Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biomolecules ; 14(4)2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38672520

RESUMEN

Ethyl (S)-4-chloro-3-hydroxybutyrate ((S)-CHBE) is an important chiral intermediate in the synthesis of the cholesterol-lowering drug atorvastatin. Studying the use of SpyTag/SpyCatcher and SnoopTag/SnoopCatcher systems for the asymmetric reduction reaction and directed coupling coenzyme regeneration is practical for efficiently synthesizing (S)-CHBE. In this study, Spy and Snoop systems were used to construct a double-enzyme directed fixation system of carbonyl reductase (BsCR) and glucose dehydrogenase (BsGDH) for converting 4-chloroacetoacetate (COBE) to (S)-CHBE and achieving coenzyme regeneration. We discussed the enzymatic properties of the immobilized enzyme and the optimal catalytic conditions and reusability of the double-enzyme immobilization system. Compared to the free enzyme, the immobilized enzyme showed an improved optimal pH and temperature, maintaining higher relative activity across a wider range. The double-enzyme immobilization system was applied to catalyze the asymmetric reduction reaction of COBE, and the yield of (S)-CHBE reached 60.1% at 30 °C and pH 8.0. In addition, the double-enzyme immobilization system possessed better operational stability than the free enzyme, and maintained about 50% of the initial yield after six cycles. In summary, we show a simple and effective strategy for self-assembling SpyCatcher/SnoopCatcher and SpyTag/SnoopTag fusion proteins, which inspires building more cascade systems at the interface. It provides a new method for facilitating the rapid construction of in vitro immobilized multi-enzyme complexes from crude cell lysate.


Asunto(s)
Enzimas Inmovilizadas , Glucosa 1-Deshidrogenasa , Glucosa 1-Deshidrogenasa/metabolismo , Glucosa 1-Deshidrogenasa/química , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Biocatálisis , Concentración de Iones de Hidrógeno , Hidroxibutiratos/química , Temperatura , Catálisis , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , Carbonil Reductasa (NADPH)/metabolismo , Carbonil Reductasa (NADPH)/química
2.
Cancer Res Commun ; 4(1): 134-151, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38112643

RESUMEN

Wnt ligand WNT4 is critical in female reproductive tissue development, with WNT4 dysregulation linked to related pathologies including breast cancer (invasive lobular carcinoma, ILC) and gynecologic cancers. WNT4 signaling in these contexts is distinct from canonical Wnt signaling yet inadequately understood. We previously identified atypical intracellular activity of WNT4 (independent of Wnt secretion) regulating mitochondrial function, and herein examine intracellular functions of WNT4. We further examine how convergent mechanisms of WNT4 dysregulation impact cancer metabolism. In ILC, WNT4 is co-opted by estrogen receptor α (ER) via genomic binding in WNT4 intron 1, while in gynecologic cancers, a common genetic polymorphism (rs3820282) at this ER binding site alters WNT4 regulation. Using proximity biotinylation (BioID), we show canonical Wnt ligand WNT3A is trafficked for secretion, but WNT4 is localized to the cytosol and mitochondria. We identified DHRS2, mTOR, and STAT1 as putative WNT4 cytosolic/mitochondrial signaling partners. Whole metabolite profiling, and integrated transcriptomic data, support that WNT4 mediates metabolic reprogramming via fatty acid and amino acid metabolism. Furthermore, ovarian cancer cell lines with rs3820282 variant genotype are WNT4 dependent and have active WNT4 metabolic signaling. In protein array analyses of a cohort of 103 human gynecologic tumors enriched for patient diversity, germline rs3820282 genotype is associated with metabolic remodeling. Variant genotype tumors show increased AMPK activation and downstream signaling, with the highest AMPK signaling activity in variant genotype tumors from non-White patients. Taken together, atypical intracellular WNT4 signaling, in part via genetic dysregulation, regulates the distinct metabolic phenotypes of ILC and gynecologic cancers. SIGNIFICANCE: WNT4 regulates breast and gynecologic cancer metabolism via a previously unappreciated intracellular signaling mechanism at the mitochondria, with WNT4 mediating metabolic remodeling. Understanding WNT4 dysregulation by estrogen and genetic polymorphism offers new opportunities for defining tumor biology, precision therapeutics, and personalized cancer risk assessment.


Asunto(s)
Neoplasias de la Mama , Neoplasias de los Genitales Femeninos , Humanos , Femenino , Ligandos , Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de los Genitales Femeninos/genética , Transducción de Señal , Neoplasias de la Mama/genética , Proteína Wnt4/genética , Carbonil Reductasa (NADPH)/metabolismo
3.
Cell Metab ; 35(11): 1897-1914.e11, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37858332

RESUMEN

Genetic studies have identified numerous loci associated with type 2 diabetes (T2D), but the functional roles of many loci remain unexplored. Here, we engineered isogenic knockout human embryonic stem cell lines for 20 genes associated with T2D risk. We examined the impacts of each knockout on ß cell differentiation, functions, and survival. We generated gene expression and chromatin accessibility profiles on ß cells derived from each knockout line. Analyses of T2D-association signals overlapping HNF4A-dependent ATAC peaks identified a likely causal variant at the FAIM2 T2D-association signal. Additionally, the integrative association analyses identified four genes (CP, RNASE1, PCSK1N, and GSTA2) associated with insulin production, and two genes (TAGLN3 and DHRS2) associated with ß cell sensitivity to lipotoxicity. Finally, we leveraged deep ATAC-seq read coverage to assess allele-specific imbalance at variants heterozygous in the parental line and identified a single likely functional variant at each of 23 T2D-association signals.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Madre Embrionarias Humanas , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células Secretoras de Insulina/metabolismo , Polimorfismo de Nucleótido Simple , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo
4.
Tissue Cell ; 82: 102078, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37060745

RESUMEN

Prostate cancer (PCa) is a prevalent cause of morbidity and mortality. DHRS2-modified human umbilical cord mesenchymal stem cells-derived exosomes (hUC-MSCs-derived exos) function in PCa. We explored the mechanism of DHRS2-modified hUC-MSCs-derived exos in PCa cell malignant behaviors. DHRS2 expression levels in WPMY-1 cells and 4 PCa cell lines were detected by RT-qPCR and Western blot. 22Rv1/DU145 cells with high/low DHRS2 expression were selected to establish the low/high DHRS2 expression models by transfection. Cell proliferation and apoptosis were detected by CCK-8, colony formation assays, and flow cytometry. hUC-MSCs were identified by oil red O, alizarin staining, and flow cytometry. Exos were extracted from hUC-MSCs by ultracentrifugation and identified by transmission electron microscopy, Nano series-Nano-ZS, and Western blot. DU145 cells were selected for in vitro study to further study the effects of DHRS2-modified exos on cell proliferation and apoptosis. The effect of DHRS2-modified exos on cell cycle distribution was detected by flow cytometry. DHRS2 was repressed in PCa cells. DHRS2 overexpression suppressed PCa cell proliferation and promoted apoptosis. Exos were successfully isolated from hUC-MSC. DHRS2-modified hUC-MSCs-derived exos carried DHRS2 into PCa cells and blocked malignant behaviors. Briefly, DHRS2 was repressed in PCa cells. DHRS2-modified hUC-MSCs-derived exos blocked PCa cell proliferation and enhanced apoptosis.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Neoplasias de la Próstata , Masculino , Humanos , Exosomas/metabolismo , Apoptosis/genética , Proliferación Celular/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Cordón Umbilical , Carbonil Reductasa (NADPH)/metabolismo
5.
Biochem Biophys Res Commun ; 663: 41-46, 2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37119764

RESUMEN

Renal cell carcinoma (RCC), also known as kidney cancer, is a common malignant tumor of the urinary system. While surgical treatment is essential, novel therapeutic targets and corresponding drugs for RCC are still needed due to the high relapse rate and low five-year survival rate. In this study, we found that SUV420H2 is overexpressed in renal cancers and that high SUV420H2 expression is associated with a poor prognosis, as evidenced by RCC RNA-seq results derived from the TCGA. SUV420H2 knockdown using siRNA led to growth suppression and cell apoptosis in the A498 cell line. Furthermore, we identified DHRS2 as a direct target of SUV420H2 in the apoptosis process through a ChIP assay with a histone 4 lysine 20 (H4K20) trimethylation antibody. Rescue experiments showed that cotreatment with siSUV420H2 and siDHRS2 attenuated cell growth suppression induced by SUV420H2 knockdown only. Additionally, treatment with the SUV420H2 inhibitor A-196 induced cell apoptosis via upregulation of DHRS2. Taken together, our findings suggest that SUV420H2 may be a potential therapeutic target for the treatment of renal cancer.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Recurrencia Local de Neoplasia/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proliferación Celular , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo
6.
Anticancer Drugs ; 33(10): 1058-1068, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36206098

RESUMEN

Long noncoding RNAs (lncRNAs) have been demonstrated to participate in various biological processes and play key roles in tumorigenesis and metastasis. Pituitary adenoma (PA) is one of the most common malignancies in central nervous system. Recently, multiple lncRNAs have been identified to regulate PA initiation, progression and metastasis. we aimed to elucidate the expression pattern and function of lncRNA MYMLR in PA development. The expression of lncRNA MYMLR in PA tissues and cells was examined by real-time quantitative PCR. Knockdown of MYMLR expression was achieved by using shRNA. The function of MYMLR and regulatory network were analyzed using CCK-8 assay, wound-healing assay, migration assay and Annexin V/PI staining. Xenograft tumor model was used to explore the function of MYMLR in vivo . Bioinformatics analysis and luciferase reporter assay were conducted to investigate the interaction between MYMLR and its regulatory network. LncRNA MYMLR was highly expressed in PA tissues compared with that in normal tissues. Knockdown of MYMLR suppressed cell proliferation, migration and invasion, while promoting PA cell apoptosis. Mechanistically, MYMLR functioned as a competing endogenous RNA (ceRNA) sponging microRNA miR-197-3p. Furthermore, miR-197-3p exerted its tumor inhibitory role via negatively regulating carbonyl reductase 1 (CBR1). Overexpression of CBR1 antagonized the inhibitory effect of lncRNA MYMLR knockdown or miR-197-3p overexpression. In addition, xenograft tumor model revealed that knockdown of lncRNA MYMLR suppressed PA tumor development in vivo via regulating CBR1. Our findings suggest a regulatory network of lncRNA MYMLR/miR-197-3p/CBR1, which benefits the understanding of PA development and provides a promising lncRNA-direct therapeutic strategy against PA.


Asunto(s)
Carbonil Reductasa (NADPH) , MicroARNs , Neoplasias Hipofisarias , ARN Largo no Codificante , Humanos , Anexina A5/genética , Anexina A5/metabolismo , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Hipofisarias/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño , Animales
7.
Cell Death Dis ; 13(10): 845, 2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36192391

RESUMEN

The short-chain dehydrogenase/reductase (SDR) superfamily has essential roles in lipid metabolism and redox sensing. In recent years, accumulating evidence highlights the emerging association between SDR family enzymes and cancer. Dehydrogenase/reductase member 2(DHRS2) belongs to the NADH/NADPH-dependent SDR family, and extensively participates in the regulation of the proliferation, migration, and chemoresistance of cancer cells. However, the underlying mechanism has not been well defined. In the present study, we have demonstrated that DHRS2 inhibits the growth and metastasis of ovarian cancer (OC) cells in vitro and in vivo. Mechanistically, the combination of transcriptome and metabolome reveals an interruption of choline metabolism by DHRS2. DHRS2 post-transcriptionally downregulates choline kinase α (CHKα) to inhibit AKT signaling activation and reduce phosphorylcholine (PC)/glycerophosphorylcholine (GPC) ratio, impeding choline metabolism reprogramming in OC. These actions mainly account for the tumor-suppressive role of DHRS2 in OC. Overall, our findings establish the mechanistic connection among metabolic enzymes, metabolites, and the malignant phenotype of cancer cells. This could result in further development of novel pharmacological tools against OC by the induction of DHRS2 to disrupt the choline metabolic pathway.


Asunto(s)
Colina Quinasa , Neoplasias Ováricas , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular , Colina/metabolismo , Colina Quinasa/genética , Colina Quinasa/metabolismo , Regulación hacia Abajo , Femenino , Glicerilfosforilcolina/metabolismo , Humanos , NAD/metabolismo , NADP/metabolismo , Neoplasias Ováricas/genética , Oxidorreductasas/genética , Fosforilcolina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
Eur J Pharmacol ; 933: 175242, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36058290

RESUMEN

Microglia-mediated neuroinflammation plays a vital role in the pathogenesis of ischemic stroke, which serves as a prime target for developing novel therapeutic agent. However, feasible and effective agents for controlling neuroinflammation are scarce. Bergapten were acknowledged to hold therapeutic potential in restricting inflammation in multiple diseases, including peripheral neuropathy, migraine headaches and osteoarthritis. Here, we aimed to investigate the impact of bergapten on microglia-mediated neuroinflammation and its therapeutic potential in ischemic stroke. Our study demonstrated that bergapten significantly reduced the expression of pro-inflammatory cytokines and the activation of NF-κB signaling pathway in LPS-stimulated primary microglia. Mechanistically, bergapten suppressed cellular potassium ion efflux by inhibiting Kv1.3 channel and inhibits the degradation of Carbonyl reductase 1 induced by LPS, which might contribute to the anti-inflammatory effect of bergapten. Furthermore, bergapten suppressed microglial activation and post-stroke neuroinflammation in an experimental stroke model, leading to reduced infarct size and improved functional recovery. Thus, our study identified that bergapten might be a potential therapeutic compound for the treatment of ischemic stroke.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular Isquémico , Canal de Potasio Kv1.3/metabolismo , 5-Metoxipsoraleno/farmacología , Antiinflamatorios/farmacología , Lesiones Encefálicas/metabolismo , Carbonil Reductasa (NADPH)/metabolismo , Citocinas/metabolismo , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Lipopolisacáridos/farmacología , Microglía , FN-kappa B/metabolismo , Enfermedades Neuroinflamatorias , Potasio/metabolismo
9.
Anim Reprod Sci ; 237: 106910, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34974394

RESUMEN

In this study, aims were to evaluate orexin A (OXA) effects on mRNA abundance of important enzymes involved in prostaglandin production, such as cyclooxygenase 2 (PTGS2), microsomal PGE2 synthase-1 (PTGES), PGF2α synthase (PGFS) and carbonyl reductase 1 (CBR1), as well as prostaglandin E2 (PGE2) and F2α (PGF2α) culture medium concentrations for endometrial and myometrial explants. Tissues were collected from gilts during specific phases of the estrogenic cycle or early gestational period. There were greater concentrations of PGE2 with OXA treatments of endometrial tissues collected on days 12-13 and 27-28, as well as PGF2α on days 10-11 of the gestational period. The PGF2α concentrations were less in tissues collected on days 27-28 of the gestational period. The OXA treatments resulted in lesser concentrations of PGE2 from myometrial tissues collected on days 10-11 and greater PGF2α on days 10-11 of the gestational period and 10-11 of the estrogenic cycle. Effects of OXA may occur due to actions at PTGS2, PTGES, PGFS and CBR1 genes because mRNA abundances for proteins encoded by these genes were affected by OXA. Results indicate there is an OXA effect on mRNA abundances and prostaglandin culture medium concentrations of uterine tissue collected at different stages of the gestational period or estrogenic cycle using different doses of OXA. It, therefore, is concluded OXA may affect de novo synthesis and secretion of PGE2 and PGF2α in the uterus of pigs.


Asunto(s)
Carbonil Reductasa (NADPH) , Dinoprost , Animales , Carbonil Reductasa (NADPH)/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprost/metabolismo , Dinoprost/farmacología , Dinoprostona/metabolismo , Endometrio/metabolismo , Femenino , Orexinas/farmacología , Embarazo , Prostaglandinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos , Útero/metabolismo
10.
J Steroid Biochem Mol Biol ; 215: 106025, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34775032

RESUMEN

Nowadays, 17ß-estradiol (E2) biodegradation pathway has still not been identified in bacteria. To bridge this gap, we have described a novel E2 degradation pathway in Rhodococcus sp. P14 in this study, which showed that estradiol could be first transferred to estrone (E1) and thereby further converted into 16-hydroxyestrone, and then transformed into opened estrogen D ring. In order to identify the genes, which may be responsible for the pathway, transcriptome analysis was performed during E2 degradation in strain P14. The results showed that the expression of a short-chain dehydrogenase (SDR) gene and a CYP123 gene in the same gene cluster could be induced significantly by E2. Based on gene analysis, this gene cluster was found to play an important role in transforming E2 to 16-hydroxyestrone. The function of CYP123 was unknown before this study, and was found to harbor the activity of 16-estrone hydratase. Moreover, the global response to E2 in strain P14 was also analyzed by transcriptome analysis. It was observed that various genes involved in the metabolism processes, like the TCA cycle, lipid and amino acid metabolism, as well as glycolysis showed a significant increase in mRNA levels in response to strain P14 that can use E2 as the single carbon source. Overall, this study provides us an in depth understanding of the E2 degradation mechanisms in bacteria and also sheds light about the ability of strain P14 to effectively use E2 as the major carbon source for promoting its growth.


Asunto(s)
Carbonil Reductasa (NADPH)/genética , Sistema Enzimático del Citocromo P-450/genética , Estradiol/metabolismo , Regulación Bacteriana de la Expresión Génica , Rhodococcus/metabolismo , Transcriptoma , Biotransformación , Carbono/metabolismo , Carbonil Reductasa (NADPH)/metabolismo , Ciclo del Ácido Cítrico/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Estrona/metabolismo , Ontología de Genes , Hidroxiestronas/metabolismo , Metabolismo de los Lípidos/genética , Anotación de Secuencia Molecular , Familia de Multigenes , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rhodococcus/clasificación , Rhodococcus/genética
11.
Chem Biol Interact ; 351: 109752, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34801537

RESUMEN

2,4,6-trinitrotoluene (TNT) is a known source of reactive oxygen species (ROS), which cause oxidative stress in aquatic ecosystems. Carbonyl reductases (CRs) are one of several possible defense mechanisms induced against ROS products, especially those that result in the 'so-called' carbonyl stress. Daphnia magna, a freshwater organism living in stagnant freshwater bodies, expresses four copies of the CR gene (Dma_CR1, Dma_CR2, Dma_CR3 and Dma_CR4). In this study, induction of all four copies of Dma_CR by 2-amino-4,6-dinitrotoluene (2-ADNT) and 4-amino-2,6-dinitrotoluene (4-ADNT), was investigated. Reverse transcription polymerase chain reaction (RT-PCR) analysis of treated daphnids revealed up-regulation of Dma_CR1 alone in response to TNT, but not 2-ADNT and 4-ADNT (which are key metabolites of TNT). This concentration- and time-dependent up-regulation in mRNA-expression was observed both in the presence and absence of light, in the same magnitude. Moreover, significant change in mRNA-expression could be observed 8 h after treatment with TNT. In the presence of TNT, the antioxidant N-acetylcysteine (NAc) could not reverse TNT-induced up-regulation of Dma_CR1 mRNA-expression. On the other hand, withdrawal of TNT from the culture medium caused a significant reduction in the TNT-induced mRNA-expression of Dma_CR1 within 24 h. These findings highlight the potential of Dma_CR1 as a biomarker for biomonitoring of TNT levels in freshwater bodies.


Asunto(s)
Carbonil Reductasa (NADPH)/metabolismo , Daphnia/efectos de los fármacos , Trinitrotolueno/farmacología , Regulación hacia Arriba/efectos de los fármacos , Contaminantes Químicos del Agua/farmacología , Compuestos de Anilina/farmacología , Animales , Biomarcadores/metabolismo , Carbonil Reductasa (NADPH)/genética
12.
Chem Biol Interact ; 348: 109634, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34506768

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) are used worldwide as antipyretic analgesics and agents for rheumatoid arthritis and osteoarthritis, but known to cause damage to the gastrointestinal mucosae as their serious adverse effects. Few studies showed the impairment of intestinal epithelial barrier function (EBF) by high concentrations (0.5-1 mM) of NSAIDs, but the underlying mechanism is not fully understood. This study is aimed at clarifying effects at a low concentration (50 µM) of three NSAIDs, loxoprofen (Lox), ibuprofen and indomethacin, on intestinal EBF using human intestinal epithelial-like Caco-2 cells. Among those NSAIDs, Lox increased the transepithelial electric resistance (TER) value, decreased the paracellular Lucifer yellow CH (LYCH) permeability, and upregulated claudin (CLDN)-1, -3 and -5, indicating that low doses of Lox enhanced EBF through increasing expression of CLDNs. Lox is known to be metabolized to a pharmacologically active metabolite, (2S,1'R,2'S)-loxoprofen alcohol (Lox-RS), by carbonyl reductase 1 (CBR1), which is highly expressed in human intestine. CBR1 was expressed in the Caco-2 cells, and the pretreatment with a CBR1 inhibitor suppressed both the Lox-evoked CLDN upregulation and EBF enhancement. In addition, the treatment of the cells with Lox-RS resulted in higher TER value and lower LYCH permeability than those with Lox. Thus, Lox-RS synthesized by CBR1 may greatly contribute to the improving efficacy of Lox on the barrier function. Since EBF is decreased in inflammatory bowel disease, we finally examined the effect of Lox on EBF using the Caco-2/THP-1 co-culture system, which is used as an in vitro inflammatory bowel disease model. Lox significantly recovered EBF which was impaired by inflammatory cytokines secreted from THP-1 macrophages. These in vitro observations suggest that Lox enhances intestinal EBF, for which the metabolism of Lox to Lox-RS by CBR1 has an important role.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Carbonil Reductasa (NADPH)/metabolismo , Diferenciación Celular/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Fenilpropionatos/farmacología , Antiinflamatorios no Esteroideos/metabolismo , Células CACO-2 , Citocinas/metabolismo , Humanos , Mucosa Intestinal/citología , Fenilpropionatos/metabolismo
13.
Biochem J ; 478(19): 3597-3611, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34542554

RESUMEN

The hetero-oligomeric retinoid oxidoreductase complex (ROC) catalyzes the interconversion of all-trans-retinol and all-trans-retinaldehyde to maintain the steady-state output of retinaldehyde, the precursor of all-trans-retinoic acid that regulates the transcription of numerous genes. The interconversion is catalyzed by two distinct components of the ROC: the NAD(H)-dependent retinol dehydrogenase 10 (RDH10) and the NADP(H)-dependent dehydrogenase reductase 3 (DHRS3). The binding between RDH10 and DHRS3 subunits in the ROC results in mutual activation of the subunits. The molecular basis for their activation is currently unknown. Here, we applied site-directed mutagenesis to investigate the roles of amino acid residues previously implied in subunit interactions in other SDRs to obtain the first insight into the subunit interactions in the ROC. The results of these studies suggest that the cofactor binding to RDH10 subunit is critical for the activation of DHRS3 subunit and vice versa. The C-terminal residues 317-331 of RDH10 are critical for the activity of RDH10 homo-oligomers but not for the binding to DHRS3. The C-terminal residues 291-295 are required for DHRS3 subunit activity of the ROC. The highly conserved C-terminal cysteines appear to be involved in inter-subunit communications, affecting the affinity of the cofactor binding site in RDH10 homo-oligomers as well as in the ROC. Modeling of the ROC quaternary structure based on other known structures of SDRs suggests that its integral membrane-associated subunits may be inserted in adjacent membranes of the endoplasmic reticulum (ER), making the formation and function of the ROC dependent on the dynamic nature of the tubular ER network.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Carbonil Reductasa (NADPH)/metabolismo , Proteínas de la Membrana/metabolismo , Retinaldehído/metabolismo , Tretinoina/metabolismo , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Secuencia de Aminoácidos , Animales , Biocatálisis , Carbonil Reductasa (NADPH)/química , Carbonil Reductasa (NADPH)/genética , Dominio Catalítico , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mutagénesis Sitio-Dirigida/métodos , Estructura Cuaternaria de Proteína , Spodoptera/citología , Relación Estructura-Actividad
14.
Genomics ; 113(3): 1057-1069, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33667649

RESUMEN

The Bromodomain and Extra-terminal domain (BET) proteins are promising targets in treating cancers. Although BET inhibitors have been in clinical trials, they are limited by lacking of suitable biomarkers to indicate drug responses in different cancers. Here we identify DHRS2, ETV4 and NOTUM as potential biomarkers to indicate drug resistance in liver cancer cells of a recently discovered BET inhibitor, Hjp-6-171. Furthermore, we confirm that reactivation of WNT pathway, the target of NOTUM, contributes to the drug sensitivity restoration in Hjp-6-171 resistant cells. Specially, combinations of Hjp-6-171 and a GSK3ß inhibitor CHIR-98014 show remarkable therapeutic effects in vitro and in vivo. Integrating RNA-seq and ChIP-seq data, we reveal the expression signature of ß-catenin regulated genes is contrary in sensitive cells to that in resistant cells. We propose WNT signaling molecules such as ß-catenin and ETV4 to be candidate biomarkers to indicate BET inhibitor responses in liver cancer patients.


Asunto(s)
Neoplasias Hepáticas , Vía de Señalización Wnt , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
15.
Eur J Pharmacol ; 898: 173972, 2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-33652058

RESUMEN

Dehydrogenase/reductase member 2 (DHRS2) belongs to the short-chain dehydrogenase/reductase (SDR) family. It was initially isolated from the nuclear extract of hepatocellular carcinoma HepG2 cells and was identified as a specific cell cycle regulator. DHRS2 is a reduced nicotinamide adenine dinucleotide phosphate (NADPH)-dependent carbonyl reductase and catalyzes the reduction of dicarbonyl compounds. It is also functionally active in lipid metabolism and acts as a metabolic enzyme of hormones. Recent studies have shown that DHRS2 reprograms lipid metabolism and redox homeostasis to regulate proliferation, migration, invasion, and drug resistance of cancer cells. Here, we describe the structure, organelle localization and function of DHRS2, and also highlight its roles in the pathologic progression of diseases.


Asunto(s)
Carbonil Reductasa (NADPH)/metabolismo , Metabolismo de los Lípidos , Neoplasias/enzimología , Animales , Antineoplásicos/farmacología , Carbonil Reductasa (NADPH)/antagonistas & inhibidores , Carbonil Reductasa (NADPH)/química , Inhibidores Enzimáticos/farmacología , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Conformación Proteica , Relación Estructura-Actividad
16.
FEBS Lett ; 594(20): 3395-3405, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32767399

RESUMEN

DNA topoisomerase II beta (TOP2B) has a role in transcriptional regulation. Here, to further investigate transcriptional regulation by TOP2B, we used RNA-sequencing and real-time PCR to analyse the differential gene expression profiles of wild-type and two independent TOP2B-null pre-B Nalm-6 cell lines, one generated by targeted insertion and the other using CRISPR-Cas9 gene editing. We identified carbonyl reductase 1 (CBR1) among the most significantly downregulated genes in these TOP2B-null cells. Reduced CBR1 expression was accompanied by loss of binding of the transcription factors USF2 and MAX to the CBR1 promoter. We describe possible mechanisms by which loss of TOP2B results in CBR1 downregulation. To our knowledge, this is the first report of a link between TOP2B and CBR1.


Asunto(s)
Carbonil Reductasa (NADPH)/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Carbonil Reductasa (NADPH)/metabolismo , Línea Celular , Epigénesis Genética , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Regiones Promotoras Genéticas
17.
Arch Toxicol ; 94(9): 3059-3068, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32588086

RESUMEN

Although novel anticancer drugs are being developed intensively, anthracyclines remain the gold standard in the treatment of acute myeloid leukaemia (AML). The reductive conversion of daunorubicin (Dau) to less active daunorubicinol (Dau-ol) is an important mechanism that contributes to the development of pharmacokinetic anthracycline resistance. Dau is a key component in many AML regimes, in which it is combined with many drugs, including all-trans-retinoic acid (ATRA), cytarabine, cladribine and prednisolone. In the present study, we investigated the influence of these anticancer drugs on the reductive Dau metabolism mediated by the aldo-keto reductases AKR1A1, 1B10, 1C3, and 7A2 and carbonyl reductase 1 (CBR1). In incubation experiments with recombinant enzymes, cladribine and cytarabine did not significantly inhibit the activity of the tested enzymes. Prednisolone inhibited AKR1C3 with an IC50 of 41.73 µM, while ATRA decreased the activity of AKR1B10 (IC50 = 78.33 µM) and AKR1C3 (IC50 = 1.17 µM). Subsequent studies showed that AKR1C3 inhibition mediated by ATRA exhibited tight binding (Kiapp = 0.54 µM). Further, the combination of 1 µM ATRA with different concentrations of Dau demonstrated synergistic effects in HCT116 and KG1a human cells expressing AKR1C3. Our results suggest that ATRA-mediated inhibition of AKR1C3 can contribute to the mechanisms that are hidden beyond the beneficial clinical outcome of the ATRA-Dau combination.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Carbonil Reductasa (NADPH)/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Oxidorreductasas de Alcohol , Antraciclinas , Antibióticos Antineoplásicos/uso terapéutico , Antineoplásicos , Daunorrubicina , Humanos
18.
Drug Dev Res ; 81(7): 885-892, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32542754

RESUMEN

Investigate the effect of flufenamic acid (FFA) on lung injury of sepsis rats. Rat sepsis model was established using cecal ligation and puncture (CLP). The pathomorphology of lung tissue was detected by Hematoxylin-eosin (H&E) staining. The expression levels of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and high mobility group box-1 (HMGB-1) in serum and TNF-α, IL-6, malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) in lung tissues. The viability of RLE-6TN cells was detected by CCK-8 assay. The expression of carbonyl reductase 1 (CBR1) in RLE-6TN cells was analyzed by Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis. The inflammatory response was obviously enhanced in CLP-constructed sepsis rats and alleviated by FFA treatment. Sepsis induced the increase of W/D ratio, promoted the levels of TNF-α, IL-6, HMGBR1, and MDA and inhibited the levels of SOD and GSH. FFA could effectively alleviate the sepsis-induced lung injury. The viability of RLE-6TN cells induced by LPS was improved with the treatment of FFA. CBR1 expression in LPS-induced RLE-6TN cells was decreased and FFA could up-regulate the CBR1 expression. In addition, LPS-induced lung injury promoted the inflammatory response in lung tissues, increased the W/D ratio and levels of TNF-α, IL-6, HMGBR1, and MDA while inhibited the levels of SOD and GSH. FFA could effectively improve the LPS-induced lung injury while the effect of FFA on LPS-induced lung injury was alleviated by CBR1 interference. FFA may alleviate sepsis-induced lung injury by up-regulating CBR1.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Carbonil Reductasa (NADPH)/metabolismo , Ácido Flufenámico/uso terapéutico , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/farmacología , Carbonil Reductasa (NADPH)/genética , Línea Celular , Ácido Flufenámico/farmacología , Glutatión/metabolismo , Interleucina-6/sangre , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Sepsis/complicaciones , Sepsis/metabolismo , Sepsis/patología , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba/efectos de los fármacos
19.
Epigenetics ; 15(1-2): 122-133, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31423895

RESUMEN

Histone deacetylases (HDACs) have been linked to a variety of cancers, and HDAC inhibitors (HDACi) are a promising class of drugs that have demonstrated anti-cancer effects. However, we have little knowledge regarding the selection and application of HDAC inhibitors to the personalized treatment of ovarian cancer (OC). Here, we report a correlation between the high expression of HDACs and poor outcomes in OC patients, which reveals that HDACi are a class of agents that show great promise for the treatment of OC. Furthermore, we found that HDACi increased both the mRNA and protein levels of DHRS2, which has been shown to be closely linked to HDACi sensitivity when it is highly expressed, especially in ovarian cancer cells. Consistently, we found that suppression of DHRS2 reduced the sensitivity of OC cells to HDAC inhibitors via attenuation of the inhibitory effects of HDAC inhibitors on Mcl-1 in vitro. Our study demonstrated that DHRS2 expression was decreased in OC tissues and that high expression of DHRS2 was correlated with better outcomes in OC patients. In addition, DHRS2 expression was closely related to the effects of chemotherapy. Our study reveals the role of DHRS2 in cell apoptosis induced by HDAC inhibitors and explores the clinical attributes of DHRS2 in OC from a new perspective, suggesting that OC patients with high DHRS2 expression may benefit from treatment with HDAC inhibitors.


Asunto(s)
Biomarcadores de Tumor/genética , Carbonil Reductasa (NADPH)/genética , Resistencia a Antineoplásicos , Neoplasias Ováricas/genética , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Carbonil Reductasa (NADPH)/metabolismo , Línea Celular Tumoral , Femenino , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Neoplasias Ováricas/patología , Pronóstico
20.
ACS Chem Biol ; 15(1): 44-51, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31860257

RESUMEN

Fusidane-type antibiotics are a group of triterpenoid antibiotics. They include helvolic acid, fusidic acid, and cephalosporin P1, among which fusidic acid has been used clinically. We have recently elucidated the biosynthesis of helvolic acid and fusidic acid, which share an early biosynthetic route involving six conserved enzymes. Here, we report two separate gene clusters for cephalosporin P1 biosynthesis. One consists of the six conserved genes, and the other contains three genes encoding a P450 enzyme (CepB4), an acetyltransferase (CepD2), and a short-chain dehydrogenase/reductase (CepC2). Introduction of these three genes into Aspergillus oryzae, which harbors the six conserved genes, produced cephalosporin P1. Stepwise introduction revealed that CepB4 not only catalyzes stereoselective dual oxidation of C6 and C7, but also monooxygenation of C6 or C7. This led to the generation of five new analogues. Using monohydroxylated products as substrates, we demonstrated that CepD2 specifically acetylates C6-OH, although both C6-OH and C7-OH acetylated analogues have been identified in nature.


Asunto(s)
Cefalosporinas/biosíntesis , Sistema Enzimático del Citocromo P-450/metabolismo , Enzimas Multifuncionales/metabolismo , Acetilación , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Aspergillus oryzae/genética , Secuencia de Bases , Carbonil Reductasa (NADPH)/genética , Carbonil Reductasa (NADPH)/metabolismo , Dominio Catalítico , Clonación Molecular , Sistema Enzimático del Citocromo P-450/genética , Ácido Fusídico/análogos & derivados , Ácido Fusídico/química , Regulación de la Expresión Génica , Hidroxilación , Estructura Molecular , Enzimas Multifuncionales/genética , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...