE(2). This change in the primary PG produced could be explained by a decrease in PGF synthase (PGFS) activity or an increase in PGE synthase activity, or by modulation of a putative PGE(2)-9-ketoreductase, which converts PGE(2) into PGF(2alpha). Therefore, we have investigated the regulation of the mRNAs for PGFS and PGE(2)-9-ketoreductase (9K-PGR), two enzymes that lead to the production of PGF(2alpha). Others have described 9K-PGR activity in uterus, ovaries, kidney, and liver of different species and have established that this enzyme could possess both 9K-PGR and 20alpha-hydroxysteroid dehydrogenase (20alpha-HSD) activity. Some have concluded that 9K-PGR and 20alpha-HSD are identical enzymes. Using primers sequences chosen from homologous nucleotide sequences of published rabbit 20alpha-HSD/9K-PGR and rat 20alpha-HSD cDNAs, a 317-base pair (bp) fragment was amplified by reverse transcription-polymerase chain reaction (RT-PCR), cloned, and sequenced. Homologies of 83% and 78% were found with rabbit and rat 20alpha-HSD, respectively. The presence of 20alpha-HSD/9K-PGR and prostaglandin F synthase (PGFS) mRNA expression was studied semiquantitatively in cultured epithelial cells using RT-PCR. Stimulation of cells with roIFN-t resulted in a biphasic response, an inhibition of PGF(2alpha) production at low dose (1 ng/ml) and a stimulation of PGE(2) at high dose (10 microg/ml). The increase of PGE(2) was accompanied by reduced 9K-PGR and PGFS mRNA gene expression. The effect of oxytocin (OT) was also studied, and the presence of OT had no effect on either 9K-PGR or PGFS gene expression. The 20alpha-HSD/9K-PGR transcript was also detected in other bovine tissues at different intensity (liver > kidney > testis > ovaries). We believe that the 9K-PGR and PGFS can be key enzymes in the regulation of specific PGs in the endometrium during the periimplantation period.


Assuntos
Endométrio/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hidroxiprostaglandina Desidrogenases/genética , Interferon Tipo I/farmacologia , Ocitocina/farmacologia , Proteínas da Gravidez/farmacologia , RNA Mensageiro/análise , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Clonagem Molecular , Dinoprosta/biossíntese , Dinoprostona/biossíntese , Feminino , Expressão Gênica , Hidroxiprostaglandina Desidrogenases/química , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA