Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
FEMS Yeast Res ; 242024 Jan 09.
Article in English | MEDLINE | ID: mdl-39009031

ABSTRACT

Lignocellulose (dry plant biomass) is an abundant cheap inedible residue of agriculture and wood industry with great potential as a feedstock for biotechnological processes. Lignocellulosic substrates can serve as valuable resources in fermentation processes, allowing the production of a wide array of chemicals, fuels, and food additives. The main obstacle for cost-effective conversion of lignocellulosic hydrolysates to target products is poor metabolism of the major pentoses, xylose and L-arabinose, which are the second and third most abundant sugars of lignocellulose after glucose. We study the oversynthesis of riboflavin in the flavinogenic yeast Candida famata and found that all major lignocellulosic sugars, including xylose and L-arabinose, support robust growth and riboflavin synthesis in the available strains of C. famata. To further increase riboflavin production from xylose and lignocellulose hydrolysate, genes XYL1 and XYL2 coding for xylose reductase and xylitol dehydrogenase were overexpressed. The resulting strains exhibited increased riboflavin production in both shake flasks and bioreactors using diluted hydrolysate, reaching 1.5 g L-1.


Subject(s)
Candida , Lignin , Metabolic Engineering , Riboflavin , Xylose , Lignin/metabolism , Riboflavin/metabolism , Riboflavin/biosynthesis , Candida/metabolism , Candida/genetics , Xylose/metabolism , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Fermentation , Bioreactors/microbiology , D-Xylulose Reductase/metabolism , D-Xylulose Reductase/genetics , Arabinose/metabolism
2.
Int J Parasitol Drugs Drug Resist ; 25: 100555, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38996597

ABSTRACT

Aldo-keto reductases (AKRs), a superfamily of NADP(H)-dependent oxidoreductases, catalyze the oxidoreduction of a wide variety of eobiotic and xenobiotic aldehydes and ketones. In mammals, AKRs play essential roles in hormone and xenobiotic metabolism, oxidative stress, and drug resistance, but little is known about these enzymes in the parasitic nematode Haemonchus contortus. In the present study, 22 AKR genes existing in the H. contortus genome were investigated and a phylogenetic analysis with comparison to AKRs in Caenorhabditis elegans, sheep and humans was conducted. The constitutive transcription levels of all AKRs were measured in eggs, larvae, and adults of H. contortus, and their expression was compared in a drug-sensitive strain (ISE) and a benzimidazole-resistant strain (IRE) previously derived from the sensitive strain by imposing benzimidazole selection pressure. In addition, the inducibility of AKRs by exposure of H. contortus adults to benzimidazole anthelmintic flubendazole in vitro was tested. Phylogenetic analysis demonstrated that the majority of AKR genes in H. contortus lack orthologues in the sheep genome, which is a favorable finding for considering AKRs as potential drug targets. Large differences in the expression levels of individual AKRs were observed, with AKR1, AKR3, AKR8, and AKR10 being the most highly expressed at most developmental stages. Significant changes in the expression of AKRs during the life cycle and pronounced sex differences were found. Comparing the IRE and ISE strains, three AKRs were upregulated, and seven AKRs were downregulated in adults. In addition, the expression of three AKRs was induced by flubendazole exposure in adults of the ISE strain. Based on these results, AKR1, AKR2, AKR3, AKR5, AKR10 and AKR19 in particular merit further investigation and functional characterization with respect to their potential involvement in drug biotransformation and anthelmintic resistance in H. contortus.


Subject(s)
Aldo-Keto Reductases , Haemonchus , Mebendazole , Phylogeny , Animals , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/metabolism , Haemonchus/genetics , Haemonchus/drug effects , Haemonchus/enzymology , Mebendazole/pharmacology , Mebendazole/analogs & derivatives , Female , Male , Drug Resistance/genetics , Sheep , Anthelmintics/pharmacology , Transcriptome , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Humans , Caenorhabditis elegans/genetics , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Benzimidazoles/pharmacology
3.
Am J Physiol Renal Physiol ; 327(3): F489-F503, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38991008

ABSTRACT

Fate mapping and genetic manipulation of renin cells have relied on either noninducible Cre lines that can introduce the developmental effects of gene deletion or bacterial artificial chromosome transgene-based inducible models that may be prone to spurious and/or ectopic gene expression. To circumvent these problems, we generated an inducible mouse model in which CreERT2 is under the control of the endogenous Akr1b7 gene, an independent marker of renin cells that is expressed in a few extrarenal tissues. We confirmed the proper expression of Cre using Akr1b7CreERT2/+;R26RmTmG/+ mice in which Akr1b7+/renin+ cells become green fluorescent protein (GFP)+ upon tamoxifen administration. In embryos and neonates, GFP was found in juxtaglomerular cells, along the arterioles, and in the mesangium, and in adults, GFP was present mainly in juxtaglomerular cells. In mice treated with captopril and a low-salt diet to induce recruitment of renin cells, GFP extended along the afferent arterioles and in the mesangium. We generated Akr1b7CreERT2/+;Ren1cFl/-;R26RmTmG/+ mice to conditionally delete renin in adult mice and found a marked reduction in kidney renin mRNA and protein and mean arterial pressure in mutant animals. When subjected to a homeostatic threat, mutant mice were unable to recruit renin+ cells. Most importantly, these mice developed concentric vascular hypertrophy ruling out potential developmental effects on the vasculature due to the lack of renin. We conclude that Akr1b7CreERT2 mice constitute an excellent model for the fate mapping of renin cells and for the spatial and temporal control of gene expression in renin cells.NEW & NOTEWORTHY Fate mapping and genetic manipulation are important tools to study the identity of renin cells. Here, we report on a novel Cre mouse model, Akr1b7CreERT2, for the spatial and temporal regulation of gene expression in renin cells. Cre is properly expressed in renin cells during development and in the adult under basal conditions and under physiological stress. Moreover, renin can be efficiently deleted in the adult, leading to the development of concentric vascular hypertrophy.


Subject(s)
Mice, Transgenic , Renin , Animals , Renin/metabolism , Renin/genetics , Mice , Juxtaglomerular Apparatus/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Captopril/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism
5.
Chem Biol Interact ; 398: 111111, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38878851

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Databases, Protein , Aldo-Keto Reductases/metabolism , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/chemistry , Humans , Internet , Aldehyde Reductase/metabolism , Aldehyde Reductase/chemistry , Aldehyde Reductase/genetics , Animals
6.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1909-1923, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38914500

ABSTRACT

Galactitol, a rare sugar alcohol, has promising potential in the food industry and pharmaceutical field. The available industrial production methods rely on harsh hydrogenation processes, which incur high costs and environmental concerns. It is urgent to develop environmentally friendly and efficient biosynthesis technologies. In this study, a xylose reductase named AnXR derived from Aspergillus niger CBS 513.88 was identified and characterized for the enzymatic properties. AnXR exhibited the highest activity at 25 ℃ and pH 8.0, and it belonged to the NADPH-dependent aldose reductase family. To engineer a strain for galactitol production, we deleted the galactokinase (GAL1) gene in Saccharomyes cerevisiae by using the recombinant gene technology, which significantly reduced the metabolic utilization of D-galactose by host cells. Subsequently, we introduced the gene encoding AnXR into this modified strain, creating an engineered strain capable of catalyzing the conversion of D-galactose into galactitol. Furthermore, we optimized the whole-cell catalysis conditions for the engineered strain, which achieved a maximum galactitol yield of 12.10 g/L. Finally, we tested the reduction ability of the strain for other monosaccharides and discovered that it could produce functional sugar alcohols such as xylitol and arabinitol. The engineered strain demonstrates efficient biotransformation capabilities for galactitol and other functional sugar alcohols, representing a significant advancement in environmentally sustainable production practices.


Subject(s)
Aldehyde Reductase , Aspergillus niger , Galactitol , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Galactitol/metabolism , Galactitol/genetics , Aspergillus niger/metabolism , Aspergillus niger/genetics , Galactose/metabolism , Metabolic Engineering/methods , Fermentation , Industrial Microbiology , Galactokinase/genetics , Galactokinase/metabolism
7.
Mar Drugs ; 22(6)2024 May 22.
Article in English | MEDLINE | ID: mdl-38921547

ABSTRACT

Clavatols exhibit a wide range of biological activities due to their diverse structures. A genome mining strategy identified an A5cla cluster from Penicillium sp. MYA5, derived from the Arctic plant Dryas octopetala, is responsible for clavatol biosynthesis. Seven clavatols, including one new clavatol derivate named penicophenone F (1) and six known clavatols (2-7), were isolated from Penicillium sp. MYA5 using a transcriptome mining strategy. These structures were elucidated by comprehensive spectroscopic analysis. Antibacterial, aldose reductase inhibition, and siderophore-producing ability assays were conducted on compounds 1-7. Compounds 1 and 2 demonstrated inhibitory effects on the ALR2 enzyme with inhibition rates of 75.3% and 71.6% at a concentration of 10 µM, respectively. Compound 6 exhibited antibacterial activity against Staphylococcus aureus and Escherichia coli with MIC values of 4.0 µg/mL and 4.0 µg/mL, respectively. Additionally, compounds 1, 5, and 6 also showed potential iron-binding ability.


Subject(s)
Anti-Bacterial Agents , Penicillium , Staphylococcus aureus , Penicillium/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Genomics/methods , Escherichia coli/drug effects , Escherichia coli/genetics , Microbial Sensitivity Tests , Transcriptome , Arctic Regions , Siderophores/pharmacology , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/genetics
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167214, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38718846

ABSTRACT

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.


Subject(s)
Aldehyde Reductase , Aldo-Keto Reductases , Carcinoma, Pancreatic Ductal , Cellular Senescence , Gemcitabine , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Animals , Humans , Mice , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/antagonists & inhibitors , Aldo-Keto Reductases/metabolism , Aldo-Keto Reductases/genetics , Carcinogenesis/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Proliferation , Cellular Senescence/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Mice, Nude , Oleanolic Acid/pharmacology , Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/drug therapy
9.
J Ethnopharmacol ; 332: 118354, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38762210

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Berberine , Diet, High-Fat , Lipid Metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Humans , Berberine/pharmacology , Berberine/therapeutic use , Hep G2 Cells , Male , Diet, High-Fat/adverse effects , Lipid Metabolism/drug effects , Mice , Aldo-Keto Reductases/metabolism , Aldo-Keto Reductases/genetics , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Glucose/metabolism , Liver/drug effects , Liver/metabolism , Insulin Resistance
10.
Dev Cell ; 59(15): 1954-1971.e7, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38776924

ABSTRACT

A significant variation in chromatin accessibility is an epigenetic feature of leukemia. The cause of this variation in leukemia, however, remains elusive. Here, we identify SMARCA5, a core ATPase of the imitation switch (ISWI) chromatin remodeling complex, as being responsible for aberrant chromatin accessibility in leukemia cells. We find that SMARCA5 is required to maintain aberrant chromatin accessibility for leukemogenesis and then promotes transcriptional activation of AKR1B1, an aldo/keto reductase, by recruiting transcription co-activator DDX5 and transcription factor SP1. Higher levels of AKR1B1 are associated with a poor prognosis in leukemia patients and promote leukemogenesis by reprogramming fructose metabolism. Moreover, pharmacological inhibition of AKR1B1 has been shown to have significant therapeutic effects in leukemia mice and leukemia patient cells. Thus, our findings link the aberrant chromatin state mediated by SMARCA5 to AKR1B1-mediated endogenous fructose metabolism reprogramming and shed light on the essential role of AKR1B1 in leukemogenesis, which may provide therapeutic strategies for leukemia.


Subject(s)
Fructose , Humans , Animals , Mice , Fructose/metabolism , Chromatin/metabolism , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Leukemia/metabolism , Leukemia/pathology , Leukemia/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin Assembly and Disassembly , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/genetics , Adenosine Triphosphatases
12.
J Hazard Mater ; 470: 134212, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583205

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Cadmium , Oryza , Oryza/genetics , Oryza/metabolism , Oryza/drug effects , Oryza/growth & development , Cadmium/toxicity , Cadmium/metabolism , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/metabolism , Aldehydes/metabolism , Catalase/metabolism , Catalase/genetics , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Malondialdehyde/metabolism , Stress, Physiological , Pyruvaldehyde/metabolism , Gene Expression Regulation, Plant/drug effects , Hydrogen Peroxide/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Mutation , Plant Roots/metabolism , Plant Roots/drug effects , Plant Roots/genetics , Inactivation, Metabolic
13.
FEMS Microbiol Lett ; 3712024 Jan 09.
Article in English | MEDLINE | ID: mdl-38664064

ABSTRACT

Thermo-acidic pretreatment of lignocellulosic biomass is required to make it amenable to microbial metabolism and results in generation of furfural due to breakdown of pentose sugars. Furfural is toxic to microbial metabolism and results in reduced microbial productivity and increased production costs. This study asks if deletion of yghZ gene which encodes a NADPH-dependent aldehyde reductase enzyme results in improved furfural tolerance in Escherichia coli host. The ∆yghZ strain-SSK201-was tested for tolerance to furfural in presence of 5% xylose as a carbon source in AM1 minimal medium. At 96 h and in presence of 1.0 g/L furfural, the culture harboring strain SSK201 displayed 4.5-fold higher biomass, 2-fold lower furfural concentration and 15.75-fold higher specific growth rate (µ) as compared to the parent strain SSK42. The furfural tolerance advantage of SSK201 was retained when the carbon source was switched to glucose in AM1 medium and was lost in rich LB medium. The findings have potential to be scaled up to a hydrolysate culture medium, which contains furan inhibitors and lack nutritionally rich components, under bioreactor cultivation and observe growth advantage of the ∆yghZ host. It harbors potential to generate robust industrial strains which can convert lignocellulosic carbon into metabolites of interest in a cost-efficient manner.


Subject(s)
Carbon , Escherichia coli Proteins , Escherichia coli , Furaldehyde , Xylose , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Biomass , Carbon/metabolism , Culture Media/chemistry , Culture Media/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Furaldehyde/metabolism , Gene Deletion , Glucose/metabolism , Xylose/metabolism
14.
Dig Dis Sci ; 69(7): 2502-2521, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38662158

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Carcinoma, Hepatocellular , Gene Expression Regulation, Neoplastic , Liver Neoplasms , RNA, Long Noncoding , Female , Humans , Male , Middle Aged , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
15.
Chem Biol Interact ; 392: 110905, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38373627

ABSTRACT

Aldose reductase is a member of the 1B1 subfamily of aldo-keto reductase gene superfamily. The action of aldose reductase (AR) has been implicated in the pathogenesis of a variety of disease states, most notably complications of diabetes mellitus including neuropathy, retinopathy, nephropathy, and cataracts. To explore for mechanistic roles for AR in disease pathogenesis, we established mutant strains produced using Crispr-Cas9 to inactivate the AKR1B3 gene in C57BL6 mice. Phenotyping AR-knock out (ARKO) strains confirmed previous reports of reduced accumulation of tissue sorbitol levels. Lens epithelial cells in ARKO mice showed markedly reduced epithelial-to-mesenchymal transition following lens extraction in a surgical model of cataract and posterior capsule opacification. A previously unreported phenotype of preputial sebaceous gland swelling was observed frequently in male ARKO mice homozygous for the mutant AKR1B3 allele. This condition, which was shown to be accompanied by infiltration of proinflammatory CD3+ lymphocytes, was not observed in WT mice or mice heterozygous for the mutant allele. Despite this condition, reproductive fitness of the ARKO strain was indistinguishable from WT mice housed under identical conditions. These studies establish the utility of a new strain of AKR1B3-null mice created to support mechanistic studies of cataract and diabetic eye disease.


Subject(s)
Capsule Opacification , Cataract , Lens, Crystalline , Animals , Male , Mice , Aldehyde Reductase/genetics , Capsule Opacification/pathology , Cataract/genetics , Cataract/pathology , Incidence , Inflammation/pathology , Lens, Crystalline/pathology , Mice, Inbred C57BL , Mice, Knockout , Sebaceous Glands
16.
Diabetes ; 73(7): 1188-1195, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38394643

ABSTRACT

Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease. Because many genes associate with DKD, multiomics approaches were used to narrow the list of functional genes, gene products, and related pathways providing insights into the pathophysiological mechanisms of DKD. The Kidney Precision Medicine Project human kidney single-cell RNA-sequencing (scRNA-seq) data set and Mendeley Data on human kidney cortex biopsy proteomics were used. The R package Seurat was used to analyze scRNA-seq data and data from a subset of proximal tubule cells. PathfindR was applied for pathway analysis in cell type-specific differentially expressed genes and the R limma package was used to analyze differential protein expression in kidney cortex. A total of 790 differentially expressed genes were identified in proximal tubule cells, including 530 upregulated and 260 downregulated transcripts. Compared with differentially expressed proteins, 24 genes or proteins were in common. An integrated analysis combining protein quantitative trait loci, genome-wide association study hits (namely, estimated glomerular filtration rate), and a plasma metabolomics analysis was performed using baseline metabolites predictive of DKD progression in our longitudinal Diabetes Heart Study samples. The aldo-keto reductase family 1 member A1 gene (AKR1A1) was revealed as a potential molecular hub for DKD cellular dysfunction in several cross-linked pathways featured by deficiency of this enzyme.


Subject(s)
Aldehyde Reductase , Biomarkers , Diabetic Nephropathies , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Humans , Biomarkers/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Proteomics/methods , Genome-Wide Association Study , Male , Kidney Tubules, Proximal/metabolism , Female , Middle Aged , Multiomics
17.
Exp Mol Med ; 56(1): 220-234, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38200154

ABSTRACT

Diabetes might be associated with increased cancer risk, with several studies reporting hyperglycemia as a primary oncogenic stimulant. Since glucose metabolism is linked to numerous metabolic pathways, it is difficult to specify the mechanisms underlying hyperglycemia-induced cancer progression. Here, we focused on the polyol pathway, which is dramatically activated under hyperglycemia and causes diabetic complications. We investigated whether polyol pathway-derived fructose facilitates hyperglycemia-induced gastric cancer metastasis. We performed bioinformatics analysis of gastric cancer datasets and immunohistochemical analyses of gastric cancer specimens, followed by transcriptomic and proteomic analyses to evaluate phenotypic changes in gastric cancer cells. Consequently, we found a clinical association between the polyol pathway and gastric cancer progression. In gastric cancer cell lines, hyperglycemia enhanced cell migration and invasion, cytoskeletal rearrangement, and epithelial-mesenchymal transition (EMT). The hyperglycemia-induced acquisition of metastatic potential was mediated by increased fructose derived from the polyol pathway, which stimulated the nuclear ketohexokinase-A (KHK-A) signaling pathway, thereby inducing EMT by repressing the CDH1 gene. In two different xenograft models of cancer metastasis, gastric cancers overexpressing AKR1B1 were found to be highly metastatic in diabetic mice, but these effects of AKR1B1 were attenuated by KHK-A knockdown. In conclusion, hyperglycemia induces fructose formation through the polyol pathway, which in turn stimulates the KHK-A signaling pathway, driving gastric cancer metastasis by inducing EMT. Thus, the polyol and KHK-A signaling pathways could be potential therapeutic targets to decrease the metastatic risk in gastric cancer patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Polymers , Stomach Neoplasms , Humans , Animals , Mice , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Proteomics , Signal Transduction , Hyperglycemia/complications , Fructokinases/genetics , Fructokinases/metabolism , Fructose/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Movement/genetics , Cell Line, Tumor , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Aldehyde Reductase/pharmacology
18.
Arch. endocrinol. metab. (Online) ; 66(1): 12-18, Jan.-Feb. 2022. tab
Article in English | LILACS | ID: biblio-1364310

ABSTRACT

ABSTRACT Objective: The AKR1B1 gene encodes an enzyme that catalyzes the reduction of glucose into sorbitol. Chronic hyperglycemia in patients with diabetes mellitus (DM) leads to increased AKR1B1 affinity for glucose and, consequently, sorbitol accumulation. Elevated sorbitol increases oxidative stress, which is one of the main pathways related to chronic complications of diabetes, including diabetic kidney disease (DKD). Accordingly, some studies have suggested the rs759853 polymorphism in the AKR1B1 gene is associated with DKD; however, findings are still contradictory. The aim was to investigate the association of the rs759853 polymorphism in the AKR1B1 gene and DKD. Materials and methods: The sample comprised 695 patients with type 2 DM (T2DM) and DKD (cases) and 310 patients with T2DM of more than 10 years' duration, but no DKD (controls). The polymorphism was genotyped by real-time PCR. Results: Allelic and genotype frequencies of this polymorphism did not differ significantly between groups. However, the A/A genotype was associated with risk for DKD after adjustment for gender, triglycerides, BMI, presence of hypertension and diabetic retinopathy, and duration of DM, under both recessive (P = 0.048) and additive (P = 0.037) inheritance models. Conclusion: Our data suggest an association between the AKR1B1 rs759853A/A genotype and risk for DKD in Brazilians T2DM patients.


Subject(s)
Humans , Aldehyde Reductase/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/complications , Diabetic Nephropathies/genetics , Case-Control Studies , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Alleles , Gene Frequency , Genotype
19.
Rev. méd. Chile ; 127(4): 399-409, abr. 1999. ilus, tab
Article in Spanish | LILACS | ID: lil-243910

ABSTRACT

Background: Recent studies suggest that polymorphisms associated to the aldose reductase gene could be related to early retinopathy in noninsulin dependent diabetics (NIDDM). There is also new interest on the genetic modulation of coagulation factors in relation to this complication. Aim: To look for a possible relationship between the rate of appearance of retinopathy and the genotype of (AC)n polymorphic marker associated to aldose reductase gene. Patients and methods: A random sample of 27 NIDDM, aged 68.1 ñ 10.6 years, with a mean diabetes duration of 20.7 ñ 4.8 years and a mean glycosilated hemoglobin of 10.6 ñ 1.6 percent, was studied. The genotype of the (AC)n, polymorphic marker associated to the 5Õ end of the aldose reductase (ALR2) gene was determined by 32P-PCR plus sequenciation. Mutations of the factor XIII-A gene were studied by single stranded conformational polymorphism, sequenciation and restriction fragment length polymorphism. Results: Four patients lacked the (AC)24 and had a higher rate of appearance of retinopathy than patients with the (AC)24 allele (0.0167 and 0.0907 score points per year respectively, p=0.047). Both groups had similar glycosilated hemoglobin (11.7 ñ 0.2 and 10.5 ñ 1.6 percent respectively). Factor XIII gene mutations were not related to the rate of appearance of retinopathy. Conclusions: Our data suggest that the absence of the (AC)24 allele of the (AC)n polymorphic marker associated to the 5Õ end of the aldose reductase gene, is associated to a five fold reduction of retinopathy appearance rate


Subject(s)
Humans , Aldehyde Reductase/genetics , Diabetic Retinopathy/genetics , Glycated Hemoglobin , Diabetes Mellitus, Type 2/complications , Electrophoresis , Alleles , Biomarkers , Polymorphism, Genetic , Diabetic Retinopathy/etiology
SELECTION OF CITATIONS
SEARCH DETAIL