Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.714
Filter
1.
Medicina (Kaunas) ; 60(9)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39336582

ABSTRACT

Background and Objectives: Diabetes is a global health issue, with approximately 50% of patients developing diabetic nephropathy (DN) and 25% experiencing early and severe forms of the disease. The genetic factors contributing to rapid disease progression in a subset of these patients are unclear. This study investigates genetic variations in the GLO-1, CBR-1, and ACE genes associated with early and severe DN. Materials and Methods: Sanger DNA sequencing of the exons of CBR1, GLO1, and ACE genes was conducted in 113 patients with early and severe DN (defined as occurring within 10 years of the diagnosis of diabetes and with eGFR < 45 mL/min/1.73 m2) and 100 controls. The impact of identified genetic variations was analyzed using computational protein models created in silico with SWISS-Model and SWISS-Dock for ligand binding interactions. Results: In GLO1, two heterozygous missense mutations, c.102G>T and c.147C>G, and one heterozygous nonsense mutation, c.148G>T, were identified in patients. The SNP rs1049346 (G>A) at location 6:38703061 (GRCh38) was clinically significant. The c.147C>G mutation (C19S) was associated with ligand binding disruption in the GLO1 protein, while the nonsense mutation resulted in a truncated, non-functional protein. In CBR1, two heterozygous variations, one missense c.358G>A, and one silent mutation c.311G>C were observed, with the former (D120N) affecting the active site. No significant changes were noted in ACE gene variants concerning protein structure or function. Conclusions: The study identifies four novel and five recurrent mutations/polymorphisms in GLO1, ACE, and CBR1 genes associated with severe DN in Pakistani patients. Notably, a nonsense mutation in GLO1 led to a truncated, non-functional protein, while missense mutations in GLO1 and CBR1 potentially disrupt enzyme function, possibly accelerating DN progression.


Subject(s)
Diabetic Nephropathies , Lactoylglutathione Lyase , Peptidyl-Dipeptidase A , Humans , Diabetic Nephropathies/genetics , Female , Male , Middle Aged , Lactoylglutathione Lyase/genetics , Peptidyl-Dipeptidase A/genetics , Aged , Adult , Sequence Analysis, DNA/methods , Polymorphism, Single Nucleotide , Mutation, Missense , Aldehyde Reductase
2.
Nutrients ; 16(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39275261

ABSTRACT

Diabetic retinopathy (DR) is a specific microvascular problem of diabetes, which is mainly caused by hyperglycemia and may lead to rapid vision loss. Dietary polyphenols have been reported to decrease the risk of DR. Apocynum venetum L. leaves are rich in polyphenolic compounds and are popular worldwide for their health benefits as a national tea drink. Building on previous findings of antioxidant activity and aldose reductase inhibition of A. venetum, this study investigated the chemical composition of polyphenol-rich extract of A. venetum leaves (AVL) and its protective mechanism on ARPE-19 cells in hyperglycemia. Ninety-three compounds were identified from AVL by LC-MS/MS, including sixty-eight flavonoids, twenty-one organic acids, and four coumarins. AVL regulated the polyol pathway by decreasing the expression of aldose reductase and the content of sorbitol, enhancing the Na+K+-ATPase activity, and weakening intracellular oxidative stress effectively; it also could regulate the expression of autophagy-related proteins via the AMPK/mTOR/ULK1 signaling pathway to maintain intracellular homeostasis. AVL could restore the polyol pathway, inhibit oxidative stress, and maintain intracellular autophagy to protect cellular morphology and improve DR. The study reveals the phytochemical composition and protective mechanisms of AVL against DR, which could be developed as a functional food and/or candidate pharmaceutical, aiming for retina protection in diabetic retinopathy.


Subject(s)
Apocynum , Autophagy , Glucose , Oxidative Stress , Plant Extracts , Plant Leaves , Polyphenols , Retinal Pigment Epithelium , Humans , Plant Extracts/pharmacology , Polyphenols/pharmacology , Polyphenols/analysis , Plant Leaves/chemistry , Autophagy/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Glucose/metabolism , Glucose/adverse effects , Apocynum/chemistry , Oxidative Stress/drug effects , Polymers , Cell Line , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Signal Transduction/drug effects , Antioxidants/pharmacology , Aldehyde Reductase/metabolism
3.
Exp Cell Res ; 442(1): 114210, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39154929

ABSTRACT

Gastric cancer is a malignant tumor associated with a high mortality rate. Recently, emerging evidence has shown that ferroptosis, a regulated form of cell death induced by iron (Fe)-dependent lipid peroxidation. Nuclear factor E2 related factor 2 (NRF2) is a key regulator of intracellular oxidation homeostasis that plays a pivotal role in controlling lipid peroxidation, which is closely related to the process of ferroptosis. However, the molecular mechanism of NRF2 on ferroptosis remains to be investigated in gastric cancer. In our study, NRF 2 was found to transcriptionally activate Aldo-keto reductase 1 member B1 (AKR1B1) expression in gastric cancer. AKR1B1 is involved in the regulation of lipid metabolism by removing the aldehyde group of glutathione. We found that AKR1B1 is highly expressed in gastric cancer and is associated with a poor prognosis of the patients. In vitro experiments found that AKR1B1 has the ability to promote the proliferation and invasion of gastric cancer cells. AKR1B1 inhibited RSL3-induced ferroptosis in gastric cancer by reducing reactive oxygen species accumulation and lipid peroxidation, as well as decreasing intracellular ferrous ion and malondialdehyde expression and increasing glutathione expression. Our study demonstrated that AKR1B1 resisted RSL3-induced ferroptosis by regulating GPX4, PTGS2 and ACSL4, which was further demonstrated in a xenograft nude mouse model. Our work reveals a critical role for the AKR1B1 in the resistance to RSL3-induced ferroptosis in gastric cancer.


Subject(s)
Cell Proliferation , Ferroptosis , NF-E2-Related Factor 2 , Stomach Neoplasms , Ferroptosis/genetics , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Cell Line, Tumor , Animals , Reactive Oxygen Species/metabolism , Mice , Gene Expression Regulation, Neoplastic , Lipid Peroxidation , Aldehyde Reductase/metabolism , Aldehyde Reductase/genetics , Mice, Nude , Male , Carbolines
4.
Int Immunopharmacol ; 140: 112812, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39094360

ABSTRACT

Diabetic nephropathy (DN) is one of the leading clinical causes of end-stage renal failure. The classical aldose reductase (AR) inhibitor epalrestat shows beneficial effect on renal dysfunction induced by DN, with metabolic profile and molecular mechanisms remains to be investigated further. In the current study, integrated untargeted metabolomics, network pharmacology and molecular dynamics approaches were applied to explore the therapeutic mechanisms of epalrestat against DN. Firstly, untargeted serum and urine metabolomics analysis based on UPLC-Q-TOF-MS was performed, revealed that epalrestat could regulate the metabolic disorders of amino acids metabolism, arachidonic acid metabolism, pyrimidine metabolism and citrate cycle metabolism pathways after DN. Subsequently, metabolomics-based network analysis was carried out to predict potential active targets of epalrestat, mainly involving AGE-RAGE signaling pathway, TNF signaling pathway and HIF-1 signaling pathway. Moreover, a 100 ns molecular dynamics approach was employed to validate the interactions between epalrestat and the core targets, showing that epalrestat could form remarkable tight binding with GLUT1 and NFκB than it with AR. Surface-plasmon resonance assay further verified epalrestat could bind GLUT1 and NFκB proteins specifically. Overall, integrated system network analysis not only demonstrated that epalrestat could attenuate DN induced metabolic disorders and renal injuries, but also revealed that it could interact with multi-targets to play a synergistic regulatory role in the treatment of DN.


Subject(s)
Diabetic Nephropathies , Metabolomics , Molecular Dynamics Simulation , Rhodanine , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Animals , Male , Rhodanine/analogs & derivatives , Rhodanine/therapeutic use , Rhodanine/pharmacology , Thiazolidines/pharmacology , Thiazolidines/therapeutic use , Humans , Aldehyde Reductase/metabolism , Aldehyde Reductase/antagonists & inhibitors , Signal Transduction/drug effects , Glucose Transporter Type 1/metabolism , NF-kappa B/metabolism , Network Pharmacology , Rats
5.
Int J Biol Macromol ; 277(Pt 1): 133816, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002911

ABSTRACT

Diabetic vascular complication including diabetic retinopathy is a major morbidity in Saudia Arabia. The polyol pathway aka aldose reductase (AR) pathway has gained significant association with diabetic retinopathy with regard to chronically enhanced glucose metabolism. Considerable research has been put forth to develop more effective therapeutic strategies to overcome the overwhelming challenges of vascular complications associated with diabetes. In this regard, constituents of Cichorium intybus can offer strong AR inhibitory potential because of their strong antidiabetic properties. Therefore, aim of this study was to investigate the AR inhibitory as well as antiglycation potential of C. intybus extract/compounds. The preliminary in vitro results showed that methanolic extract of C. intybus could significantly inhibit AR enzyme and advanced glycation end product formation. Eventually, based on previous studies and reviews, we selected one hundred fifteen C. intybus root constituents and screened them through Lipinski's rule of five and ADMET analysis. Later, after molecular docking analysis of eight compounds, five best were selected for molecular dynamics simulation to deduce their binding affinity with the AR enzyme. Finally, three out of five compounds were further tested in vitro for their AR inhibitory potential and antiglycation properties. Enzyme assay and kinetic studies showed that all the three tested compounds were having potent AR inhibitory properties, although to a lesser extent than ellagic acid and tolrestat. Similarly, kaempferol showed strong antiglycation property equivalent to ellagic acid, but greater than aminoguanidine. Intriguingly, significant reduction in sorbitol accumulation in RBCs by the tested compounds substantiated strong AR inhibition by these compounds. Moreover, decrease in sorbitol accumulation under high glucose environment also signifies the potential application of these compounds in diabetic retinopathy and other vascular complications. Thus, in sum, the in silico and in vitro studies combinedly showed that C. intybus root is a treasure for therapeutic compounds and can be explored further for drug development against diabetic retinopathy.


Subject(s)
Aldehyde Reductase , Cichorium intybus , Diabetic Retinopathy , Enzyme Inhibitors , Plant Extracts , Humans , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism , Cichorium intybus/chemistry , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Glycation End Products, Advanced/metabolism , Glycosylation/drug effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Phytochemicals/pharmacology , Phytochemicals/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry
6.
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
7.
Oncol Res ; 32(8): 1287-1308, 2024.
Article in English | MEDLINE | ID: mdl-39055885

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Antineoplastic Agents , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Aldo-Keto Reductases/metabolism , Aldo-Keto Reductases/genetics , Aldo-Keto Reductases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacology , Precision Medicine , Signal Transduction , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism
8.
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
10.
Bioorg Chem ; 151: 107666, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39067420

ABSTRACT

Design and virtual screening of a set of non-acidic 4-methyl-4-phenyl-benzenesulfonate-based aldose reductase 2 inhibitors had been developed followed by chemical synthesis. Based on the results, the synthesized compounds 2, 4a,b, 7a-c, 9a-c, 10a-c, 11b,c and 14a-c inhibited the ALR2 enzymatic activity in a submicromolar range (99.29-417 nM) and among them, the derivatives 2, 9b, 10a and 14b were able to inhibit ALR2 by IC50 of 160.40, 165.20, 99.29 and 120.6 nM, respectively. Moreover, kinetic analyses using Lineweaver-Burk plot revealed that the most active candidate 10a inhibited ALR2 potently via a non-competitive mechanism. In vivo studies showed that 10 mg/kg of compound 10a significantly lowered blood glucose levels in alloxan-induced diabetic mice by 46.10 %. Moreover, compound 10a showed no toxicity up to a concentration of 50 mg/kg and had no adverse effects on liver and kidney functions. It significantly increased levels of GSH and SOD while decreasing MDA levels, thereby mitigating oxidative stress associated with diabetes and potentially attenuating diabetic complications. Furthermore, the binding mode of compound 10a was confirmed through MD simulation. Noteworthy, compounds 2 and 14b showed moderate antimicrobial activity against the two fungi Aspergillus fumigatus and Aspergillus niger. Finally, we report the thiazole derivative 10a as a new promising non-acidic aldose reductase inhibitor that may be beneficial in treating diabetic complications.


Subject(s)
Aldehyde Reductase , Drug Design , Enzyme Inhibitors , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism , Animals , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Structure-Activity Relationship , Molecular Structure , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/chemically induced , Dose-Response Relationship, Drug , Molecular Docking Simulation , Male , Humans , Benzenesulfonates/pharmacology , Benzenesulfonates/chemistry , Benzenesulfonates/chemical synthesis , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry
11.
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
12.
J Neurotrauma ; 41(17-18): 2186-2198, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39083435

ABSTRACT

Spinal cord contusion injury results in Wallerian degeneration of spinal cord axonal tracts, which are necessary for locomotor function. Axonal swelling and loss of axonal density at the contusion site, characteristic of Wallerian degeneration, commence within hours of injury. Tempol, a superoxide dismutase mimetic, was previously shown to reduce the loss of spinal cord white matter and improve locomotor function in an experimental model of spinal cord contusion, suggesting that tempol treatment might inhibit Wallerian degeneration of spinal cord axons. Here, we report that tempol partially inhibits Wallerian degeneration, resulting in improved locomotor recovery. We previously reported that Wallerian degeneration is reduced by inhibitors of aldose reductase (AR), which converts glucose to sorbitol in the polyol pathway. We observed that tempol inhibited sorbitol production in the injured spinal cord to the same extent as the AR inhibitor, sorbinil. Tempol also prevented post-contusion upregulation of AR (AKR1B10) protein expression within degenerating axons, as previously observed for AR inhibitors. Additionally, we hypothesized that tempol inhibits axonal degeneration by preventing loss of the glutathione pool due to polyol pathway activity. Consistent with our hypothesis, tempol treatment resulted in greater glutathione content in the injured spinal cord, which was correlated with increased expression and activity of gamma glutamyl cysteine ligase (γGCL; EC 6.3.2.2), the rate-limiting enzyme for glutathione synthesis. Administration of the γGCL inhibitor buthionine sulfoximine abolished all observed effects of tempol administration. Together, these results support a pathological role for polyol pathway activation in glutathione depletion, resulting in Wallerian degeneration after spinal cord injury (SCI). Interestingly, methylprednisolone, oxandrolone, and clenbuterol, which are known to spare axonal tracts after SCI, were equally effective in inhibiting polyol pathway activation. These results suggest that prevention of AR activation is a common target of many disparate post-SCI interventions.


Subject(s)
Aldehyde Reductase , Cyclic N-Oxides , Glutathione , Spin Labels , Spinal Cord Injuries , Wallerian Degeneration , Animals , Wallerian Degeneration/metabolism , Wallerian Degeneration/drug therapy , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism , Cyclic N-Oxides/pharmacology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Rats , Glutathione/metabolism , Rats, Sprague-Dawley , Female , Enzyme Activation/drug effects , Neuroprotective Agents/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase/drug effects , Antioxidants/pharmacology , Disease Models, Animal
13.
Bioprocess Biosyst Eng ; 47(10): 1659-1668, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38969832

ABSTRACT

Erythritol is a natural non-caloric sweetener, which is produced by fermentation and extensively applied in food, medicine and chemical industries. The final step of the erythritol synthesis pathway is involved in erythritol reductase, whose activity and NADPH-dependent become the limiting node of erythritol production efficiency. Herein, we implemented a strategy combining molecular docking and thermal stability screening to construct an ER mutant library. And we successfully obtained a double mutant ERK26N/V295M (ER*) whose catalytic activity was 1.48 times that of wild-type ER. Through structural analysis and MD analysis, we found that the catalytic pocket and the enzyme stability of ER* were both improved. We overexpressed ER* in the engineered strain ΔKU70 to obtain the strain YLE-1. YLE-1 can produce 39.47 g/L of erythritol within 144 h, representing a 35% increase compared to the unmodified strain, and a 10% increase compared to the strain overexpressing wild-type ER. Considering the essentiality of NADPH supply, we further co-expressed ER* with two genes from the oxidative phase of PPP, ZWF1 and GND1. This resulted in the construction of YLE-3, which exhibited a significant increase in production, producing 47.85 g/L of erythritol within 144 h, representing a 63.90% increase compared to the original chassis strain. The productivity and the yield of the engineered strain YLE-3 were 0.33 g/L/h and 0.48 g/g glycerol, respectively. This work provided an ER mutation with excellent performance, and also proved the importance of cofactors in the process of erythritol synthesis, which will promote the industrial production of erythritol by metabolic engineering of Y. lipolytica.


Subject(s)
Erythritol , Yarrowia , Erythritol/biosynthesis , Erythritol/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Yarrowia/enzymology , Fungal Proteins/genetics , Fungal Proteins/biosynthesis , Fungal Proteins/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Aldehyde Reductase/biosynthesis , Protein Engineering/methods , Metabolic Engineering/methods , Molecular Docking Simulation
14.
Faraday Discuss ; 252(0): 157-173, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-38836629

ABSTRACT

A scarcity of cofactors, necessary metabolites or substrates for in vivo enzymatic reactions, is among the major barriers for product synthesis in metabolically engineered cells. This work compares our recently developed cofactor-boosting strategy, which uses xylose reductase (XR) and lactose to increase the intracellular levels of reduced or oxidized nicotinamide adenine dinucleotide (phosphate) (NAD(P)H), adenosine triphosphate (ATP) and acetyl coenzymeA (acetyl-CoA), with other previously reported methods. We demonstrated that the XR/lactose approach enhances levels of sugar alcohols and sugar phosphates, which leads to elevated levels of crucial cofactors required by specific metabolic pathways. The patterns of cofactor enhancement are not uniform and depend upon the specific pathway components that are overexpressed. We term this model the "user-pool" model. Here, we investigated metabolite alteration in the fatty-alcohol-producing system in the presence of XR/lactose within an early time frame (5 min after the bioconversion started). All metabolite data were analyzed using untargeted metabolomics. We found that the XR/lactose system could improve fatty-alcohol production as early as 5 min after the bioconversion started. The enhancement of key cofactors and intermediates, such as hexitol, NAD(P)H, ATP, 3-phosphoglycerate, acetyl-CoA, 6-phosphogluconate (6-PG) and glutathione, was consistent with those previously reported on a longer time scale (after 1 h). However, measurements performed at the early time reported here showed detectable differences in metabolite enhancement patterns, such as those of ATP, NADPH, acetyl-CoA and glutathione. These data could serve as a basis for future analysis of metabolic flux alteration by the XR/lactose system. Comparative analysis of the cofactor enhancement by XR and other methods suggests that XR/lactose can serve as a simple tool to increase levels of various cofactors for microbial cell factories.


Subject(s)
Biocatalysis , Aldehyde Reductase/metabolism , NADP/metabolism , NADP/chemistry , Lactose/metabolism , Lactose/chemistry , Coenzymes/metabolism , Coenzymes/chemistry , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/chemistry , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Metabolic Engineering
15.
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
16.
Lipids Health Dis ; 23(1): 201, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937844

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases , Curcumin , Non-alcoholic Fatty Liver Disease , Triglycerides , Curcumin/pharmacology , Curcumin/analogs & derivatives , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Humans , Hep G2 Cells , Aldo-Keto Reductases/metabolism , Rats , Male , Triglycerides/blood , Triglycerides/metabolism , Acetyl-CoA Carboxylase/metabolism , Aldehyde Reductase/metabolism , Aldehyde Reductase/antagonists & inhibitors , Diet, High-Fat/adverse effects , Molecular Docking Simulation , Liver/drug effects , Liver/metabolism , Metformin/pharmacology , Rats, Sprague-Dawley , Disease Models, Animal , Rhodanine/analogs & derivatives , Thiazolidines
17.
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
18.
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
19.
J Biol Chem ; 300(7): 107479, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38879006

ABSTRACT

Glucoselysine (GL) is an unique advanced glycation end-product derived from fructose. The main source of fructose in vivo is the polyol pathway, and an increase in its activity leads to diabetic complications. Here, we aimed to demonstrate that GL can serve as an indicator of the polyol pathway activity. Additionally, we propose a novel approach for detecting GL in peripheral blood samples using liquid chromatography-tandem mass spectrometry and evaluate its clinical usefulness. We successfully circumvent interference from fructoselysine, which shares the same molecular weight as GL, by performing ultrafiltration and hydrolysis without reduction, successfully generating adequate peaks for quantification in serum. Furthermore, using immortalized aldose reductase KO mouse Schwann cells, we demonstrate that GL reflects the downstream activity of the polyol pathway and that GL produced intracellularly is released into the extracellular space. Clinical studies reveal that GL levels in patients with type 2 diabetes are significantly higher than those in healthy participants, while Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)ornithine (MG-H1) levels are significantly lower. Both GL and MG-H1 show higher values among patients with vascular complications; however, GL varies more markedly than MG-H1 as well as hemoglobin A1c, fasting plasma glucose, and estimated glomerular filtration rate. Furthermore, GL remains consistently stable under various existing drug treatments for type 2 diabetes, whereas MG-H1 is impacted. To the best of our knowledge, we provide important insights in predicting diabetic complications caused by enhanced polyol pathway activity via assessment of GL levels in peripheral blood samples from patients.


Subject(s)
Diabetes Mellitus, Type 2 , Glycation End Products, Advanced , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Humans , Animals , Glycation End Products, Advanced/metabolism , Mice , Male , Middle Aged , Female , Lysine/metabolism , Ornithine/metabolism , Ornithine/blood , Ornithine/analogs & derivatives , Aldehyde Reductase/metabolism , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/blood , Polymers/chemistry , Aged , Mice, Knockout , Imidazoles
SELECTION OF CITATIONS
SEARCH DETAIL