Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Microb Cell Fact ; 23(1): 134, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724934

ABSTRACT

BACKGROUND: Lovastatin has widespread applications thanks to its multiple pharmacological effects. Fermentation by filamentous fungi represents the major way of lovastatin production. However, the current lovastatin productivity by fungal fermentation is limited and needs to be improved. RESULTS: In this study, the lovastatin-producing strains of Aspergillus terreus from marine environment were screened, and their lovastatin productions were further improved by genetic engineering. Five strains of A. terreus were isolated from various marine environments. Their secondary metabolites were profiled by metabolomics analysis using Ultra Performance Liquid Chromatography-Mass spectrometry (UPLC-MS) with Global Natural Products Social Molecular Networking (GNPS), revealing that the production of secondary metabolites was variable among different strains. Remarkably, the strain of A. terreus MJ106 could principally biosynthesize the target drug lovastatin, which was confirmed by High Performance Liquid Chromatography (HPLC) and gene expression analysis. By one-factor experiment, lactose was found to be the best carbon source for A. terreus MJ106 to produce lovastatin. To improve the lovastatin titer in A. terreus MJ106, genetic engineering was applied to this strain. Firstly, a series of strong promoters was identified by transcriptomic and green fluorescent protein reporter analysis. Then, three selected strong promoters were used to overexpress the transcription factor gene lovE encoding the major transactivator for lov gene cluster expression. The results revealed that compared to A. terreus MJ106, all lovE over-expression mutants exhibited significantly more production of lovastatin and higher gene expression. One of them, LovE-b19, showed the highest lovastatin productivity at a titer of 1512 mg/L, which represents the highest production level reported in A. terreus. CONCLUSION: Our data suggested that combination of strain screen and genetic engineering represents a powerful tool for improving the productivity of fungal secondary metabolites, which could be adopted for large-scale production of lovastatin in marine-derived A. terreus.


Subject(s)
Aspergillus , Fermentation , Genetic Engineering , Lovastatin , Lovastatin/biosynthesis , Lovastatin/metabolism , Aspergillus/metabolism , Aspergillus/genetics , Aquatic Organisms/metabolism , Aquatic Organisms/genetics
2.
Toxicol In Vitro ; 97: 105806, 2024 May.
Article in English | MEDLINE | ID: mdl-38432573

ABSTRACT

INTRODUCTION: Statins have demonstrated chondroprotective effects by reducing inflammation and mitigating extracellular matrix degradation. However, statins are also reported to be cytotoxic to several types of cells. Early-onset osteoarthritis (OA) is characterized by synovial inflammation, which adversely affects hyaluronan (HA) production in fibroblast-like synoviocytes (FLSs). Nevertheless, the precise effects of statins on the synovium remain unclear. METHODS: This study investigated the impact of lovastatin on human FLSs, and HA secretion-related genes, signaling pathways, and production were evaluated. RESULTS: The findings revealed that high doses of lovastatin (20 or 40 µM) decreased FLS viability and increased cell death. FLS proliferation ceased when cultured in a medium containing 5 or 10 µM lovastatin. mRNA expression analysis demonstrated that lovastatin (5 and 10 µM) upregulated the gene level of hyaluronan synthase 1 (HAS1), HAS2, and proteoglycan 4 (PRG4), but not HAS3. While the expression of multidrug resistance-associated protein 5 transporter gene remained unaffected, both inward-rectifying potassium channel and acid-sensing ion channel 3 were upregulated. Western blot further confirmed that lovastatin increased the production of HAS1 and PRG4, and activated the PKC-α, ERK1/2, and p38-MAPK signaling pathways. Additionally, lovastatin elevated intracellular cAMP levels and HA production in FLSs. CONCLUSION: Lovastatin impairs cellular proliferation but enhances HA production in human FLSs.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Synoviocytes , Humans , Synoviocytes/metabolism , Hyaluronic Acid/metabolism , Lovastatin/pharmacology , Lovastatin/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Fibroblasts/metabolism , Cell Proliferation , Inflammation/metabolism , Cells, Cultured
3.
Appl Microbiol Biotechnol ; 108(1): 154, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38240803

ABSTRACT

Monascus pilosus has been used to produce lipid-lowering drugs rich in monacolin K (MK) for a long period. Genome mining reveals there are still many potential genes worth to be explored in this fungus. Thereby, efficient genetic manipulation tools will greatly accelerate this progress. In this study, we firstly developed the protocol to prepare protoplasts for recipient of CRISPR/Cas9 system. Subsequently, the vector and donor DNA were co-transformed into recipients (106 protoplasts/mL) to produce 60-80 transformants for one test. Three genes (mpclr4, mpdot1, and mplig4) related to DNA damage response (DDR) were selected to compare the gene replacement frequencies (GRFs) of Agrobacterium tumefaciens-mediated transformation (ATMT) and CRISPR/Cas9 gene editing system (CGES) in M. pilosus MS-1. The results revealed that GRF of CGES was approximately five times greater than that of ATMT, suggesting that CGES was superior to ATMT as a targeting gene editing tool in M. pilosus MS-1. The inactivation of mpclr4 promoted DDR via the non-homologous end-joining (NHEJ) and increased the tolerances to DNA damaging agents. The inactivation of mpdot1 blocked DDR and led to the reduced tolerances to DNA damaging agents. The inactivation of mplig4 mainly blocked the NHEJ pathway and led to obviously reduced tolerances to DNA damaging agents. The submerged fermentation showed that the ability to produce MK in strain Δmpclr4 was improved by 52.6% compared to the wild type. This study provides an idea for more effective exploration of gene functions in Monascus strains. KEY POINTS: • A protocol of high-quality protoplasts for CGES has been developed in M. pilosus. • The GRF of CGES was about five times that of ATMT in M. pilosus. • The yield of MK for Δmpclr4 was enhanced by 52.6% compared with the wild type.


Subject(s)
Gene Editing , Monascus , Monascus/genetics , Monascus/metabolism , CRISPR-Cas Systems , Gene Targeting/methods , Lovastatin/metabolism , Agrobacterium tumefaciens/genetics , DNA/metabolism
4.
Stem Cell Reports ; 18(4): 969-984, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37044069

ABSTRACT

The spermatogonial stem cell (SSC) niche is critical for SSC maintenance and subsequent spermatogenesis. Numerous reproductive hazards impair the SSC niche, thereby resulting in aberrant SSC self-renewal and male infertility. However, promising agents targeting the impaired SSC niche to promote SSC self-renewal are still limited. Here, we screen out and assess the effects of Lovastatin on the self-renewal of mouse SSCs (mSSCs). Mechanistically, Lovastatin promotes the self-renewal of mSSCs and inhibits its inflammation and apoptosis through the regulation of isoprenoid intermediates. Remarkably, treatment by Lovastatin could promote the proliferation of undifferentiated spermatogonia in the male gonadotoxicity model generated by busulfan injection. Of note, we demonstrate that Lovastatin could enhance the proliferation of primate undifferentiated spermatogonia. Collectively, our findings uncover that lovastatin could promote the self-renewal of both murine and primate SSCs and have implications for the treatment of certain types of male infertility using small compounds.


Subject(s)
Infertility, Male , Lovastatin , Mice , Animals , Male , Humans , Lovastatin/pharmacology , Lovastatin/metabolism , Stem Cells/metabolism , Cell Proliferation , Spermatogonia/metabolism , Spermatogenesis , Primates , Infertility, Male/chemically induced , Infertility, Male/metabolism
5.
Yeast ; 40(1): 42-52, 2023 01.
Article in English | MEDLINE | ID: mdl-36514193

ABSTRACT

When Monascus purpureus was co-cultured with Saccharomyces cerevisiae, we noted significant changes in the secondary metabolism and morphological development of Monascus. In yeast co-culture, although the pH was not different from that of a control, the Monascus mycelial biomass increased during fermentation, and the Monacolin K yield was significantly enhanced (up to 58.87% higher). However, pigment production did not increase. Co-culture with S. cerevisiae significantly increased the expression levels of genes related to Monacolin K production (mokA-mokI), especially mokE, mokF, and mokG. Linoleic acid, that has been implicated in playing a regulating role in the secondary metabolism and morphology of Monascus, was hypothesized to be the effector. Linoleic acid was detected in the co-culture, and its levels changed during fermentation. Addition of linoleic acid increased Monacolin K production and caused similar morphological changes in Monascus spores and mycelia. Exogenous linoleic acid also significantly upregulated the transcription levels of all nine genes involved in the biosynthesis of Monacolin K (up to 69.50% higher), consistent with the enhanced Monacolin K yield. Taken together, our results showed the effect of S. cerevisiae co-culture on M. purpureus and suggested linoleic acid as a specific quorum-sensing molecule in Saccharomyces-Monascus co-culture.


Subject(s)
Linoleic Acid , Monascus , Linoleic Acid/metabolism , Linoleic Acid/pharmacology , Monascus/genetics , Monascus/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Coculture Techniques , Fermentation , Lovastatin/metabolism , Lovastatin/pharmacology
6.
J Biomol Struct Dyn ; 41(6): 2431-2447, 2023 04.
Article in English | MEDLINE | ID: mdl-35098894

ABSTRACT

Microbial biofilms have been recognized for a vital role in antibiotic resistance and chronic microbial infections for 2-3 decades; still, there are no 'anti-biofilm drugs' available for human applications. There is an urgent need to develop novel 'anti-biofilms' therapeutics to manage biofilm-associated infectious diseases. Several reports have suggested that targeting molecules involved in quorum sensing or biofilm-specific transcription may inhibit biofilm formation. However, the possibility of targeting other vital components of microbial biofilms, especially the extracellular matrix (ECM) components, has remained largely unexplored. Here we report targeting TasA(28-261), the major proteinaceous component of Bacillus subtilis ECM with two small molecule inhibitors (lovastatin and simvastatin) identified through virtual screening and drug repurposing, resulted in complete inhibition of biofilm. In molecular docking and dynamics simulation studies, lovastatin was observed to make stable interactions with TasA(28-261), whereas the simvastatin - TasA(28-261) interactions were relatively less stable. However, in subsequent in vitro studies, both lovastatin and simvastatin successfully inhibited B. subtilis biofilm formation at MIC values of < 10 µg/ml. Besides, these potential inhibitors also caused the disintegration of pre-formed biofilms. Results presented here provide 'proof of concept' for the hypothesis that targeting the extracellular matrix's vital component(s) could be one of the most efficient approaches for inhibiting microbial biofilms and disintegrating the pre-formed biofilms. We propose that a similar approach targeting ECM-associated proteins with FDA-approved drugs could be implemented to develop novel anti-biofilm therapeutic strategies against biofilm-forming chronic microbial pathogens.Communicated by Ramaswamy H. Sarma.


Subject(s)
Bacillus subtilis , Biofilms , Humans , Bacillus subtilis/physiology , Molecular Docking Simulation , Lovastatin/metabolism , Simvastatin , Bacterial Proteins/metabolism
7.
J Appl Microbiol ; 133(6): 3585-3595, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36000236

ABSTRACT

AIMS: Lovastatin has been indicated to impair growth and development of Phytophthora sojae. Therefore, this study was performed to understand the inhibitory mechanism of lovastatin and investigate the metabolic pathway potentially served as a new control target for this plant pathogen. METHODS AND RESULTS: Whole transcriptome analysis of lovastatin-treated P. sojae was performed by RNA-sequencing. The results revealed that 84 genes were upregulated and 58 were downregulated with more than fourfold changes under treatment. Kyoto Encyclopaedia of Genes and Genomes analysis indicated that the branched-chain amino acids (BCAAs) biosynthesis pathway was abundantly enriched. All enzymes in the BCAAs biosynthesis pathway were identified in the P. sojae genome. Moreover, the study found that the herbicide flumetsulam targeting acetohydroxyacid synthase (AHAS) of the BCAAs biosynthesis pathway could effectively inhibit mycelial growth of P. sojae. CONCLUSIONS: Lovastatin treatment significantly influences the BCAAs biosynthesis pathway in P. sojae. Moreover, the herbicide flumetsulam targets AHAS and inhibits growth of P. sojae. SIGNIFICANCE AND IMPACT OF THE STUDY: The present study revealed that BCAAs biosynthesis pathway was influenced by lovastatin treatment and its key enzyme AHAS was identified as a potential new control target, which provides clues for exploring more oomycetes to control plant diseases caused by P. sojae.


Subject(s)
Herbicides , Phytophthora , Phytophthora/genetics , Transcriptome , Amino Acids, Branched-Chain/metabolism , Lovastatin/pharmacology , Lovastatin/metabolism , Plant Diseases/prevention & control , Herbicides/pharmacology , Glycine max/metabolism
8.
J Chem Neuroanat ; 125: 102148, 2022 11.
Article in English | MEDLINE | ID: mdl-36031087

ABSTRACT

BACKGROUND: Lovastatin, as a drug of statins subgroup, has been conceptualized to have anti-inflammatory, antioxidant, and anti-apoptotic properties. Accordingly, the present study aimed to investigate the neuroprotective ramification of lovastatin on spinal cord injury (SCI). MATERIAL AND METHODS: Seventy-five female adult Wistar rats were divided into five groups (n = 15). In addition to non-treated (Control group) and laminectomy alone (Sham group), SCI animals were randomly assigned to non-treated spinal cord injury (SCI group), treated with 2 mg/kg of lovastatin (Lova 2 group), and treated with 5 mg/kg of lovastatin (Lova 5 group). At the end of the study, to evaluate the treatments, MDA, CAT, SOD, and GSH factors were evaluated biochemically, apoptosis and gliosis were assessed by immunohistochemical while measuring caspase-3 and GFAP antibodies, and inflammation was estimated by examining the expression of IL-10, TNF-α, and IL-1ß genes. The stereological method was used to appraise the total volume of the spinal cord at the site of injury, the volume of the central cavity created, and the density of neurons and glial cells in the traumatic area. In addition, Basso-Beattie-Bresnehan (BBB) and narrow beam test (NBT) were utilized to rate neurological functions. RESULTS: Our results exposed the fact that biochemical factors (except MDA), stereological parameters, and neurological functions were significantly ameliorated in both lovastatin-treated groups, especially in Lova 5 ones, compared to the SCI group. The expression of the IL-10 gene was significantly upregulated in both lovastatin-treated groups compared to the SCI group and was considerably heighten in Lova 5 group. Expression of TNF-α and IL-1ß, as well as the rate of apoptosis and GFAP positive cells significantly decreased in both lovastatin treated groups compared to the SCI group, and it was more pronounced in the Lova 5 ones. CONCLUSION: Overall, using lovastatin, especially at a dose of 5 mg/kg, has a dramatic neuroprotective impact on SCI treatment.


Subject(s)
Neuroprotective Agents , Spinal Cord Injuries , Animals , Female , Rats , Apoptosis , Disease Models, Animal , Interleukin-10/metabolism , Lovastatin/pharmacology , Lovastatin/therapeutic use , Lovastatin/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats, Wistar , Spinal Cord/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Acta Pharmacol Sin ; 43(11): 2905-2916, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35459869

ABSTRACT

Anterior gradient 2 (AGR2), a protein disulfide isomerase (PDI), is a multifunctional protein under physiological and pathological conditions. In this study we investigated the roles of AGR2 in regulating cholesterol biogenesis, lipid-lowering efficiency of lovastatin as well as in protection against hypercholesterolemia/statin-induced liver injury. We showed that AGR2 knockout significantly decreased hepatic and serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in mice with whole-body or hepatocyte-specific Agr2-null mutant, compared with the levels in their wild-type littermates fed a normal chow diet (NCD) or high-fat diet (HFD). In contrast, mice with AGR2 overexpression (Agr2/Tg) exhibited an increased cholesterol level. Mechanistic studies revealed that AGR2 affected cholesterol biogenesis via activation of AKT/sterol regulatory element-binding protein-2 (SREBP2), to some extent, in a PDI motif-dependent manner. Moreover, elevated AGR2 led to a significant decrease in the lipid-lowering efficacy of lovastatin (10 mg· kg-1· d-1, ip, for 2 weeks) in mice with hypercholesterolemia (hyperCho), which was validated by results obtained from clinical samples in statin-treated patients. We showed that lovastatin had limited effect on AGR2 expression, but AGR2 was inducible in Agr2/Tg mice fed a HFD. Further investigations demonstrated that drug-induced liver toxicity and inflammatory reactions were alleviated in hypercholesterolemic Agr2/Tg mice, suggesting the dual functions of AGR2 in lipid management and hyperCho/statin-induced liver injury. Importantly, the AGR2-reduced lipid-lowering efficacy of lovastatin was attenuated, at least partially, by co-administration of a sulfhydryl-reactive compound allicin (20 mg· kg-1· d-1, ip, for 2 weeks). These results demonstrate a novel role of AGR2 in cholesterol metabolism, drug resistance and liver protection, suggesting AGR2 as a potential predictor for selection of lipid-lowering drugs in clinic.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Mice , Animals , Lovastatin/pharmacology , Lovastatin/therapeutic use , Lovastatin/metabolism , Hypercholesterolemia/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Cholesterol, LDL , Liver/metabolism
10.
J Biosci Bioeng ; 133(4): 362-368, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35105506

ABSTRACT

Monascus purpureus have been used for making koji and other fermented foods and supplements. M. purpureus characteristically produces monacolin K (MK), a secondary metabolite that competitively inhibits cholesterol synthesis. Synchrotron light irradiation was applied to induce mutation in the strain KUPM5 to improve the MK-producing ability of M. purpureus strain KUPM5. Screening by a bioassay utilizing sensitivities to yeast Saccharomyces cerevisiae from 936 colonies allows isolating three mutant strains: SC01, SC02, and SC03. These mutant strains and the parental strain KUPM5 were subjected to make koji using rice, and their metabolites were compared. All strains SC01, SC02, and SC03 in koji showed higher production of MK than the strain KUPM5. Particularly, the SC02 strain produced MK threefold higher than KUPM5 and maintained the production capabilities of other metabolites, including red, yellow, and orange pigments, mycelial contents, and α-amylase activity comparable to those of the strain KUPM5. Comparative genome analysis among strain KUPM5 and the mutants revealed that synchrotron light irradiation introduced mutations in approximately 90% of the total genes, including SNV, MNV, and indel mutations. The frequencies of SNV substitution in the whole genome occupied 68.96% of all mutations, of which 92.38% were transversions and 7.62% were transitions. This study, therefore, proved the synchrotron light irradiation was highly efficient for the strain improvement of a filamentous fungus, M. purpureus, and provided insights into the properties of mutation in the fungus by this mutagen.


Subject(s)
Fermented Foods , Monascus , Fermentation , Lovastatin/metabolism , Monascus/metabolism , Pigments, Biological , Synchrotrons
11.
Hepatology ; 76(5): 1466-1481, 2022 11.
Article in English | MEDLINE | ID: mdl-35102596

ABSTRACT

BACKGROUND AND AIMS: NASH is associated with high levels of cholesterol and triglyceride (TG) in the liver; however, there is still no approved pharmacological therapy. Synthesis of cholesterol and TG is controlled by sterol regulatory element-binding protein (SREBP), which is found to be abnormally activated in NASH patients. We aim to discover small molecules for treating NASH by inhibiting the SREBP pathway. APPROACH AND RESULTS: Here, we identify a potent SREBP inhibitor, 25-hydroxylanosterol (25-HL). 25-HL binds to insulin-induced gene (INSIG) proteins, stimulates the interaction between INSIG and SCAP, and retains them in the endoplasmic reticulum, thereby suppressing SREBP activation and inhibiting lipogenesis. In NASH mouse models, 25-HL lowers levels of cholesterol and TG in serum and the liver, enhances energy expenditure to prevent obesity, and improves insulin sensitivity. 25-HL dramatically ameliorates hepatic steatosis, inflammation, ballooning, and fibrosis through down-regulating the expression of lipogenic genes. Furthermore, 25-HL exhibits both prophylactic and therapeutic efficacies of alleviating NASH and atherosclerosis in amylin liver NASH model diet-treated Ldlr-/- mice, and reduces the formation of cholesterol crystals and associated crown-like structures of Kupffer cells. Notably, 25-HL lowers lipid contents in serum and the liver to a greater extent than lovastatin or obeticholic acid. 25-HL shows a good safety and pharmacokinetics profile. CONCLUSIONS: This study provides the proof of concept that inhibiting SREBP activation by targeting INSIG to lower lipids could be a promising strategy for treating NASH. It suggests the translational potential of 25-HL in human NASH and demonstrates the critical role of SREBP-controlled lipogenesis in the progression of NASH by pharmacological inhibition.


Subject(s)
Insulins , Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Lipogenesis/physiology , Sterol Regulatory Element Binding Proteins , Sterol Regulatory Element Binding Protein 1/metabolism , Islet Amyloid Polypeptide/metabolism , Liver/metabolism , Triglycerides/metabolism , Cholesterol/metabolism , Lovastatin/metabolism , Insulins/metabolism , Mice, Inbred C57BL
12.
Mol Biotechnol ; 64(1): 90-99, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34546548

ABSTRACT

Lovastatin is an anti-cholesterol medicine that is commonly prescribed to manage cholesterol levels, and minimise the risk of suffering from heart-related diseases. Aspergillus terreus (ATCC 20542) supplied with carbohydrates or sugar alcohols can produce lovastatin. The present work explored the application of metabolic engineering in A. terreus to re-route the precursor flow towards the lovastatin biosynthetic pathway by simultaneously overexpressing the gene for acetyl-CoA carboxylase (acc) to increase the precursor flux, and eliminate ( +)-geodin biosynthesis (a competing secondary metabolite) by removing the gene for emodin anthrone polyketide synthase (gedC). Alterations to metabolic flux in the double mutant (gedCΔ*accox) strain and the effects of using two different substrate formulations were examined. The gedCΔ*accox strain, when cultivated with a mixture of glycerol and lactose, significantly (p < 0.05) increased the levels of metabolic precursors malonyl-CoA (48%) and acetyl-CoA (420%), completely inhibited the (+)-geodin biosynthesis, and increased the level of lovastatin [152 mg/L; 143% higher than the wild-type (WT) strain]. The present work demonstrated how the manipulation of A. terreus metabolic pathways could increase the efficiency of carbon flux towards lovastatin, thus elevating its overall production and enabling the use of glycerol as a substrate source. As such, the present work also provides a framework model for other medically or industrially important fungi to synthesise valuable compounds using sustainable carbon sources.


Subject(s)
Aspergillus/metabolism , Lovastatin/metabolism , Metabolic Engineering , Acetyl Coenzyme A/metabolism , Aspergillus/genetics , Benzofurans/metabolism , Biosynthetic Pathways , Fermentation , Glycerol/metabolism , Kinetics , Lactose/metabolism , Malonyl Coenzyme A/metabolism
13.
Planta Med ; 88(13): 1190-1198, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34875697

ABSTRACT

Phloridzin is a lead compound of the prestigious antidiabetic gliflozins. The present study found that phloridzin highly accumulated in Malus rockii Rehder. The content of phloridzin in M. rockii was the highest among wild plants, with the percentage of 15.54% in the dry leaves. The structure of phloridzin was revised by proton exchange experiments and extensive 2D NMR spectra. Phloridzin exhibited significant hypolipidemic activity in golden Syrian hamsters maybe by increasing the expression of CYP7A1, at the doses of 50 mg/kg and 200 mg/kg. The total performance of anti-hyperlipidemic effect of phloridzin may be superior to that of lovastatin, though lovastatin was more active than phloridzin. In addition, phloridzin exhibited moderate antimalarial activity with inhibition ratio of 31.3 ± 10.9% at a dose of 25 mg/kg/day, and showed moderate analgesic activity with 28.0% inhibition at a dose of 50 mg/kg.


Subject(s)
Antimalarials , Malus , Sodium-Glucose Transporter 2 Inhibitors , Phlorhizin/pharmacology , Phlorhizin/chemistry , Malus/chemistry , Sodium-Glucose Transporter 2 Inhibitors/metabolism , Protons , Lovastatin/metabolism
14.
Neurochem Int ; 148: 105078, 2021 09.
Article in English | MEDLINE | ID: mdl-34048842

ABSTRACT

As a general anesthetic widely used in surgical, sevoflurane has been shown to cause cognitive and memory deficits in the elderly. It's important to find out agents that can counteract sevoflurane-induced cognitive dysfunction. This study is aimed to investigate the effect of lovastatin on sevoflurane-induced cognitive impairment in aged rats and reveal the potential mechanisms. BV-2 cells, rat hippocampal neurons or male aged rats were exposed to 2% sevoflurane for 5 h. The cells were pretreated with 10 µM lovastatin. The rats were intraperitoneally injected with 5 mg/kg/day lovastatin for three days. The results showed that lovastatin enhanced exosomal IDE secretion from sevoflurane-exposed BV-2 cells and promoted Aß degradation. Lovastatin treatment also inhibited the increased expressions of ß-secretase 1 (BACE1) and γ-secretase in hippocampal neurons under sevoflurane exposure in vitro. In animal experiments, the discrimination index in novel object recognition test and percentage of spontaneous alternation in Y-maze test were significantly elevated after lovastatin administration. In addition, Aß plaque area and contents of soluble Aß1-40 and Aß1-42 in the hippocampal tissues were decreased upon lovastatin treatment. Furthermore, lovastatin reversed sevoflurane-induced Aß accumulation via up-regulating IDE expression, and down-regulating amyloid precursor protein (APP)-related protein expression (ß-C-terminal fragment (CTF), BACE1 and γ-secretase). In conclusion, lovastatin alleviates sevoflurane-induced cognitive deficient in aged rats via promoting Aß degradation and reducing Aß production. Lovastatin may be beneficial in preventing anesthetic-induced cognitive impairment.


Subject(s)
Aging/physiology , Cognitive Dysfunction/metabolism , Lovastatin/pharmacology , Plaque, Amyloid/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition/drug effects , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Hippocampus/metabolism , Lovastatin/metabolism , Mice , Rats, Sprague-Dawley , Sevoflurane/metabolism , Sevoflurane/pharmacology
15.
J Sci Food Agric ; 101(5): 1782-1791, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-32892346

ABSTRACT

BACKGROUND: Ginkgo biloba seeds are well known for the significant curative effects on relieving cough and asthma. However, the development of products from ginkgo seeds still falls behind at present, resulting in a great waste of ginkgo seeds' resource. In this work, submerged fermentation of ginkgo seed powder using Eurotium cristatum was studied to investigate its feasibility as a new processing method. RESULTS: To promote the growth of E. cristatum, the optimum fermentation medium was 80.0 g L-1 of ginkgo seed powder with addition of 5.0 g L-1 calcium chloride (CaCl2 ), 4.0 g L-1 magnesium sulfate (MgSO4 ), 1.25 g L-1 zinc sulfate (ZnSO4 ) and 0.65 g L-1 iron(II) sulfate (FeSO4 ). The optimum fermentation conditions were pH 5.8 ± 0.1, inoculum size 5.1 × 106 CFU mL-1 , liquid medium volume 100 mL in 250-mL Erlenmeyer flask and fermentation 4 days. Through fermentation, the production of lovastatin in fermentation broth could reach up to 32.97 ± 0.17 µg mL-1 and the total antioxidant capacity was improved by more than two-fold. In addition, 40.15% of the ginkgotoxin in ginkgo seed powder was degraded while the entire degradation of ginkgolic acids was obtained. Moreover, fermented ginkgo seed powder suspension presented pleasant fragrances, and the activities of amylase and protease were enhanced to 11.30 ± 0.10 U mL-1 and 23.01 ± 0.20 U mL-1 , respectively. CONCLUSIONS: Submerged fermentation using E. cristatum could significantly enhance the functional value and safety of ginkgo seed powder, and had great potential to become a novel processing method for the development of ginkgo seeds fermented products. © 2020 Society of Chemical Industry.


Subject(s)
Eurotium/metabolism , Fermented Foods/microbiology , Ginkgo biloba/microbiology , Antioxidants/analysis , Antioxidants/metabolism , Fermentation , Fermented Foods/analysis , Food Microbiology , Ginkgo biloba/chemistry , Ginkgo biloba/metabolism , Lovastatin/analysis , Lovastatin/metabolism , Powders/chemistry , Pyridoxine/analogs & derivatives , Pyridoxine/analysis , Pyridoxine/metabolism , Salicylates/analysis , Salicylates/metabolism , Seeds/chemistry , Seeds/metabolism , Seeds/microbiology
16.
Eur J Pharm Biopharm ; 154: 338-347, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32739535

ABSTRACT

A novel liposome-micelle-hybrid (LMH) carrier system was developed as a superior oral drug delivery platform compared to conventional liposome or micelle formulations. The optimal LMH system was engineered by encapsulating TPGS micelles in the aqueous core of liposomes and its efficacy for oral delivery was demonstrated using lovastatin (LOV) as a model poorly soluble drug with P-gp (permeability glycoprotein) limited intestinal absorption. LOV-LMH was characterised as unilamellar, spherical vesicles encapsulating micellar structures within the interior aqueous core and showing an average diameter below 200 nm. LMH demonstrated enhanced drug loading, water apparent solubility and extended/controlled release of LOV compared to conventional liposomes and micelles. LMH exhibited enhanced LOV absorption and transportation in a Caco-2 cell monolayer model of the intestine by inhibiting the P-gp transporter system compared to free LOV. The LMH system is a promising novel oral delivery approach for enhancing bioavailability of poorly water-soluble drugs, especially those presenting P-gp effluxes limited absorption.


Subject(s)
Drug Delivery Systems/methods , Intestinal Absorption/physiology , Lovastatin/administration & dosage , Lovastatin/metabolism , Micelles , Water/metabolism , Administration, Oral , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Intestinal Absorption/drug effects , Liposomes , Solubility/drug effects
17.
J Appl Microbiol ; 129(5): 1410-1418, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32357272

ABSTRACT

AIMS: Red yeast rice (RYR), produced by inoculating Monascus strains to steamed rice, contains many kinds of physiologically bioactive compounds, among which monacolin K can be used as an antihypercholesterolaemic agent. However, RYR can be polluted by the mycotoxin citrinin, which has nephrotoxic and hepatotoxic activities. To avoid the risk of citrinin contamination in Monascus fermented products, it is important to screen for Monascus strains that produce no or low citrinin. METHODS AND RESULTS: Five autochthonous Monascus strains with high-yield monacolin K and undetectable citrinin were obtained using high-performance liquid chromatography (HPLC). All five strains were identified as Monascus ruber based on Genealogical Concordance Phylogenetic Species Recognition criteria. Polymerase chain reaction revealed that citrinin polyketide synthase (pksCT) gene was found in these strains, but transcriptional regulator (ctnA) was not found. CONCLUSIONS: Five strains are potential strains for producing high-quality RYR. The distribution of the pksCT gene was not restricted to Monascus purpureus and Monascus sanguineus, and M. ruber strains were diverse in pksCT and ctnA genes. SIGNIFICANCE AND IMPACT OF THE STUDY: The integration of citrinin HPLC analysis and pksCT and ctnA genes amplification could provide a complementary approach in valuable Monascus strains screening.


Subject(s)
Citrinin/metabolism , Lovastatin/metabolism , Monascus/isolation & purification , Monascus/metabolism , Chromatography, High Pressure Liquid , Fermentation , Industrial Microbiology , Monascus/classification , Monascus/genetics , Phylogeny , Polyketide Synthases/genetics , Polymerase Chain Reaction , Trans-Activators/genetics
18.
J Invertebr Pathol ; 171: 107334, 2020 03.
Article in English | MEDLINE | ID: mdl-32006551

ABSTRACT

FK506 binding proteins (FKBPs) participate in regulation of diverse biological processes. However, the role of these proteins in insect-pathogenic fungi is far from well understood. To investigate the functions of FKBPs in Beauveria bassiana, a widely used entomopathogenic fungus for control of insect pests, we identify three putative FKBP genes, Bbfkbp12, Bbfkbp15, and Bbfkbp50, in the fungus. Gene-disruption experiments show that loss of Bbfkbp12 results in a significant increase of resistance of B. bassiana against the immunosuppressive compounds FK506 and rapamycin, while loss of Bbfkbp50 leads to the resistance to the ergosterol synthesis inhibitor lovastatin. Transcription assays of calcineurin (CaN)- and mTOR (mammalian target of rapamycin)-downstream target genes confirm that BbFKBP12 is the target of both FK506 and rapamycin, associated with CaN- and mTOR-signal pathways in B. bassiana. GFP-tagging of the proteins shows that BbFKBP12 and BbFKBP15 localize in cytoplasm while BbFKBP50 in nucleus. Our results provide useful information for the study of functions of CaN- and mTOR-mediated signaling, and ergosterol synthesis in the entomopathogenic fungi.


Subject(s)
Beauveria/growth & development , Ergosterol/biosynthesis , Fungal Proteins/genetics , Signal Transduction , Tacrolimus Binding Proteins/genetics , Antifungal Agents/metabolism , Beauveria/metabolism , Fungal Proteins/metabolism , Lovastatin/metabolism , Sirolimus/metabolism , Tacrolimus/metabolism , Tacrolimus Binding Proteins/metabolism
19.
Clin Chem Lab Med ; 58(5): 664-672, 2020 04 28.
Article in English | MEDLINE | ID: mdl-31665111

ABSTRACT

Background Statins are used to treat and prevent cardiovascular diseases (CVDs) by reducing the total serum cholesterol concentration. Unfortunately, dose-related side effects and sub-optimal response, attributed to non-adherence amongst others, were described. Therefore, a fast and sensitive liquid chromatography-high-resolution tandem mass spectrometry (LC-HRMS/MS) method for adherence testing and therapeutic drug monitoring of all currently marketed statins and their active metabolites in human blood plasma should be developed, validated and tested for applicability. Methods Atorvastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin, as well as ortho- and para-hydroxy-atorvastatin, lovastatin hydroxy acid and simvastatin hydroxy acid were included and several internal standards (IS) tested. Validation was performed according to the guideline of the European Medicines Agency including selectivity, carry-over, accuracy, precision, matrix effects, dilution integrity and analyte stability. Finally, applicability was tested using 14 patient samples submitted for regular toxicological analysis. Results Due to an analytical interference of atorvastatin-d5, diazepam-d5 and pentobarbital-d5 were chosen as IS for positive and negative ionization mode, respectively. All statins and metabolites fulfilled the validation acceptance criteria except for fluvastatin, which could not be quantified reliably and reproducibly, most probably due to instability. Analyses of human plasma samples revealed concentrations of statins and metabolites below the reference plasma concentrations in the case of eight patients. However, nothing was known concerning patients' adherence and time between intake and sampling. Conclusions An LC-HRMS/MS method for identification and quantification of atorvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin and four active metabolites was successfully developed and applicability demonstrated.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Medication Adherence , Tandem Mass Spectrometry/methods , Atorvastatin/blood , Atorvastatin/metabolism , Atorvastatin/standards , Atorvastatin/therapeutic use , Cardiovascular Diseases/drug therapy , Chromatography, High Pressure Liquid/standards , Guidelines as Topic , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/standards , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Limit of Detection , Lovastatin/blood , Lovastatin/metabolism , Lovastatin/standards , Lovastatin/therapeutic use , Pravastatin/blood , Pravastatin/standards , Pravastatin/therapeutic use , Quality Control , Reference Standards , Tandem Mass Spectrometry/standards
20.
Xenobiotica ; 50(5): 593-601, 2020 May.
Article in English | MEDLINE | ID: mdl-31505985

ABSTRACT

Lovastatin shows low bioavailability (lower than 5%) after oral administration because of the poor aqueous solubility and widely metabolized by CYP3A4.Lovastatin solid dispersion was designed to enhance the dissolution. The in vitro intestinal absorption study indicated an increase in the apparent permeability of different intestinal segments compared with crude lovastatin. In the range of 12.5-50 µg/ml, the absorption of both lovastatin and lovastatin solid dispersion were found to be a passive process in rat's jejunum and ileum, but not endocytosis process. CYP3A4 inhibitor (ketoconazole) significantly increased the intestinal absorption of lovastatin and lovastatin solid dispersion. However, P-glycoprotein efflux inhibitor (verapamil) had little effect on them.The absolute bioavailability of lovastatin and lovastatin acid after oral administration of lovastatin solid dispersion were increased by about 2.01-fold and 1.40-fold than that of lovastatin suspension. The oral bioavailability of lovastatin and lovastatin acid after oral administration of lovastatin solid dispersion with 10 mg/kg kaempferol (CYP3A4 inhibitor) were increased about 3.79-fold and 2.51-fold than that of lovastatin suspension, and the absolute bioavailability of lovastatin was up to 33.0%.As a result, co-administration of lovastatin solid dispersion with kaempferol could be a promising delivery system to improve the oral bioavailability of lovastatin.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Kaempferols/metabolism , Lovastatin/metabolism , Administration, Oral , Animals , Biological Availability , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Intestinal Absorption , Kaempferols/administration & dosage , Lovastatin/administration & dosage , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...