Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
PLoS One ; 17(10): e0271602, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36301939

RESUMO

The aberrant expression of aldo keto reductases (AKR1B1 & AKR1B10) has been extensively studied in different types of cancer especially the colon cancer but a very few studies have yet been reported regarding the discovery of inhibitors for the treatment of colon cancer by targeting these isozymes. Therefore, there is a need of selective inhibitors of both targets for the eradication of colon cancer. Currently, the study is focused on the exploration of two quinolone compounds i.e., (S)-(6-Methoxyquinolin-4-yl)[(1S,2R,4S,5R)-5-vinylquinuclidin-2-yl]methanol (Quinidine) and (R)-(6-Methoxyquinolin-4-yl)[(1S,2S,4S,5R)-5-vinylquinuclidin-2-yl]methanol (Quinine) as the potential inhibitors of AKR1B1 and AKR1B10 via detailed in-silico approach. The structural properties including vibrational frequencies, dipole moment, polarizability and the optimization energies were estimated using density functional theory (DFT) calculations; where both compounds were found chemically reactive. After that, the optimized structures were used for the molecular docking studies and here quinidine was found more selective towards AKR1B1 and quinine exhibited maximum inhibition of AKR1B10. The results of molecular docking studies were validated by molecular dynamics simulations which provided the deep insight of stability of protein ligand complex. At the end, the ADMET properties were determined to demonstrate the druglikeness properties of both selected compounds. These findings suggested further exploration of both compounds at molecular level using different in-vivo and in-vitro approaches that will lead to the designing of potential inhibitor of AKR1B1/AKR1B10 for curing colon cancer and related malignancies.


Assuntos
Aldeído Redutase , Aldo-Ceto Redutases , Neoplasias do Colo , Quinidina , Quinina , Humanos , Aldeído Redutase/antagonistas & inibidores , Aldo-Ceto Redutases/antagonistas & inibidores , Neoplasias do Colo/tratamento farmacológico , Simulação de Acoplamento Molecular , Quinidina/farmacologia , Quinina/farmacologia
2.
Molecules ; 27(13)2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35807227

RESUMO

Both members of the aldo-keto reductases (AKRs) family, AKR1B1 and AKR1B10, are over-expressed in various type of cancer, making them potential targets for inflammation-mediated cancers such as colon, lung, breast, and prostate cancers. This is the first comprehensive study which focused on the identification of phenylcarbamoylazinane-1, 2,4-triazole amides (7a−o) as the inhibitors of aldo-keto reductases (AKR1B1, AKR1B10) via detailed computational analysis. Firstly, the stability and reactivity of compounds were determined by using the Guassian09 programme in which the density functional theory (DFT) calculations were performed by using the B3LYP/SVP level. Among all the derivatives, the 7d, 7e, 7f, 7h, 7j, 7k, and 7m were found chemically reactive. Then the binding interactions of the optimized compounds within the active pocket of the selected targets were carried out by using molecular docking software: AutoDock tools and Molecular operation environment (MOE) software, and during analysis, the Autodock (academic software) results were found to be reproducible, suggesting this software is best over the MOE (commercial software). The results were found in correlation with the DFT results, suggesting 7d as the best inhibitor of AKR1B1 with the energy value of −49.40 kJ/mol and 7f as the best inhibitor of AKR1B10 with the energy value of −52.84 kJ/mol. The other potent compounds also showed comparable binding energies. The best inhibitors of both targets were validated by the molecular dynamics simulation studies where the root mean square value of <2 along with the other physicochemical properties, hydrogen bond interactions, and binding energies were observed. Furthermore, the anticancer potential of the potent compounds was confirmed by cell viability (MTT) assay. The studied compounds fall into the category of drug-like properties and also supported by physicochemical and pharmacological ADMET properties. It can be suggested that the further synthesis of derivatives of 7d and 7f may lead to the potential drug-like molecules for the treatment of colon cancer associated with the aberrant expression of either AKR1B1 or AKR1B10 and other associated malignancies.


Assuntos
Aldo-Ceto Redutases , Amidas , Neoplasias do Colo , Triazóis , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/metabolismo , Amidas/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Humanos , Simulação de Acoplamento Molecular , Triazóis/farmacologia
3.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-35163833

RESUMO

Preventing the cytokine storm observed in COVID-19 is a crucial goal for reducing the occurrence of severe acute respiratory failure and improving outcomes. Here, we identify Aldo-Keto Reductase 1B10 (AKR1B10) as a key enzyme involved in the expression of pro-inflammatory cytokines. The analysis of transcriptomic data from lung samples of patients who died from COVID-19 demonstrates an increased expression of the gene encoding AKR1B10. Measurements of the AKR1B10 protein in sera from hospitalised COVID-19 patients suggests a significant link between AKR1B10 levels and the severity of the disease. In macrophages and lung cells, the over-expression of AKR1B10 induces the expression of the pro-inflammatory cytokines Interleukin-6 (IL-6), Interleukin-1ß (IL-1ß) and Tumor Necrosis Factor a (TNFα), supporting the biological plausibility of an AKR1B10 involvement in the COVID-19-related cytokine storm. When macrophages were stressed by lipopolysaccharides (LPS) exposure and treated by Zopolrestat, an AKR1B10 inhibitor, the LPS-induced production of IL-6, IL-1ß, and TNFα is significantly reduced, reinforcing the hypothesis that the pro-inflammatory expression of cytokines is AKR1B10-dependant. Finally, we also show that AKR1B10 can be secreted and transferred via extracellular vesicles between different cell types, suggesting that this protein may also contribute to the multi-organ systemic impact of COVID-19. These experiments highlight a relationship between AKR1B10 production and severe forms of COVID-19. Our data indicate that AKR1B10 participates in the activation of cytokines production and suggest that modulation of AKR1B10 activity might be an actionable pharmacological target in COVID-19 management.


Assuntos
Aldo-Ceto Redutases/fisiologia , COVID-19/genética , Síndrome da Liberação de Citocina/genética , Síndrome do Desconforto Respiratório/genética , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Animais , COVID-19/complicações , COVID-19/metabolismo , COVID-19/patologia , Estudos de Casos e Controles , Células Cultivadas , Síndrome da Liberação de Citocina/metabolismo , Síndrome da Liberação de Citocina/patologia , Síndrome da Liberação de Citocina/virologia , Citocinas/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Gravidade do Paciente , Células RAW 264.7 , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/virologia , SARS-CoV-2/fisiologia , Transcriptoma
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166319, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34954342

RESUMO

Non-alcoholic steatohepatitis (NASH) is a clinically important spectrum of non-alcoholic fatty liver disease (NAFLD) in humans. NASH is a stage of NAFLD progression wherein liver steatosis accompanies inflammation and pro-fibrotic events. Presently, there are no approved drugs for NASH, which has become a leading cause of liver transplant worldwide. To discover novel drug targets for NASH, we analyzed a human transcriptomic NASH dataset and found Aldo-keto reductase family 1 member B10 (AKR1B10) as a significantly upregulated gene in livers of human NASH patients. Similarly murine Akr1b10 and Aldo-keto reductase family 1 member B8 (Akr1b8) gene, which is a murine ortholog of human AKR1B10, were also found to be upregulated in a mouse model of diet-induced NASH. Furthermore, pharmacological inhibitors of AKR1B10 significantly reduced the pathological features of NASH such as steatosis, inflammation and fibrosis in mouse. In addition, genetic silencing of both mouse Akr1b10 and Akr1b8 significantly reduced the expression of proinflammatory cytokines from hepatocytes. These results, thus, underscore the involvement of murine AKR1B10 and AKR1B8 in the pathogenesis of murine NASH and raise an intriguing possibility of a similar role of AKR1B10 in human NASH.


Assuntos
Oxirredutases do Álcool/metabolismo , Aldo-Ceto Redutases/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Oxirredutases do Álcool/antagonistas & inibidores , Oxirredutases do Álcool/genética , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sulindaco/uso terapêutico
5.
Biochem Pharmacol ; 192: 114710, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34339712

RESUMO

Bcr-Abl tyrosine kinase inhibitors significantly improved Philadelphia chromosome-positive leukaemia therapy. Apart from Bcr-Abl kinase, imatinib, dasatinib, nilotinib, bosutinib and ponatinib are known to have additional off-target effects that might contribute to their antitumoural activities. In our study, we identified aldo-keto reductase 1B10 (AKR1B10) as a novel target for dasatinib. The enzyme AKR1B10 is upregulated in several cancers and influences the metabolism of chemotherapy drugs, including anthracyclines. AKR1B10 reduces anthracyclines to alcohol metabolites that show less antineoplastic properties and tend to accumulate in cardiac tissue. In our experiments, clinically achievable concentrations of dasatinib selectively inhibited AKR1B10 both in experiments with recombinant enzyme (Ki = 0.6 µM) and in a cellular model (IC50 = 0.5 µM). Subsequently, the ability of dasatinib to attenuate AKR1B10-mediated daunorubicin (Daun) resistance was determined in AKR1B10-overexpressing cells. We have demonstrated that dasatinib can synergize with Daun in human cancer cells and enhance its therapeutic effectiveness. Taken together, our results provide new information on how dasatinib may act beyond targeting Bcr-Abl kinase, which may help to design new chemotherapy regimens, including those with anthracyclines.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Dasatinibe/administração & dosagem , Daunorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Inibidores de Proteínas Quinases/administração & dosagem , Células A549 , Aldo-Ceto Redutases/química , Aldo-Ceto Redutases/metabolismo , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteínas de Fusão bcr-abl/química , Proteínas de Fusão bcr-abl/metabolismo , Células HCT116 , Humanos , Simulação de Acoplamento Molecular , Estrutura Secundária de Proteína
6.
J Pharmacol Sci ; 147(1): 1-8, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34294359

RESUMO

Tyrosine kinase inhibitors (TKIs) are widely utilized in clinical practice to treat carcinomas, but secondary tumor resistance during chronic treatment can be problematic. AKR1B1 and AKR1B10 of the aldo-keto reductase (AKR) superfamily are highly expressed in cancer cells and are believed to be involved in drug resistance. The aim of this study was to understand how TKI treatment of chronic myelogenous leukemia (CML) cells changes their glucose metabolism and if inhibition of AKRs can sensitize CML cells to TKIs. K562 cells were treated with the TKIs imatinib, nilotinib, or bosutinib, and the effects on glucose metabolism, cell death, glutathione levels, and AKR levels were assessed. To assess glucose dependence, cells were cultured in normal and low-glucose media. Pretreatment with AKR inhibitors, including epalrestat, were used to determine AKR-dependence. Treatment with TKIs increased intracellular glucose, AKR1B1/10 levels, glutathione oxidation, and nuclear translocation of nuclear factor erythroid 2-related factor 2, but with minimal cell death. These effects were dependent on intracellular glucose accumulation. Pretreatment with epalrestat, or a selective inhibitor of AKR1B10, exacerbated TKI-induced cell death, suggesting that especially AKR1B10 was involved in protection against TKIs. Thus, by disrupting cell protective mechanisms, AKR inhibitors may render CML more susceptible to TKI treatments.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Aldeído Redutase , Aldo-Ceto Redutases/metabolismo , Aldo-Ceto Redutases/fisiologia , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Glucose/metabolismo , Humanos , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Rodanina/análogos & derivados , Rodanina/farmacologia , Rodanina/uso terapêutico , Tiazolidinas/farmacologia , Tiazolidinas/uso terapêutico
7.
Eur J Pharmacol ; 883: 173167, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32485245

RESUMO

Abnormal vitamin A (retinol) metabolism plays an important role in the occurrence of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). In this study, NAFLD and NASH models were established to investigate the effects of food additives glycyrrhizic acid (GL) on retinol metabolism in NAFLD/NASH mice. Potential targets of GL and its active metabolite glycyrrhetinic acid (GA) were analyzed by RNA sequence, bioinformatics, and molecular docking analyses. Gene transfection and enzymatic kinetics were used to identify the target of GL. The results showed that GL could resolve the fatty and inflammatory lesions in the mouse liver, thereby improving the disorder of retinol metabolism. RNA sequence analysis of model mice liver revealed significant changes in AKR1B10 (retinol metabolic enzymes). Bioinformatics and molecular docking analyses showed that AKR1B10 is a potential target of GA but not GL. GA could inhibit AKR1B10 activity, which then affects retinol metabolism, whereas GL only had the same effect after hydrolysis into GA. In AKR1B10-KO hepatocytes, GA, GL, and hydrolysates of GL had no regulatory effect on retinol metabolism. Therefore, GA, the active metabolite of GL, as a novel AKR1B10 inhibitor, could promote retinoic acid synthesis. GL restored the balance of retinol metabolism in NAFLD/NASH mice by metabolizing to GA.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ácido Glicirretínico/farmacologia , Ácido Glicirrízico/farmacologia , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Vitamina A/metabolismo , Aldo-Ceto Redutases/genética , Aldo-Ceto Redutases/metabolismo , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Ácido Glicirretínico/metabolismo , Ácido Glicirrízico/metabolismo , Células Hep G2 , Humanos , Hidrólise , Cinética , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/patologia
8.
Org Biomol Chem ; 18(25): 4788-4801, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32530010

RESUMO

Human aldo-keto reductases (AKRs) are enzymes involved in the reduction, among other substrates, of all-trans-retinal to all-trans-retinol (vitamin A), thus contributing to the control of the levels of retinoids in organisms. Structure-activity relationship studies of a series of C11-to-C14 methyl-shifted (relative to natural C13-methyl) all-trans-retinal analogues as putative substrates of AKRs have been reported. The synthesis of these retinoids was based on the formation of a C10-C11 single bond of the pentaene skeleton starting from a trienyl iodide and the corresponding dienylstannanes and dienylsilanes, using the Stille-Kosugi-Migita and Hiyama-Denmark cross-coupling reactions, respectively. Since these reagents differ by the location and presence of methyl groups at the dienylorganometallic fragment, the study also provided insights into the ability of the different positional isomers to undergo cross-coupling and the sensitivity of these processes to steric hindrance. The resulting C11-to-C14 methyl-shifted all-trans-retinal analogues were found to be active substrates when tested with AKR1B1 and AKR1B10 enzymes, although relevant differences in substrate specificities were noted. For AKR1B1, all analogues exhibited higher catalytic efficiency (kcat/Km) than parent all-trans-retinal. In addition, only all-trans-11-methylretinal, the most hydrophobic derivative, showed a higher value of kcat/Km = 106 000 ± 23 200 mM-1 min-1 for AKR1B10, which is in fact the highest value from all known retinoid substrates of this enzyme. The novel structures, identified as efficient AKR substrates, may serve in the design of selective inhibitors with potential pharmacological interest.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Tretinoína/farmacologia , Aldo-Ceto Redutases/metabolismo , Humanos , Estrutura Molecular , Tretinoína/síntese química , Tretinoína/química
9.
FEBS Lett ; 594(3): 564-580, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31573681

RESUMO

Tolrestat and epalrestat have been characterized as noncompetitive inhibitors of aldo-ketone reductase 1B1 (AKR1B1), a leading drug target for the treatment of type 2 diabetes complications. However, clinical applications are limited for most AKR1B1 inhibitors due to adverse effects of cross-inhibition with other AKRs. Here, we report an atypical competitive binding and inhibitory effect of tolrestat on the thermostable AKR Tm1743 from Thermotoga maritima. Analysis of the Tm1743 crystal structure in complex with tolrestat alone and epalrestat-NADP+ shows that tolrestat, but not epalrestat, binding triggers dramatic conformational changes in the anionic site and cofactor binding pocket that prevents accommodation of NADP+ . Enzymatic and molecular dynamics simulation analyses further confirm tolrestat as a competitive inhibitor of Tm1743.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Naftalenos/farmacologia , Temperatura , Thermotoga maritima/enzimologia , Aldo-Ceto Redutases/química , Aldo-Ceto Redutases/metabolismo , Sítios de Ligação , Inibidores Enzimáticos/metabolismo , Estabilidade Enzimática , Simulação de Dinâmica Molecular , Naftalenos/metabolismo , Conformação Proteica
10.
Cell Death Dis ; 10(12): 902, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31780644

RESUMO

The incidence of melanoma is increasing over the years with a still poor prognosis and the lack of a cure able to guarantee an adequate survival of patients. Although the new immuno-based coupled to target therapeutic strategy is encouraging, the appearance of targeted/cross-resistance and/or side effects such as autoimmune disorders could limit its clinical use. Alternative therapeutic strategies are therefore urgently needed to efficiently kill melanoma cells. Ferroptosis induction and execution were evaluated in metastasis-derived wild-type and oncogenic BRAF melanoma cells, and the process responsible for the resistance has been dissected at molecular level. Although efficiently induced in all cells, in an oncogenic BRAF- and ER stress-independent way, most cells were resistant to ferroptosis execution. At molecular level we found that: resistant cells efficiently activate NRF2 which in turn upregulates the early ferroptotic marker CHAC1, in an ER stress-independent manner, and the aldo-keto reductases AKR1C1 ÷ 3 which degrades the 12/15-LOX-generated lipid peroxides thus resulting in ferroptotic cell death resistance. However, inhibiting AKRs activity/expression completely resensitizes resistant melanoma cells to ferroptosis execution. Finally, we found that the ferroptotic susceptibility associated with the differentiation of melanoma cells cannot be applied to metastatic-derived cells, due to the EMT-associated gene expression reprogramming process. However, we identified SCL7A11 as a valuable marker to predict the susceptibility of metastatic melanoma cells to ferroptosis. Our results identify the use of pro-ferroptotic drugs coupled to AKRs inhibitors as a new valuable strategy to efficiently kill human skin melanoma cells.


Assuntos
Aldo-Ceto Redutases/metabolismo , Estresse do Retículo Endoplasmático , Ferroptose , Melanoma/enzimologia , Melanoma/patologia , Aldo-Ceto Redutases/antagonistas & inibidores , Araquidonato 15-Lipoxigenase/metabolismo , Biomarcadores Tumorais/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ferroptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxidos Lipídicos/metabolismo , Melanoma/genética , Fator 2 Relacionado a NF-E2/metabolismo , Metástase Neoplásica , Oncogenes , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Regulação para Cima/efeitos dos fármacos , gama-Glutamilciclotransferase/metabolismo
11.
Arch Biochem Biophys ; 674: 108096, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31479646

RESUMO

A mouse gene, Akr1cl, encodes a member of the aldo-keto reductase 1C subfamily (AKR1CL), whose function, however, remains unknown. Here, we show that the recombinant AKR1CL is an NADPH-dependent reductase of prostaglandin (PG) D2 (Km 3.2 µM, kcat 5.6 min-1) and oxidizes 9α,11ß-PGF2 (Km 30 µM, kcat 3.3 min-1) in the reverse reaction. In contrast, it did not exhibit oxidoreductase activity towards other PGs (E2, A1, B2 and F2α), steroids and nonsteroidal carbonyls and alcohols, which are substrates of other mammalian AKR1C subfamily enzymes. The enzyme activity was inhibited by estradiol, quercetin, benzbromarone, ethacrynic acid and flufenamic acid, of which estradiol was the most potent competitive inhibitor (Ki 3.2 µM). The mRNA for AKR1CL was expressed abundantly in mouse testis, ovary and adrenal gland, and at low levels in the brain, lung, small intestine and prostate. Thus, AKR1CL is the first PGD2 11-ketoreductase with strict substrate specificity in mammals. The site-directed mutagenesis of P85 in AKR1CL to the corresponding residue, W, in other mammalian AKR1C subfamily enzymes resulted in broad substrate specificity for nonsteroidal carbonyls and alcohols, suggesting that P85 plays a critical role in the strict specificity for PGD2 and 9α,11ß-PGF2.


Assuntos
Aldo-Ceto Redutases/metabolismo , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Sequência de Aminoácidos , Animais , Inibidores Enzimáticos/química , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Especificidade por Substrato/genética , Distribuição Tecidual
12.
J Agric Food Chem ; 67(30): 8382-8392, 2019 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-31271032

RESUMO

Staphylococcus aureus is a common pathogen that can cause life-threatening infections. Treatment of antibiotic-resistant S. aureus infection needs effective antibacterial agents. Thymol, a generally recognized safe natural compound, has potential as an alternative to treat S. aureus infections. However, the targets and mechanisms of action of thymol were not fully understood. Bioinformatics analysis showed that IolS, a predicted aldo-keto reductase (AKR) in S. aureus, could be a potential target of thymol. Isothermal titration calorimetry (ITC) analysis demonstrated that thymol directly binds IolS and amino acid residues (Y30 and L33) are essential for such binding. Deletion of IolS or mutation of Y30A and L33A reduced the bactericidal activity of thymol at the concentration of 200 µg/mL, suggesting that thymol mediates bactericidal activity via binding with IolS. Biochemical analysis showed that addition of thymol significantly increased AKR activity of IolS from 1.6 ± 0.1 to 2.4 ± 0.2 U (p < 0.05). The content of NADPH within S. aureus cells decreased significantly from 105 ± 5 to 72 ± 3 pmol/108 cells (p < 0.05) following thymol treatment at the concentration of 200 µg/mL. Importantly, addition of NADPH could alleviate the bactericidal effect of thymol on S. aureus, indicating that the depletion of NADPH is responsible for thymol-mediated bactericidal activity. Overall, these results demonstrated that thymol could directly bind IolS and increase its AKR activity, leading to the depletion of NADPH and bactericidal effect. AKR activity of IolS could be a promising target for the development of new antimicrobials.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , NADP/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia , Timol/farmacologia , Aldo-Ceto Redutases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
13.
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
14.
Biochem Pharmacol ; 163: 391-403, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30862505

RESUMO

E-cadherin transcriptional activator EP300 is down-regulated in metaplastic breast carcinoma, a rare form of triple negative and E-cadherin-negative aggressive breast cancer with a poor clinical outcome. In order to shed light on the regulation of E-cadherin by EP300 in breast cancer we analyzed by immunohistochemistry 41 cases of invasive breast cancer with both E-cadherinhigh and E-cadherinlow expression levels, together with 20 non-malignant breast tissues. EP300 and E-cadherin showed a positive correlation in both non-malignant and cancer cases and both markers together were better predictors of lymph node metastasis than E-cadherin alone. These data support a metastasis suppressor role for EP300 in breast cancer. However, some reports suggest an oncogenic role for EP300. We generated a breast cancer cell model to study E-cadherin-independent effects of EP300 by over-expression of EP300 in HS578T cells which have E-cadherin promoter hypermethylated. In this cell system, EP300 led to up-regulation of mesenchymal (vimentin, Snail, Slug, Zeb1) and stemness (ALDH+ and CD44high/CD24low) markers, increases in migration, invasion, anchorage-independent growth and drug resistance. Genome-wide expression profiling identified aldo-keto reductases AKR1C1-3 as effectors of stemness and drug resistance, since their pharmacological inhibition with flufenamic acid restored both doxorubicin and paclitaxel sensitivity and diminished mammosphere formation. Thus, in cells with a permissive E-cadherin promoter, EP300 acts as a tumour/metastasis supressor by up-regulating E-cadherin expression, maintenance of the epithelial phenotype and avoidance of an epithelial-to-mesenchymal transition. In cells in which the E-cadherin promoter is hypermethylated, EP300 functions as an oncogene via up-regulation of aldo-keto reductases. This offers the rationale of using current aldo-keto reductase inhibitors in breast cancer treatment.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Neoplasias da Mama/enzimologia , Proteína p300 Associada a E1A/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antineoplásicos/farmacologia , Biomarcadores Tumorais , Caderinas , Linhagem Celular Tumoral , Movimento Celular , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Imuno-Histoquímica , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Paclitaxel/farmacologia
15.
Chem Biol Interact ; 302: 101-107, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30703376

RESUMO

Buparlisib is a pan-class I phosphoinositide 3-kinase (PI3K) inhibitor and is currently under clinical evaluation for the treatment of different cancers. Because PI3K signalling is related to cell proliferation and resistance to chemotherapy, new therapeutic approaches are focused on combining PI3K inhibitors with other anti-cancer therapeutics. Carbonyl-reducing enzymes catalyse metabolic detoxification of anthracyclines and reduce their cytotoxicity. In the present work, the effects of buparlisib were tested on six human recombinant carbonyl-reducing enzymes: AKR1A1, AKR1B1, AKR1B10, AKR1C3, and AKR7A2 from the aldo-keto reductase superfamily and CBR1 from the short-chain dehydrogenase/reductase superfamily, all of which participate in the metabolism of daunorubicin. Buparlisib exhibited the strongest inhibitory effect on recombinant AKR1C3, with a half-maximal inhibitory concentration (IC50) of 9.5 µM. Its inhibition constant Ki was found to be 14.0 µM, and the inhibition data best fitted a mixed-type mode with α = 0.6. The same extent of inhibition was observed at the cellular level in the human colorectal carcinoma HCT 116 cell line transfected with a plasmid encoding the AKR1C3 transcript (IC50 = 7.9 µM). Furthermore, we performed an analysis of flexible docking between buparlisib and AKR1C3 and found that buparlisib probably occupies a part of the binding site for a cofactor most likely via the trifluoromethyl group of buparlisib interacting with catalytic residue Tyr55. In conclusion, our results show a novel PI3K-independent effect of buparlisib that may improve therapeutic efficacy and safety of daunorubicin by preventing its metabolism by AKR1C3.


Assuntos
Membro C3 da Família 1 de alfa-Ceto Redutase/metabolismo , Aminopiridinas/farmacologia , Daunorrubicina/metabolismo , Morfolinas/farmacologia , Membro C3 da Família 1 de alfa-Ceto Redutase/antagonistas & inibidores , Membro C3 da Família 1 de alfa-Ceto Redutase/química , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Aldo-Ceto Redutases/metabolismo , Aminopiridinas/química , Aminopiridinas/metabolismo , Sítios de Ligação , Domínio Catalítico , Células HCT116 , Humanos , Concentração Inibidora 50 , Cinética , Simulação de Acoplamento Molecular , Morfolinas/química , Morfolinas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
16.
Zhonghua Nan Ke Xue ; 25(8): 734-738, 2019 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-32227718

RESUMO

The androgen receptor signaling pathway is a key factor in the development and progression of prostate cancer. Aldo-keto reductases AKR1C1-AKR1C4 play an important role in the synthesis and metabolism of androgens in the body, and their expressions influence the androgen receptor signaling pathway and consequently the development and progression of prostate cancer. For the treatment of androgen-resistant prostate cancer, which cannot be cured currently, Chinese medicine and phytotherapy are receiving more and more attention for the mild, long-lasting and multi-target advantages of the small molecules of traditional Chinese medicine. This review summarizes the roles of aldo-keto reductases in the progression of prostate cancer and compares the anti-tumor activities of small molecules in Chinese medicine targeting aldo-keto reductases, hoping to provide a basis for the discovery of new targets for prostate cancer and the development of anti-tumor drugs.


Assuntos
Aldo-Ceto Redutases , Medicina Tradicional Chinesa , Neoplasias de Próstata Resistentes à Castração/enzimologia , Aldo-Ceto Redutases/antagonistas & inibidores , Androgênios , Humanos , Masculino
17.
Br J Cancer ; 118(7): 985-994, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29515258

RESUMO

BACKGROUND: Despite chemotherapy intensification, a subgroup of high-risk paediatric T-cell acute lymphoblastic leukemia (T-ALL) patients still experience treatment failure. In this context, we hypothesised that therapy resistance in T-ALL might involve aldo-keto reductase 1C (AKR1C) enzymes as previously reported for solid tumors. METHODS: Expression of NRF2-AKR1C signaling components has been analysed in paediatric T-ALL samples endowed with different treatment outcomes as well as in patient-derived xenografts of T-ALL. The effects of AKR1C enzyme modulation has been investigated in T-ALL cell lines and primary cultures by combining AKR1C inhibition, overexpression, and gene silencing approaches. RESULTS: We show that T-ALL cells overexpress AKR1C1-3 enzymes in therapy-resistant patients. We report that AKR1C1-3 enzymes play a role in the response to vincristine (VCR) treatment, also ex vivo in patient-derived xenografts. Moreover, we demonstrate that the modulation of AKR1C1-3 levels is sufficient to sensitise T-ALL cells to VCR. Finally, we show that T-ALL chemotherapeutics induce overactivation of AKR1C enzymes independent of therapy resistance, thus establishing a potential resistance loop during T-ALL combination treatment. CONCLUSIONS: Here, we demonstrate that expression and activity of AKR1C enzymes correlate with response to chemotherapeutics in T-ALL, posing AKR1C1-3 as potential targets for combination treatments during T-ALL therapy.


Assuntos
Aldo-Ceto Redutases/fisiologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , 20-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 20-Hidroxiesteroide Desidrogenases/fisiologia , Idade de Início , Membro C3 da Família 1 de alfa-Ceto Redutase/antagonistas & inibidores , Membro C3 da Família 1 de alfa-Ceto Redutase/fisiologia , Aldo-Ceto Redutases/antagonistas & inibidores , Animais , Criança , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Hidroxiesteroide Desidrogenases/fisiologia , Isoenzimas/fisiologia , Acetato de Medroxiprogesterona/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Oxirredutases/antagonistas & inibidores , Oxirredutases/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/epidemiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Células Tumorais Cultivadas , Vincristina/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Enzyme Inhib Med Chem ; 33(1): 607-614, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29532688

RESUMO

Xanthohumol (XN), a prenylated chalcone unique to hops (Humulus lupulus) and two derived prenylflavanones, isoxanthohumol (IX) and 8-prenylnaringenin (8-PN) gained increasing attention as potential anti-diabetic and cancer preventive compounds. Two enzymes of the aldo-keto reductase (AKR) superfamily are notable pharmacological targets in cancer therapy (AKR1B10) and in the treatment of diabetic complications (AKR1B1). Our results show that XN, IX and 8-PN are potent uncompetitive, tight-binding inhibitors of human aldose reductase AKR1B1 (Ki = 15.08 µM, 0.34 µM, 0.71 µM) and of human AKR1B10 (Ki = 20.11 µM, 2.25 µM, 1.95 µM). The activity of the related enzyme AKR1A1 was left unaffected by all three compounds. This is the first time these three substances have been tested on AKRs. The results of this study may provide a basis for further quantitative structure?activity relationship models and promising scaffolds for future anti-diabetic or carcinopreventive drugs.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Flavanonas/farmacologia , Flavonoides/farmacologia , Humulus/química , Propiofenonas/farmacologia , Xantonas/farmacologia , Aldo-Ceto Redutases/metabolismo , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Flavanonas/química , Flavonoides/química , Humanos , Estrutura Molecular , Propiofenonas/química , Relação Estrutura-Atividade , Xantonas/química
19.
J Med Chem ; 60(20): 8441-8455, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-28976752

RESUMO

Aldo-keto reductase 1B10 (AKR1B10) is overexpressed in several extraintestinal cancers, particularly in non-small-cell lung cancer, where AKR1B10 is a potential diagnostic marker and therapeutic target. Selective AKR1B10 inhibitors are required because compounds should not inhibit the highly related aldose reductase that is involved in monosaccharide and prostaglandin metabolism. Currently, 7-hydroxy-2-(4-methoxyphenylimino)-2H-chromene-3-carboxylic acid benzylamide (HMPC) is known to be the most potent competitive inhibitor of AKR1B10, but it is nonselective. In this study, derivatives of HMPC were synthesized by removing the 4-methoxyphenylimino moiety and replacing the benzylamide with phenylpropylamide. Among them, 4c and 4e showed higher AKR1B10 inhibitory potency (IC50 4.2 and 3.5 nM, respectively) and selectivity than HMPC. The treatments with the two compounds significantly suppressed not only migration, proliferation, and metastasis of lung cancer A549 cells but also metastatic and invasive potentials of cisplatin-resistant A549 cells.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Neoplasias Pulmonares/patologia , Metástase Neoplásica/patologia , Células A549 , Aldo-Ceto Redutases/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias Pulmonares/enzimologia , Mutação
20.
Invest New Drugs ; 35(3): 375-385, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28283780

RESUMO

Anthracycline antibiotics (ANT), such as doxorubicin or daunorubicin, are a class of anticancer drugs that are widely used in oncology. Although highly effective in cancer therapy, their usefulness is greatly limited by their cardiotoxicity. Possible mechanisms of ANT cardiotoxicity include their conversion to secondary alcohol metabolites (i.e. doxorubicinol, daunorubicinol) catalyzed by carbonyl reductases (CBR) and aldo-keto reductases (AKR). These metabolites are suspected to be more cardiotoxic than their parent compounds. Moreover, overexpression of ANT-reducing enzymes (CBR and AKR) are found in many ANT-resistant cancers. The secondary metabolites show decreased cytotoxic properties and are more susceptible to ABC-mediated efflux than their parent compounds; thus, metabolite formation is considered one of the mechanisms of cancer resistance. Inhibitors of CBR and AKR were found to reduce the cardiotoxicity of ANT and the resistance of cancer cells, and therefore are being investigated as prospective cardioprotective and chemosensitizing drug candidates. In this review, the significance of a two-electron reduction of ANT, including daunorubicin, epirubicin, idarubicin, valrubicin, amrubicin, aclarubicin, and especially doxorubicin, is described with respect to toxicity and efficacy of therapy. Additionally, CBR and AKR inhibitors, including monoHER, curcumin, (-)-epigallocatechin gallate, resveratrol, berberine or pixantrone, and their modulating effect on the activity of ANT is characterized and discussed as potential mechanism of action for novel therapeutics in cancer treatment.


Assuntos
Aldo-Ceto Redutases/antagonistas & inibidores , Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Carbonil Redutase (NADPH)/antagonistas & inibidores , Cardiotônicos/farmacologia , Cardiotoxicidade/prevenção & controle , Animais , Antraciclinas/farmacocinética , Antibióticos Antineoplásicos/farmacocinética , Cardiotoxicidade/etiologia , Resistencia a Medicamentos Antineoplásicos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA