Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 138
Filtrer
1.
Oncol Res ; 32(8): 1287-1308, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055885

RÉSUMÉ

Aldo-keto reductases (AKRs) are a superfamily of enzymes that play crucial roles in various cellular processes, including the metabolism of xenobiotics, steroids, and carbohydrates. A growing body of evidence has unveiled the involvement of AKRs in the development and progression of various cancers. AKRs are aberrantly expressed in a wide range of malignant tumors. Dysregulated expression of AKRs enables the acquisition of hallmark traits of cancer by activating oncogenic signaling pathways and contributing to chemoresistance. AKRs have emerged as promising oncotherapeutic targets given their pivotal role in cancer development and progression. Inhibition of aldose reductase (AR), either alone or in combination with chemotherapeutic drugs, has evolved as a pragmatic therapeutic option for cancer. Several classes of synthetic aldo-keto reductase (AKR) inhibitors have been developed as potential anticancer agents, some of which have shown promise in clinical trials. Many AKR inhibitors from natural sources also exhibit anticancer effects. Small molecule inhibitors targeting specific AKR isoforms have shown promise in preclinical studies. These inhibitors disrupt the activation of oncogenic signaling by modulating transcription factors and kinases and sensitizing cancer cells to chemotherapy. In this review, we discuss the physiological functions of human AKRs, the aberrant expression of AKRs in malignancies, the involvement of AKRs in the acquisition of cancer hallmarks, and the role of AKRs in oncogenic signaling, and drug resistance. Finally, the potential of aldose reductase inhibitors (ARIs) as anticancer drugs is summarized.


Sujet(s)
Aldo-keto reductases , Antinéoplasiques , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Aldo-keto reductases/antagonistes et inhibiteurs , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux , Antienzymes/usage thérapeutique , Antienzymes/pharmacologie , Médecine de précision , Transduction du signal , Aldose reductase/antagonistes et inhibiteurs , Aldose reductase/métabolisme
2.
Yale J Biol Med ; 97(2): 179-204, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38947111

RÉSUMÉ

Aldo-keto reductases (AKRs) are a superfamily of promiscuous enzymes that have been chiseled by evolution to act as catalysts for numerous regulatory pathways in humans. However, they have not lost their promiscuity in the process, essentially making them a double-edged sword. The superfamily is involved in multiple metabolic pathways and are linked to chronic diseases such as cataracts, diabetes, and various cancers. Unlike other detoxifying enzymes such as cytochrome P450s (CYP450s), short-chain dehydrogenases (SDRs), and medium-chain dehydrogenases (MDRs), that participate in essential pathways, AKRs are more widely distributed and have members with interchangeable functions. Moreover, their promiscuity is ubiquitous across all species and participates in the resistance of pathogenic microbes. Moreover, the introduction of synthetic substrates, such as synthetic molecules and processed foods, results in unwanted "toxification" due to enzyme promiscuity, leading to chronic diseases.


Sujet(s)
Aldo-keto reductases , Cataracte , Tumeurs , Humains , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Cataracte/enzymologie , Cataracte/génétique , Cataracte/métabolisme , Maladie chronique , Tumeurs/enzymologie , Tumeurs/génétique
3.
Lipids Health Dis ; 23(1): 201, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937844

RÉSUMÉ

BACKGROUND: Nonalcoholic steatohepatitis (NASH) is a prevalent chronic liver condition. However, the potential therapeutic benefits and underlying mechanism of nicotinate-curcumin (NC) in the treatment of NASH remain uncertain. METHODS: A rat model of NASH induced by a high-fat and high-fructose diet was treated with nicotinate-curcumin (NC, 20, 40 mg·kg- 1), curcumin (Cur, 40 mg·kg- 1) and metformin (Met, 50 mg·kg- 1) for a duration of 4 weeks. The interaction between NASH, Cur and Aldo-Keto reductase family 1 member B10 (AKR1B10) was filter and analyzed using network pharmacology. The interaction of Cur, NC and AKR1B10 was analyzed using molecular docking techniques, and the binding energy of Cur and NC with AKR1B10 was compared. HepG2 cells were induced by Ox-LDL (25 µg·ml- 1, 24 h) in high glucose medium. NC (20µM, 40µM), Cur (40µM) Met (150µM) and epalrestat (Epa, 75µM) were administered individually. The activities of ALT, AST, ALP and the levels of LDL, HDL, TG, TC and FFA in serum were quantified using a chemiluminescence assay. Based on the changes in the above indicators, score according to NAS standards. The activities of Acetyl-CoA and Malonyl-CoA were measured using an ELISA assay. And the expression and cellular localization of AKR1B10 and Acetyl-CoA carboxylase (ACCα) in HepG2 cells were detected by Western blotting and immunofluorescence. RESULTS: The results of the animal experiments demonstrated that NASH rat model induced by a high-fat and high-fructose diet exhibited pronounced dysfunction in liver function and lipid metabolism. Additionally, there was a significant increase in serum levels of FFA and TG, as well as elevated expression of AKR1B10 and ACCα, and heightened activity of Acetyl-CoA and Malonyl-CoA in liver tissue. The administration of NC showed to enhance liver function in rats with NASH, leading to reductions in ALT, AST and ALP levels, and decrease in blood lipid and significant inhibition of FFA and TG synthesis in the liver. Network pharmacological analysis identified AKR1B10 and ACCα as potential targets for NASH treatment. Molecular docking studies revealed that both Cur and NC are capable of binding to AKR1B10, with NC exhibiting a stronger binding energy to AKR1B10. Western blot analysis demonstrated an upregulation in the expression of AKR1B10 and ACCα in the liver tissue of NASH rats, accompanied by elevated Acetyl-CoA and Malonyl-CoA activity, and increased levels of FFA and TG. The results of the HepG2 cell experiments induced by Ox-LDL suggest that NC significantly inhibited the expression and co-localization of AKR1B10 and ACCα, while also reduced levels of TC and LDL-C and increased level of HDL-C. These effects are accompanied by a decrease in the activities of ACCα and Malonyl-CoA, and levels of FFA and TG. Furthermore, the impact of NC appears to be more pronounced compared to Cur. CONCLUSION: NC could effectively treat NASH and improve liver function and lipid metabolism disorder. The mechanism of NC is related to the inhibition of AKR1B10/ACCα pathway and FFA/TG synthesis of liver.


Sujet(s)
Aldo-keto reductases , Curcumine , Stéatose hépatique non alcoolique , Triglycéride , Curcumine/pharmacologie , Curcumine/analogues et dérivés , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Animaux , Humains , Cellules HepG2 , Aldo-keto reductases/métabolisme , Rats , Mâle , Triglycéride/sang , Triglycéride/métabolisme , Acetyl-coA carboxylase/métabolisme , Aldose reductase/métabolisme , Aldose reductase/antagonistes et inhibiteurs , Alimentation riche en graisse/effets indésirables , Simulation de docking moléculaire , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Metformine/pharmacologie , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Rhodanine/analogues et dérivés , Thiazolidines
4.
Int J Biol Macromol ; 274(Pt 1): 133264, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38901517

RÉSUMÉ

Chiral alcohols are essential building blocks of numerous pharmaceuticals and fine chemicals. Aldo-keto reductases (AKRs) constitute a superfamily of oxidoreductases that catalyze the reduction of aldehydes and ketones to their corresponding alcohols using NAD(P)H as a coenzyme. Knowledge about the crucial roles of AKRs immobilization in the biocatalytic synthesis of chiral alcohols is expanding. Herein, we reviewed the characteristics of various AKRs immobilization approaches, the applications of different immobilization materials, and the prospects of continuous flow bioreactor construction by employing these immobilized biocatalysts for synthesizing chiral alcohols. Finally, the opportunities and ongoing challenges for AKR immobilization are discussed and the outlook for this emerging area is analyzed.


Sujet(s)
Alcools , Aldo-keto reductases , Biocatalyse , Enzymes immobilisées , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Aldo-keto reductases/composition chimique , Alcools/composition chimique , Alcools/métabolisme , Enzymes immobilisées/composition chimique , Enzymes immobilisées/métabolisme , Stéréoisomérie , Bioréacteurs
5.
Chem Biol Interact ; 398: 111111, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38878851

RÉSUMÉ

The aldo-keto reductase (AKR) superfamily is a large family of proteins found across the kingdoms of life. Shared features of the family include 1) structural similarities such as an (α/ß)8-barrel structure, disordered loop structure, cofactor binding site, and a catalytic tetrad, and 2) the ability to catalyze the nicotinamide adenine dinucleotide (phosphate) reduced (NAD(P)H)-dependent reduction of a carbonyl group. A criteria of family membership is that the protein must have a measured function, and thus, genomic sequences suggesting the transcription of potential AKR proteins are considered pseudo-members until evidence of a functionally expressed protein is available. Currently, over 200 confirmed AKR superfamily members are reported to exist. A systematic nomenclature for the AKR superfamily exists to facilitate family and subfamily designations of the member to be communicated easily. Specifically, protein names include the root "AKR", followed by the family represented by an Arabic number, the subfamily-if one exists-represented by a letter, and finally, the individual member represented by an Arabic number. The AKR superfamily database has been dedicated to tracking and reporting the current knowledge of the AKRs since 1997, and the website was last updated in 2003. Here, we present an updated version of the website and database that were released in 2023. The database contains genetic, functional, and structural data drawn from various sources, while the website provides alignment information and family tree structure derived from bioinformatics analyses.


Sujet(s)
Aldo-keto reductases , Bases de données de protéines , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Aldo-keto reductases/composition chimique , Humains , Internet , Aldose reductase/métabolisme , Aldose reductase/composition chimique , Aldose reductase/génétique , Animaux
6.
J Ethnopharmacol ; 332: 118354, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38762210

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Berberine (BBR) is the main active component from Coptidis rhizome, a well-known Chinese herbal medicine used for metabolic diseases, especially diabetes for thousands of years. BBR has been reported to cure various metabolic disorders, such as nonalcoholic fatty liver disease (NAFLD). However, the direct proteomic targets and underlying molecular mechanism of BBR against NAFLD remain less understood. AIM OF THE STUDY: To investigate the direct target and corresponding molecular mechanism of BBR on NAFLD is the aim of the current study. MATERIALS AND METHODS: High-fat diet (HFD)-fed mice and oleic acid (OA) stimulated HepG2 cells were utilized to verify the beneficial impacts of BBR on glycolipid metabolism profiles. The click chemistry in proteomics, DARTS, CETSA, SPR and fluorescence co-localization analysis were conducted to identify the targets of BBR for NAFLD. RNA-seq and shRNA/siRNA were used to investigate the downstream pathways of the target. RESULTS: BBR improved hepatic steatosis, ameliorated insulin resistance, and reduced TG levels in the NAFLD models. Importantly, Aldo-keto reductase 1B10 (AKR1B10) was first proved as the target of BBR for NAFLD. The gene expression of AKR1B10 increased significantly in the NAFLD patients' liver tissue. We further demonstrated that HFD and OA increased AKR1B10 expression in the C57BL/6 mice's liver and HepG2 cells, respectively, whereas BBR decreased the expression and activities of AKR1B10. Moreover, the knockdown of AKR1B10 by applying shRNA/siRNA profoundly impacted the beneficial effects on the pathogenesis of NAFLD by BBR. Meanwhile, the changes in various proteins (ACC1, CPT-1, GLUT2, etc.) are responsible for hepatic lipogenesis, fatty acid oxidation, glucose uptake, etc. by BBR were reversed by the knockdown of AKR1B10. Additionally, RNA-seq was used to identify the downstream pathway of AKR1B10 by examining the gene expression of liver tissues from HFD-fed mice. Our findings revealed that BBR markedly increased the protein levels of PPARα while downregulating the expression of PPARγ. However, various proteins of PPAR signaling pathways remained unaffected post the knockdown of AKR1B10. CONCLUSIONS: BBR alleviated NAFLD via mediating PPAR signaling pathways through targeting AKR1B10. This study proved that AKR1B10 is a novel target of BBR for NAFLD treatment and helps to find new targets for the treatment of NAFLD by using active natural compounds isolated from traditional herbal medicines as the probe.


Sujet(s)
Aldo-keto reductases , Berbérine , Alimentation riche en graisse , Métabolisme lipidique , Souris de lignée C57BL , Stéatose hépatique non alcoolique , Animaux , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Humains , Berbérine/pharmacologie , Berbérine/usage thérapeutique , Cellules HepG2 , Mâle , Alimentation riche en graisse/effets indésirables , Métabolisme lipidique/effets des médicaments et des substances chimiques , Souris , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Aldose reductase/métabolisme , Aldose reductase/génétique , Glucose/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Insulinorésistance
7.
Sci Rep ; 14(1): 12149, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38802416

RÉSUMÉ

Hepatocellular carcinoma (HCC) represents a major global health threat with diverse and complex pathogenesis. Aldo-keto reductase family 1 member B10 (AKR1B10), a tumor-associated enzyme, exhibits abnormal expression in various cancers. However, a comprehensive understanding of AKR1B10's role in HCC is lacking. This study aims to explore the expression characteristics of AKR1B10 in HCC and its correlation with clinicopathological features, survival prognosis, and tumor immune microenvironment, further investigating its role and potential regulatory mechanisms in HCC. This study conducted comprehensive analyses using various bioinformatics tools and databases. Initially, differentially expressed genes related to HCC were identified from the GEO database, and the expression of AKR1B10 in HCC and other cancers was compared using TIMER and GEPIA databases, with validation of its specificity in HCC tissue samples using the HPA database. Furthermore, the relationship of AKR1B10 expression with clinicopathological features (age, gender, tumor size, staging, etc.) of HCC patients was analyzed using the TCGA database's LIHC dataset. The impact of AKR1B10 expression levels on patient prognosis was evaluated using Kaplan-Meier survival analysis and the Cox proportional hazards model. Additionally, the correlation of AKR1B10 expression with tumor biology-related signaling pathways and tumor immune microenvironment was studied using databases like GSEA, Targetscan, and others, identifying microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) that regulate AKR1B10 expression to explore potential regulatory mechanisms. Elevated AKR1B10 expression was significantly associated with gender, primary tumor size, and fibrosis stage in HCC tissues. High AKR1B10 expression indicated poor prognosis and served as an independent predictor for patient outcomes. Detailed mechanism analysis revealed a positive correlation between high AKR1B10 expression, immune cell infiltration, and pro-inflammatory cytokines, suggesting a potential DANCR-miR-216a-5p-AKR1B10 axis regulating the tumor microenvironment and impacting HCC development and prognosis. The heightened expression of AKR1B10 in HCC is not only related to significant clinical-pathological traits but may also influence HCC progression and prognosis by activating key signaling pathways and altering the tumor immune microenvironment. These findings provide new insights into the role of AKR1B10 in HCC pathogenesis and highlight its potential as a biomarker and therapeutic target.


Sujet(s)
Aldo-keto reductase family 1 member B10 , Carcinome hépatocellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Microenvironnement tumoral , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/métabolisme , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/immunologie , Tumeurs du foie/mortalité , Tumeurs du foie/métabolisme , Mâle , Femelle , Pronostic , Aldo-keto reductase family 1 member B10/génétique , Aldo-keto reductase family 1 member B10/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Adulte d'âge moyen , Estimation de Kaplan-Meier , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , Analyse de profil d'expression de gènes , Biologie informatique/méthodes
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167214, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38718846

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC), is characteristic by a heterogeneous tumor microenvironment and gene mutations, conveys a dismal prognosis and low response to chemotherapy and immunotherapy. Here, we found that checkpoint suppressor 1 (CHES1) served as a tumor repressor in PDAC and was associated with patient prognosis. Functional experiments indicated that CHES1 suppressed the proliferation and invasion of PDAC by modulating cellular senescence. To further identify the downstream factor of CHES1 in PDAC, label-free quantitative proteomics analysis was conducted, which showed that the oncogenic Aldo-keto reductase 1B10 (AKR1B10) was transcriptionally repressed by CHES1 in PDAC. And AKR1B10 facilitated the malignant activity and repressed senescent phenotype of PDAC cells. Moreover, pharmaceutical inhibition of AKR1B10 with Oleanolic acid (OA) significantly induced tumor regression and sensitized PDAC cells to gemcitabine, and this combined therapy did not cause obvious side effects. Rescued experiments revealed that CHES1 regulated the tumorigenesis and gemcitabine sensitivity through AKR1B10-mediated senescence in PDAC. In summary, this study revealed that the CHES1/AKR1B10 axis modulated the progression and cellular senescence in PDAC, which might provide revenues for drug-targeting and senescence-inducing therapies for PDAC.


Sujet(s)
Aldose reductase , Aldo-keto reductases , Carcinome du canal pancréatique , Vieillissement de la cellule , , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Animaux , Humains , Souris , Aldose reductase/métabolisme , Aldose reductase/génétique , Aldose reductase/antagonistes et inhibiteurs , Aldo-keto reductases/métabolisme , Aldo-keto reductases/génétique , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude , Acide oléanolique/pharmacologie , Acide oléanolique/analogues et dérivés , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux
9.
Scand J Immunol ; 100(2): e13390, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38769661

RÉSUMÉ

Inflammation is an important pathophysiological process in many diseases; it has beneficial and harmful effects. When exposed to various stimuli, the body triggers an inflammatory response to eliminate invaded pathogens and damaged tissues to maintain homeostasis. However, uncontrollable persistent or excessive inflammatory responses may damage tissues and induce various diseases, such as metabolic diseases (e.g. diabetes), autoimmune diseases, nervous system-related diseases, digestive system-related diseases, and even tumours. Aldo-keto reductase 1B10 (AKR1B10) is an important player in the development and progression of multiple diseases, such as tumours and inflammatory diseases. AKR1B10 is upregulated in solid tumours, such as hepatocellular carcinoma (HCC), non-small cell lung carcinoma, and breast cancer, and is a reliable serum marker. However, information on the role of AKR1B10 in inflammation is limited. In this study, we summarized the role of AKR1B10 in inflammatory diseases, including its expression, functional contribution to inflammatory responses, and regulation of signalling pathways related to inflammation. We also discussed the role of AKR1B10 in glucose and lipid metabolism and oxidative stress. This study provides novel information and increases the understanding of clinical inflammatory diseases.


Sujet(s)
Aldo-keto reductases , Inflammation , Humains , Inflammation/immunologie , Aldo-keto reductases/métabolisme , Animaux , Stress oxydatif , Transduction du signal , Métabolisme lipidique , Glucose/métabolisme
10.
Lipids Health Dis ; 23(1): 137, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38720280

RÉSUMÉ

BACKGROUND: Evidence suggests that hepatocyte mitochondrial dysfunction leads to abnormal lipid metabolism, redox imbalance, and programmed cell death, driving the onset and progression of non-alcoholic steatohepatitis (NASH). Identifying hub mitochondrial genes linked to NASH may unveil potential therapeutic targets. METHODS: Mitochondrial hub genes implicated in NASH were identified via analysis using 134 algorithms. RESULTS: The Random Forest algorithm (RF), the most effective among the 134 algorithms, identified three genes: Aldo-keto reductase family 1 member B10 (AKR1B10), thymidylate synthase (TYMS), and triggering receptor expressed in myeloid cell 2 (TREM2). They were upregulated and positively associated with genes promoting inflammation, genes involved in lipid synthesis, fibrosis, and nonalcoholic steatohepatitis activity scores in patients with NASH. Moreover, using these three genes, patients with NASH were accurately categorized into cluster 1, exhibiting heightened disease severity, and cluster 2, distinguished by milder disease activity. CONCLUSION: These three genes are pivotal mitochondrial genes implicated in NASH progression.


Sujet(s)
Algorithmes , Apprentissage machine , Stéatose hépatique non alcoolique , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/anatomopathologie , Humains , Mitochondries/génétique , Mitochondries/métabolisme , Métabolisme lipidique/génétique , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , Gènes de mitochondrie
11.
Dig Dis Sci ; 69(7): 2502-2521, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38662158

RÉSUMÉ

BACKGROUND: Long noncoding RNAs (lncRNAs) have been shown to be related to the occurrence and development of a variety of cancers including hepatocellular carcinoma (HCC). However, a large number of potential HCC-related lncRNAs remain undiscovered and are yet to be fully understood. METHODS: Differentially expressed lncRNAs were first obtained from the tumor tissues and adjacent normal tissues of five HCC patients using high-throughput microarray chips. Then the expression levels of 10 differentially expressed lncRNAs were verified in 50 pairs of tissue samples from patients with HCC by quantitative real-time PCR (qRT-PCR). The oncogenic effects of lncRNA-4045 (ENST00000524045.6) in HCC cell lines were verified through a series of in vitro experiments including CCK-8 assay, plate clone formation assay, transwell assay, scratch assay, and flow cytometry. Subsequently, the potential target genes of lncRNA-4045 were predicted by bioinformatics analysis, fluorescence in situ hybridization assay, and RNA sequencing. The mechanism of lncRNA-4045 in HCC was explored by WB assay as well as rescue and enhancement experiments. RESULTS: The results from microarray chips showed 1,708 lncRNAs to have been significantly upregulated and 2725 lncRNAs to have been significantly downregulated in HCC tissues. Via validation in 50 HCC patients, a novel lncRNA lncRNA-4045 was found significantly upregulated in HCC tissues. Additionally, a series of in vitro experiments showed that lncRNA-4045 promoted the proliferation, invasion, and migration of HCC cell lines, and inhibited the apoptosis of HCC cell lines. The results of qRT-PCR in HCC tissues showed that the expression levels of AKR1B10 were significantly positively correlated with lncRNA-4045. LncRNA-4045 knockdown significantly down-regulated AKR1B10 protein expression, and overexpression of lncRNA-4045 led to significant up-regulation of AKR1B10 protein in HCC cell lines. Lastly, down-regulation of AKR1B10 could partially eliminate the enhancement of cell proliferation induced by lncRNA-4045 overexpression, while up-regulation of AKR1B10 was shown to enhance those effects. CONCLUSION: LncRNA-4045 may promote HCC via enhancement of the expression of AKR1B10 protein.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Prolifération cellulaire/génétique , Aldose reductase/génétique , Aldose reductase/métabolisme , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , Lignée cellulaire tumorale , Mâle , Mouvement cellulaire/génétique , Évolution de la maladie , Femelle , Adulte d'âge moyen
12.
J Hazard Mater ; 470: 134212, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38583205

RÉSUMÉ

Elevated levels of cadmium (Cd) have the ability to impede plant development. Aldo-keto reductases (AKRs) have been demonstrated in a number of plant species to improve tolerance to a variety of abiotic stresses by scavenging cytotoxic aldehydes; however, only a few AKRs have been identified to improve Cd tolerance. The OsAKR1 gene was extracted and identified from rice here. After being exposed to Cd, the expression of OsAKR1 dramatically rose in both roots and shoots, although more pronounced in roots. According to a subcellular localization experiment, the nucleus and cytoplasm are where OsAKR1 is primarily found. Mutants lacking OsAKR1 exhibited Cd sensitive phenotype than that of the wild-type (WT) Nipponbare (Nip), and osakr1 mutants exhibited reduced capacity to scavenge methylglyoxal (MG). Furthermore, osakr1 mutants exhibited considerably greater hydrogen peroxide (H2O2) and malondialdehyde (MDA) levels, and increased catalase (CAT) activity in comparison to Nip. The expression of three isomeric forms of CAT was found to be considerably elevated in osakr1 mutants during Cd stress, as demonstrated by quantitative real-time PCR analysis, when compared to Nip. These results imply that OsAKR1 controlled rice's ability to withstand Cd by scavenging harmful aldehydes and turning on the reactive oxygen species (ROS) scavenging mechanism.


Sujet(s)
Aldo-keto reductases , Cadmium , Oryza , Oryza/génétique , Oryza/métabolisme , Oryza/effets des médicaments et des substances chimiques , Oryza/croissance et développement , Cadmium/toxicité , Cadmium/métabolisme , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , Aldéhydes/métabolisme , Catalase/métabolisme , Catalase/génétique , Aldose reductase/génétique , Aldose reductase/métabolisme , Malonaldéhyde/métabolisme , Stress physiologique , Méthylglyoxal/métabolisme , Régulation de l'expression des gènes végétaux/effets des médicaments et des substances chimiques , Peroxyde d'hydrogène/métabolisme , Protéines végétales/génétique , Protéines végétales/métabolisme , Mutation , Racines de plante/métabolisme , Racines de plante/effets des médicaments et des substances chimiques , Racines de plante/génétique , Inactivation métabolique
13.
Angew Chem Int Ed Engl ; 63(22): e202403539, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38556813

RÉSUMÉ

The design and orderly layered co-immobilization of multiple enzymes on resin particles remain challenging. In this study, the SpyTag/SpyCatcher binding pair was fused to the N-terminus of an alcohol dehydrogenase (ADH) and an aldo-keto reductase (AKR), respectively. A non-canonical amino acid (ncAA), p-azido-L-phenylalanine (p-AzF), as the anchor for covalent bonding enzymes, was genetically inserted into preselected sites in the AKR and ADH. Employing the two bioorthogonal counterparts of SpyTag/SpyCatcher and azide-alkyne cycloaddition for the immobilization of AKR and ADH enabled sequential dual-enzyme coating on porous microspheres. The ordered dual-enzyme reactor was subsequently used to synthesize (S)-1-(2-chlorophenyl)ethanol asymmetrically from the corresponding prochiral ketone, enabling the in situ regeneration of NADPH. The reactor exhibited a high catalytic conversion of 74 % and good reproducibility, retaining 80 % of its initial activity after six cycles. The product had 99.9 % ee, which that was maintained in each cycle. Additionally, the double-layer immobilization method significantly increased the enzyme loading capacity, which was approximately 1.7 times greater than that of traditional single-layer immobilization. More importantly, it simultaneously enabled both the purification and immobilization of multiple enzymes on carriers, thus providing a convenient approach to facilitate cascade biocatalysis.


Sujet(s)
Alcohol dehydrogenase , Biocatalyse , Enzymes immobilisées , Enzymes immobilisées/composition chimique , Enzymes immobilisées/métabolisme , Alcohol dehydrogenase/métabolisme , Alcohol dehydrogenase/composition chimique , Alcohol dehydrogenase/génétique , Ingénierie des protéines , Aldo-keto reductases/métabolisme , Aldo-keto reductases/composition chimique , Aldo-keto reductases/génétique , Phénylalanine/composition chimique , Phénylalanine/métabolisme , Phénylalanine/analogues et dérivés , Azotures/composition chimique
14.
Nat Commun ; 15(1): 2128, 2024 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-38459030

RÉSUMÉ

Modulation of protein function through allosteric regulation is central in biology, but biomacromolecular systems involving multiple subunits and ligands may exhibit complex regulatory mechanisms at different levels, which remain poorly understood. Here, we discover an aldo-keto reductase termed AKRtyl and present its three-level regulatory mechanism. Specifically, by combining steady-state and transient kinetics, X-ray crystallography and molecular dynamics simulation, we demonstrate that AKRtyl exhibits a positive synergy mediated by an unusual Monod-Wyman-Changeux (MWC) paradigm of allosteric regulation at low concentrations of the cofactor NADPH, but an inhibitory effect at high concentrations is observed. While the substrate tylosin binds at a remote allosteric site with positive cooperativity. We further reveal that these regulatory mechanisms are conserved in AKR12D subfamily, and that substrate cooperativity is common in AKRs across three kingdoms of life. This work provides an intriguing example for understanding complex allosteric regulatory networks.


Sujet(s)
Protéines , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , Site allostérique , Régulation allostérique , NADP/métabolisme , Cinétique
15.
Phys Chem Chem Phys ; 26(12): 9295-9308, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38469695

RÉSUMÉ

Understanding selectivity mechanisms of inhibitors towards highly homologous proteins is of paramount importance in the design of selective candidates. Human aldo-keto reductases (AKRs) pertain to a superfamily of monomeric oxidoreductases, which serve as NADPH-dependent cytosolic enzymes to catalyze the reduction of carbonyl groups to primary and secondary alcohols using electrons from NADPH. Among AKRs, AKR1B1 is emerging as a promising target for cancer treatment and diabetes, despite its high structural similarity with AKR1B10, which leads to severe adverse events. Therefore, it is crucial to understand the selectivity mechanisms of AKR1B1 and AKR1B10 to discover safe anticancer candidates with optimal therapeutic efficacy. In this study, multiple computational strategies, including sequence alignment, structural comparison, Protein Contacts Atlas analysis, molecular docking, molecular dynamics simulation, MM-GBSA calculation, alanine scanning mutagenesis and pharmacophore modeling analysis were employed to comprehensively understand the selectivity mechanisms of AKR1B1/10 inhibition based on selective inhibitor lidorestat and HAHE. This study would provide substantial evidence in the design of potent and highly selective AKR1B1/10 inhibitors in future.


Sujet(s)
Antienzymes , Simulation de dynamique moléculaire , Humains , Simulation de docking moléculaire , NADP/métabolisme , Aldo-keto reductases/métabolisme , Antienzymes/pharmacologie , Aldose reductase/métabolisme
16.
Appl Environ Microbiol ; 90(4): e0015024, 2024 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-38551341

RÉSUMÉ

Avilamycins, which possess potent inhibitory activity against Gram-positive bacteria, are a group of oligosaccharide antibiotics produced by Streptomyces viridochromogenes. Among these structurally related oligosaccharide antibiotics, avilamycin A serves as the main bioactive component in veterinary drugs and animal feed additives, which differs from avilamycin C only in the redox state of the two-carbon branched-chain of the terminal octose moiety. However, the mechanisms underlying assembly and modification of the oligosaccharide chain to diversify individual avilamycins remain poorly understood. Here, we report that AviZ1, an aldo-keto reductase in the avilamycin pathway, can catalyze the redox conversion between avilamycins A and C. Remarkably, the ratio of these two components produced by AviZ1 depends on the utilization of specific redox cofactors, namely NADH/NAD+ or NADPH/NADP+. These findings are inspired by gene disruption and complementation experiments and are further supported by in vitro enzymatic activity assays, kinetic analyses, and cofactor affinity studies on AviZ1-catalyzed redox reactions. Additionally, the results from sequence analysis, structure prediction, and site-directed mutagenesis of AviZ1 validate it as an NADH/NAD+-favored aldo-keto reductase that primarily oxidizes avilamycin C to form avilamycin A by utilizing abundant NAD+ in vivo. Building upon the biological function and catalytic activity of AviZ1, overexpressing AviZ1 in S. viridochromogenes is thus effective to improve the yield and proportion of avilamycin A in the fermentation profile of avilamycins. This study represents, to our knowledge, the first characterization of biochemical reactions involved in avilamycin biosynthesis and contributes to the construction of high-performance strains with industrial value.IMPORTANCEAvilamycins are a group of oligosaccharide antibiotics produced by Streptomyces viridochromogenes, which can be used as veterinary drugs and animal feed additives. Avilamycin A is the most bioactive component, differing from avilamycin C only in the redox state of the two-carbon branched-chain of the terminal octose moiety. Currently, the biosynthetic pathway of avilamycins is not clear. Here, we report that AviZ1, an aldo-keto reductase in the avilamycin pathway, can catalyze the redox conversion between avilamycins A and C. More importantly, AviZ1 exhibits a unique NADH/NAD+ preference, allowing it to efficiently catalyze the oxidation of avilamycin C to form avilamycin A using abundant NAD+ in cells. Thus, overexpressing AviZ1 in S. viridochromogenes is effective to improve the yield and proportion of avilamycin A in the fermentation profile of avilamycins. This study serves as an enzymological guide for rational strain design, and the resulting high-performance strains have significant industrial value.


Sujet(s)
NAD , Streptomyces , Médicaments vétérinaires , NAD/métabolisme , Aldo-keto reductases/métabolisme , Oligosaccharides , Oxydoréduction , Antibactériens , Carbone/métabolisme , NADP/métabolisme , Aldose reductase/métabolisme
17.
Chem Biol Interact ; 391: 110896, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38301882

RÉSUMÉ

Aldo-keto reductase-7A (AKR7A) subfamily belongs to the AKR superfamily and is associated with detoxification of aldehydes and ketones by reducing them to the corresponding alcohols. So far five members of ARK7A subfamily are identified: two human members-AKR7A2 and AKR7A3, two rat members-AKR7A1 and AKR7A4, and one mouse member-AKR7A5, which are implicated in several diseases including neurodegenerative diseases and cancer. AKR7A members share similar crystal structures and protein functional domains, but have different substrate specificity, inducibility and biological functions. This review will summarize the research progress of AKR7A members in substrate specificity, tissue distribution, inducibility, crystal structure and biological function. The significance of AKR7A members in the occurrence and development of diseases will also be discussed.


Sujet(s)
Aldose reductase , Foie , Rats , Souris , Animaux , Humains , Aldo-keto reductases/métabolisme , Foie/métabolisme , Aldose reductase/métabolisme , Alcohol oxidoreductases/métabolisme , Spécificité du substrat
18.
Life Sci ; 336: 122336, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38092142

RÉSUMÉ

AIMS: Akr1A1 is a glycolytic enzyme catalyzing the reduction of aldehyde to alcohol. This study aims to delineate the role of Akr1A1 in regulating the adipo-osteogenic lineage differentiation of mesenchymal stem cells (MSCs). MAIN METHODS: MSCs derived from human bone marrow and Wharton Jelly together with gain- and loss-of-function analysis as well as supplementation with the S-Nitrosoglutathione reductase (GSNOR) inhibitor N6022 were used to study the function of Akr1A1 in controlling MSC lineage differentiation into osteoblasts and adipocytes. KEY FINDINGS: Akr1A1 expression, PKM2 activity, and lactate production were found to be decreased in osteoblast-committed MSCs, but PGC-1α increased to induce mitochondrial oxidative phosphorylation. Increased Akr1A1 inhibited the SIRT1-dependent pathway for decreasing the expressions of PGC-1α and TAZ but increasing PPAR γ in adipocyte-committed MSCs, hence promoting glycolysis in adipogenesis. In contrast, Akr1A1 expression, PKM2 activity and lactate production were all increased in adipocyte-differentiated cells with decreased PGC-1α for switching energy utilization to glycolytic metabolism. Reduced Akr1A1 expression in osteoblast-committed cells relieves its inhibition of SIRT1-mediated activation of PGC-1α and TAZ for facilitating osteogenesis and mitochondrial metabolism. SIGNIFICANCE: Several metabolism-involved regulators including Akr1A1, SIRT1, PPARγ, PGC-1α and TAZ were differentially expressed in osteoblast- and adipocyte-committed MSCs. More importantly, Akr1A1 was identified as a new key regulator for controlling the MSC lineage commitment in favor of adipogenesis but detrimental to osteogenesis. Such information should be useful to develop perspective new therapeutic agents to reverse the adipo-osteogenic differentiation of BMSCs, in a way to increase in osteogenesis but decrease in adipogenesis.


Sujet(s)
Adipogenèse , Cellules souches mésenchymateuses , Humains , Adipogenèse/physiologie , Ostéogenèse/physiologie , Sirtuine-1/métabolisme , Différenciation cellulaire/physiologie , Lactates/métabolisme , Aldo-keto reductases/métabolisme
19.
Microb Cell Fact ; 22(1): 213, 2023 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-37840127

RÉSUMÉ

Enantio-pure α-hydroxy amides are valuable intermediates for the synthesis of chiral pharmaceuticals. The asymmetric reduction of α-keto amides to generate chiral α-hydroxy amides is a difficult and challenging task in biocatalysis. In this study, iolS, an aldo-keto reductase from Bacillus subtilis 168 was exhibited as a potential biocatalyst, which could catalyze the reduction of diaryl α-keto amide such as 2-oxo-N, 2-diphenyl-acetamide (ONDPA) with moderate S-selectivity (76.1%, ee) and 60.5% conversion. Through semi-rational engineering, two stereocomplementary variants (I57F/F126L and N21A/F126A) were obtained with ee value of 97.6% (S) and 99.9% (R) toward ONDPA (1a), respectively, delivering chiral α-hydroxy amide with > 98% conversions. Moreover, the excellent S- and R-preference variants displayed improved stereoselectivities toward the other α-keto amide compounds. Molecular dynamic and docking analysis revealed that the two key residues at 21 and 126 were identified as the "switch", which specifically controlled the stereopreference of iolS by regulating the shape of substrate binding pocket as well as the substrate orientation. Our results offer an effective strategy to obtain α-hydroxy amides with high optical purity and provide structural insights into altering the stereoselectivity of AKRs.


Sujet(s)
Aldose reductase , Amides , Aldo-keto reductases/génétique , Aldo-keto reductases/composition chimique , Aldo-keto reductases/métabolisme , Spécificité du substrat , Biocatalyse , Catalyse , Aldose reductase/métabolisme
20.
Drug Metab Dispos ; 51(12): 1569-1577, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37722844

RÉSUMÉ

Enzymes catalyzing the reduction reaction of xenobiotics are mainly members of the aldo-keto reductase (AKR) and short-chain dehydrogenase/reductase (SDR) superfamilies. The intestine, together with the liver, is responsible for first-pass effects and is an organ that determines the bioavailability of orally administered drugs. In this study, we evaluated the mRNA and protein expression levels of 12 AKR isoforms (AKR1A1, AKR1B1, AKR1B10, AKR1B15, AKR1C1, AKR1C2, AKR1C3, AKR1C4, AKR1D1, AKR1E2, AKR7A2, and AKR7A3) and 7 SDR isoforms (CBR1, CBR3, CBR4, DCXR, DHRS4, HSD11B1, and HSD17B12) in each region of the human intestine using next-generation sequencing and data-independent acquisition proteomics. At both the mRNA and protein levels, most AKR isoforms were highly expressed in the upper regions of the intestine, namely the duodenum and jejunum, and then declined toward the rectum. Among the members in the SDR superfamily, CBR1 and DHRS4 were highly expressed in the upper regions, whereas the expression levels of the other isoforms were almost uniform in all regions. Significant positive correlations between mRNA and protein levels were observed in AKR1A1, AKR1B1, AKR1B10, AKR1C3, AKR7A2, AKR7A3, CBR1, and CBR3. The mRNA level of AKR1B10 was highest, followed by AKR7A3 and CBR1, each accounting for more than 10% of the sum of all AKR and SDR levels in the small intestine. This expression profile in the human intestine was greatly different from that in the human liver, where AKR1C isoforms are predominantly expressed. SIGNIFICANCE STATEMENT: In this study comprehensively determined the mRNA and protein expression profiles of aldo-keto reductase (AKR) and short-chain dehydrogenase/reductase isoforms involved in xenobiotic metabolism in the human intestine and found that most of them are highly expressed in the upper region, where AKR1B10, AKR7A3, and CBR1 are predominantly expressed. Since the intestine is significantly involved in the metabolism of orally administered drugs, the information provided here is valuable for pharmacokinetic studies in drug development.


Sujet(s)
Short chain dehydrogenase-reductases , Humains , Aldo-keto reductases/génétique , Aldo-keto reductases/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Aldose reductase/génétique , Aldose reductase/métabolisme , Isoformes de protéines/génétique , Oxidoreductases/génétique , Oxidoreductases/métabolisme , Intestins
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE