Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
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.
Molecules ; 28(9)2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37175180

RESUMEN

Anthracycline antibiotics (ANT) are among the most widely used anticancer drugs. Unfortunately, their use is limited due to the development of drug resistance and cardiotoxicity. ANT metabolism, performed mainly by two enzymes-aldo-keto reductase 1C3 (AKR1C3) and carbonyl reductase 1 (CBR1)-is one of the proposed mechanisms generated by the described effects. In this study, we evaluated the CBR1 inhibitory properties of ASP9521, a compound already known as potent AKR1C3 inhibitor. First, we assessed the possibility of ASP9521 binding to the CBR1 catalytic site using molecular docking and molecular dynamics. The research revealed a potential binding mode of ASP9521. Moderate inhibitory activity against CBR1 was observed in studies with recombinant enzymes. Finally, we examined whether ASP9521 can improve the cytotoxic activity of daunorubicin against human lung carcinoma cell line A549 and assessed the cardioprotective properties of ASP9521 in a rat cardiomyocytes model (H9c2) against doxorubicin- and daunorubicin-induced toxicity. The addition of ASP9521 ameliorated the cytotoxic activity of daunorubicin and protected rat cardiomyocytes from the cytotoxic effect of both applied drugs. Considering the favorable bioavailability and safety profile of ASP9521, the obtained results encourage further research. Inhibition of both AKR1C3 and CBR1 may be a promising method of overcoming ANT resistance and cardiotoxicity.


Asunto(s)
Antineoplásicos , Carbonil Reductasa (NADPH) , Humanos , Ratas , Animales , Simulación del Acoplamiento Molecular , Cardiotoxicidad , Antraciclinas/farmacología , Antraciclinas/metabolismo , Antibióticos Antineoplásicos/farmacología , Daunorrubicina/farmacología , Antineoplásicos/farmacología , Antibacterianos
5.
Chem Biol Interact ; 381: 110572, 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37247810

RESUMEN

A porcine gene, LOC100622246, encodes carbonyl reductase [NADPH] 1 (pCBR-N1), whose function remains unknown. Previously, three porcine carbonyl reductases, carbonyl reductase 1 (pCBR1), 3α/ß-hydroxysteroid dehydrogenase (p3α/ß-HSD) and prostaglandine-9-keto reductase (pPG-9-KR), were purified from neonatal testis, adult testis and adult kidney, respectively. However, the relationship of pCBR-N1 with the three enzymes is still unknown. Here, we compare the properties of the recombinant pCBR-N1 and pCBR1. The two enzymes reduced various carbonyl compounds including 5α-dihydrotestosterone, which was converted to its 3α- and 3ß-hydroxy-metabolites. Compared to pCBR1, pCBR-N1 exhibited higher Km and kcat values for most substrates, but more efficiently reduced prostaglandin E2. pCBR-N1 was inhibited by known inhibitors of p3α/ß-HSD (hexestrol and indomethacin), but not by pCBR1 inhibitors. pCBR-N1 was highly expressed than pCBR1 in the several tissues of adult domestic and microminiature pigs. The results, together with partial amino acid sequence match between pCBR-N1 and pPG-9-KR, reveal that pCBR-N1 is identical to p3α/ß-HSD and pPG-9-KR. Notably, pCBR-N1, but not pCBR1, reduced S-nitrosoglutathione and glutathione-adducts of alkenals including 4-oxo-2-nonenal with Km of 8.3-32 µM, and its activity toward non-glutathionylated substrates was activated 2- to 9-fold by 1 mM glutathione. Similar activation by glutathione was also observed for human CBR1. Site-directed mutagenesis revealed that the differences in kinetic constants and glutathione-mediated activation between pCBR-N1 and pCBR1 are due to differences in residue 236 and two glutathione-binding residues (at positions 97 and 193), respectively. Thus, pCBR-N1 is a glutathione-activated carbonyl reductase that functions in the metabolism of endogenous and xenobiotic carbonyl compounds.


Asunto(s)
Oxidorreductasas de Alcohol , Carbonil Reductasa (NADPH) , Animales , Humanos , Masculino , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hidroxiesteroide Deshidrogenasas/metabolismo , Porcinos
6.
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
7.
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
8.
FEBS J ; 290(8): 2127-2145, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36421037

RESUMEN

Reactive oxygen species (ROS) are considered a major cause of ageing and ageing-related diseases through protein carbonylation. Little is known about the molecular mechanisms that confer protection against ROS. Here, we observed that, compared with nondiapause-destined pupae, high protein carbonyl levels are present in the brains of diapause-destined pupae, which is a 'non-ageing' phase in the moth Helicoverpa armigera. Protein carbonyl levels respond to ROS and decrease metabolic activity to induce diapause in order to extend lifespan. However, protein carbonylation in the brains of diapause-destined pupae still occurs at a physiological level compared to young adult brains. We find that ROS activate Akt, and Akt then phosphorylates the transcription factor CREB to facilitate its nuclear import. CREB binds to the promoter of carbonyl reductase 1 (CBR1) and regulates its expression. High CBR1 levels reduce protein carbonyl levels to maintain physiological levels. This is the first report showing that the moth brain can naturally control protein carbonyl levels through a distinct ROS-Akt-CREB-CBR1 pathway to extend lifespan.


Asunto(s)
Mariposas Nocturnas , Proteínas Proto-Oncogénicas c-akt , Animales , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Carbonil Reductasa (NADPH) , Longevidad/fisiología , Carbonilación Proteica , Mariposas Nocturnas/genética , Mariposas Nocturnas/metabolismo , Pupa/metabolismo
9.
Drug Metab Dispos ; 51(1): 17-28, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36310032

RESUMEN

Enzymes of the aldo-keto reductase (AKR) and short-chain dehydrogenase/reductase superfamilies are involved in the reduction of compounds containing a ketone group. In most cases, multiple isoforms appear to be involved in the reduction of a compound, and the enzyme(s) that are responsible for the reaction in the human liver have not been elucidated. The purpose of this study was to quantitatively evaluate the contribution of each isoform to reduction reactions in the human liver. Recombinant cytosolic isoforms were constructed, i.e., AKR1C1, AKR1C2, AKR1C3, AKR1C4, and carbonyl reductase 1 (CBR1), and a microsomal isoform, 11ß-hydroxysteroid dehydrogenase type 1 (HSD11B1), and their contributions to the reduction of 10 compounds were examined by extrapolating the relative expression of each reductase protein in human liver preparations to recombinant systems quantified by liquid chromatography-mass spectrometry. The reductase activities for acetohexamide, doxorubicin, haloperidol, loxoprofen, naloxone, oxcarbazepine, and pentoxifylline were predominantly catalyzed by cytosolic isoforms, and the sum of the contributions of individual cytosolic reductases was almost 100%. Interestingly, AKR1C3 showed the highest contribution to acetohexamide and loxoprofen reduction, although previous studies have revealed that CBR1 mainly metabolizes them. The reductase activities of bupropion, ketoprofen, and tolperisone were catalyzed by microsomal isoform(s), and the contributions of HSD11B1 were calculated to be 41%, 32%, and 104%, respectively. To our knowledge, this is the first study to quantitatively evaluate the contribution of each reductase to the reduction of drugs in the human liver. SIGNIFICANCE STATEMENT: To our knowledge, this is the first study to determine the contribution of aldo-keto reductase (AKR)-1C1, AKR1C2, AKR1C3, AKR1C4, carbonyl reductase 1, and 11ß-hydroxysteroid dehydrogenase type 1 to drug reductions in the human liver by utilizing the relative expression factor approach. This study found that AKR1C3 contributes to the reduction of compounds at higher-than-expected rates.


Asunto(s)
Cetonas , Deshidrogenasas-Reductasas de Cadena Corta , Humanos , Aldo-Ceto Reductasas/metabolismo , Carbonil Reductasa (NADPH) , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Acetohexamida , Hígado/metabolismo , Oxidorreductasas/metabolismo , Isoformas de Proteínas
10.
Mol Biol Rep ; 50(1): 531-540, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36352178

RESUMEN

BACKGROUND: Carbonyl reductase 1 (CBR1) is a nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductase with broad substrate specificity. CBR1 catalyzes the reduction of numerous carbonyl compounds, including quinones, prostaglandins, menadione, and multiple xenobiotics, while also participating in various cellular processes, such as carcinogenesis, apoptosis, signal transduction, and drug resistance. In this study, we aimed to generate transgenic mice overexpressing mouse Cbr1 (mCbr1), characterize the mCbr1 expression in different organs, and identify changes in protein expression patterns. METHODS AND RESULTS: To facilitate a deeper understanding of the functions of CBR1, we generated transgenic mice overexpressing CBR1 throughout the body. These transgenic mice overexpress 3xFLAG-tagged mCbr1 (3xFLAG-mCbr1) under the CAG promoter. Two lines of transgenic mice were generated, one with 3xFLAG-mCbr1 expression in multiple tissues, and the other, with specific expression of 3xFLAG-mCbr1 in the heart. Pathway and network analysis using transgenic mouse hearts identified 73 proteins with levels of expression correlating with mCbr1 overexpression. The expression of voltage-gated anion channels, which may be directly related to calcium ion-related myocardial contraction, was also upregulated. CONCLUSION: mCbr1 transgenic mice may be useful for further in vivo analyses of the molecular mechanisms regulated by Cbr1; such analyses will provide a better understanding of its effects on carcinogenesis and cardiotoxicity of certain cancer drugs.


Asunto(s)
Antineoplásicos , Carbonil Reductasa (NADPH) , Ratones , Animales , Ratones Transgénicos , Antineoplásicos/farmacología , Apoptosis , Carcinogénesis , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo
11.
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
12.
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
13.
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
14.
Pharm Biol ; 60(1): 1616-1624, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35980105

RESUMEN

CONTEXT: Aidi injection (ADI), a traditional Chinese medicine antitumor injection, is usually combined with doxorubicin (DOX) for the treatment of malignant tumours. The cardiotoxicity of DOX is ameliorated by ADI in the clinic. However, the relevant mechanism is unknown. OBJECTIVE: To investigate the effects of ADI on DOX-induced cardiotoxicity and its mechanism. MATERIALS AND METHODS: ICR mice were randomly divided into six groups: control, ADI-L, ADI-H, DOX, DOX + ADI-L and DOX + ADI-H. DOX (i.p., 0.03 mg/10 g) was administered in the presence or absence of ADI (i.p., 0.1 or 0.2 mL/10 g) for two weeks. Heart pathology and levels of AST, LDH, CK, CK-MB and BNP were assessed. H9c2 cells were treated with DOX in the presence or absence of ADI (1, 4, 10%). Cell viability, caspase-3 activity, nuclear morphology, and CBR1 expression were then evaluated. DOX and doxorubicinol (DOXol) concentrations in heart, liver, kidneys, serum, and cells were analysed by UPLC-MS/MS. RESULTS: High-dose ADI significantly reduced DOX-induced pathological changes and the levels of AST, LDH, CK, CK-MB and BNP to normal. Combined treatment with ADI (1, 4, 10%) improved the cell viability, and IC50 increased from 68.51 µM (DOX alone) to 83.47, 176.9, and 310.8 µM, reduced caspase-3 activity by 39.17, 43.96, and 61.82%, respectively. High-dose ADI inhibited the expression of CBR1 protein by 32.3%, reduced DOXol levels in heart, serum and H9c2 cells by 59.8, 72.5 and 48.99%, respectively. DISCUSSION AND CONCLUSIONS: ADI reduces DOX-induced cardiotoxicity by inhibiting CBR1 expression, which provides a scientific basis for the rational use of ADI.


Asunto(s)
Carbonil Reductasa (NADPH) , Cardiotoxicidad , Animales , Antibióticos Antineoplásicos/toxicidad , Cardiotoxicidad/metabolismo , Caspasa 3 , Cromatografía Liquida , Forma MB de la Creatina-Quinasa , Doxorrubicina/toxicidad , Ratones , Ratones Endogámicos ICR , Espectrometría de Masas en Tándem
15.
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
16.
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
17.
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
18.
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
19.
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
20.
Transl Stroke Res ; 12(5): 711-724, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33964000

RESUMEN

Oxidative stress and neuroinflammatory response after the ischemic injury are important pathophysiologic mechanisms that cause brain tissue loss and neurological deficit. This study aims to observe the expression and role of carbonyl reductase 1 (CBR1), an NADPH-dependent oxidoreductase with specificity for carbonyl compounds such as 4-hydroxynonenal (4-HNE), in the brain after ischemic injury and to investigate the influence of CBR1 on ischemia-induced neuroinflammation. CBR1 expresses in the neurons, astrocyte, and microglia in the normal brain. The expression of CBR1 decreased in the ischemic regions following cerebral ischemia, and also reduced in primary neurons after OGD (oxygen-glucose deprivation); however, the expression of CBR1 significantly increased in microglia in the ischemic penumbra. Furthermore, TAT-CBR1 fusion protein played neuroprotective effects in reducing the infarct volume and improving neurological outcomes after ischemic injury. Mechanistically, CBR1 decreased the levels of 4-HNE in the brain after stroke; it also modulated microglial polarization toward the M2 phenotype, which was well-known to confer neuroprotection after ischemic injury. Our results demonstrate that CBR1 provides neuroprotection against ischemic injury by reducing oxidative stress and neuroinflammation, making a promising agent for cerebral ischemia treatment.


Asunto(s)
Oxidorreductasas de Alcohol , Lesiones Encefálicas , Isquemia Encefálica , Fármacos Neuroprotectores , Carbonil Reductasa (NADPH) , Humanos , Microglía , Enfermedades Neuroinflamatorias , Estrés Oxidativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...