Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.305
Filter
1.
Nat Prod Res ; 38(4): 696-700, 2024.
Article in English | MEDLINE | ID: mdl-36896764

ABSTRACT

Fungal endophytes are valued for biosynthesizing chemically diverse metabolic cascade with interesting biological activities. In the current investigation, two compounds were isolated from Penicillium polonicum, an endophyte of Zingiber officinale. The active moieties, glaucanic acid (1) and dihydrocompactin acid (2) were isolated from the ethyl acetate extract of P. polonicum and characterized by NMR and mass spectroscopy. Further, bioactive potential of the isolated compounds was evaluated by antimicrobial, antioxidant and cytotoxicity assays. Compounds 1 and 2 displayed antifungal activity against phytopathogen Colletotrichum gloeosporioides with more than 50% reduction in its growth. Both the compounds exhibited antioxidant activity against free radicals (DPPH and ABTS) and cytotoxicity activity against cancer cell lines respectively. The compounds, glaucanic acid and dihydrocompactin acid are being reported for the first time from an endophytic fungus. This is the first report on the biological activities of Dihydrocompactin acid produced by endophytic fungal strain.


Subject(s)
Lovastatin/analogs & derivatives , Penicillium , Zingiber officinale , Penicillium/chemistry , Fungi , Antioxidants/pharmacology , Antioxidants/metabolism , Endophytes/chemistry
2.
J Phys Chem B ; 126(37): 7088-7103, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36083294

ABSTRACT

One of the most important areas of medical science is oncology, which is responsible for both the diagnostics and treatment of cancer diseases. Over the years, there has been an intensive development of cancer diagnostics and treatment. This paper shows the comparison of normal (CCD-18Co) and cancerous (CaCo-2) cell lines of the human gastrointestinal tract on the basis of nanomechanical and biochemical properties to obtain information on cancer biomarkers useful in oncological diagnostics. The research techniques used were Raman spectroscopy and imaging and atomic force microscopy (AFM). In addition, the studies also included the effect of the statin compounds─mevastatin, lovastatin, and simvastatin─and their influence on biochemical and nanomechanical changes of cell properties using Raman imaging and AFM techniques. The cytotoxicity of statins was determined using XTT tests.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Simvastatin , Biomarkers, Tumor , Caco-2 Cells , Colon , Dietary Supplements , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Microscopy, Atomic Force/methods , Simvastatin/pharmacology
3.
Spectrochim Acta A Mol Biomol Spectrosc ; 270: 120726, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-34979441

ABSTRACT

One of the most important areas of medical science is oncology, which is responsible for both the diagnostics and treatment of cancer diseases. Simultaneously one of the main challenges of oncology is the development of modern drugs effective in the fight against cancer. Statins are a group of biologically active compounds with the activity of 3-hydroxy-3-methyl glutaryl-CoA reductase inhibitors, an enzyme catalyzing the reduction of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) to mevalonic acid. By acting on this enzyme, statins inhibit the endogenous cholesterol synthesis which in turn causes the reduction of its systemic concentrations. However, in vitro and in vivo studies confirm also the cytostatic and cytotoxic effects of statins against various types of cancer cells including colon cancer. In the presented studies the influence of mevastatin on cancerous colon cells CaCo-2 by Raman spectroscopy and imaging is discussed and compared with biochemistry characteristic for normal colon cells CCD-18Co. Based on vibrational features of colon cells: normal cells CCD-18Co, cancerous cells CaCo-2 and cancerous cells CaCo-2 treated by mevastatin in different concentrations and incubation times we have confirmed the influence of this statin on biochemistry composition of cancerous human colon cells. Moreover, the spectroscopic results for colon normal cells and cancerous cells based on data typical for nucleic acids, proteins, lipids have been compared. The cytotoxisity of mevastatin was determined by using XTT tests.


Subject(s)
Colonic Neoplasms , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Caco-2 Cells , Colonic Neoplasms/drug therapy , Humans , Lovastatin/analogs & derivatives
4.
Biomed Res Int ; 2021: 6687551, 2021.
Article in English | MEDLINE | ID: mdl-34104650

ABSTRACT

In the present study, we examined the synergetic effect of forskolin and mevastatin administration on lipid profile and lipid metabolism in omental adipose tissue in dyslipidemic rats. The study was conducted on forty male albino rats. The rats were randomly classified into four main groups of ten animals in each group as follows: group A, served as control nontreated; group B, rats that received Triton WR 1339 (500 mg/kg); group C, rats that received Triton WR 1339 with forskolin (100% FSK extract 0.5 mg/kg/day) for four weeks; and group D, dyslipidemic rats received both mevastatin and forskolin. At the end of the experimental period, blood and omental adipose tissue samples were collected, preserved, and used for biochemical determination of lipid profile and mRNA expression profile of adenylate cyclase (AC), hormone-sensitive lipase, respectively (HSL), and adenosine monophosphate-activated protein kinase (AMPK). The results showed a significant decline in the serum concentration of total cholesterol, LDL-cholesterol, and triglycerides, although there was a significant increase in serum levels of HDL-cholesterol and glycerol in rats received forskolin alone or with mevastatin when compared with control and dyslipidemic groups. The mRNA expression levels of AC, HSL, and AMPK were significantly increased in omental adipose tissue of rats received forskolin when compared with other groups. In conclusion, forskolin acts synergistically with mevastatin to lower lipid profile and improve lipid metabolism in dyslipidemic rats through upregulation of AMPK expression.


Subject(s)
Adenosine Monophosphate/metabolism , Colforsin/pharmacology , Dyslipidemias/drug therapy , Lipid Metabolism/drug effects , Lipids/physiology , Lovastatin/analogs & derivatives , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Cholesterol, HDL/metabolism , Diet, High-Fat/adverse effects , Dyslipidemias/metabolism , Liver/drug effects , Liver/metabolism , Lovastatin/pharmacology , Male , Rats , Triglycerides/metabolism , Up-Regulation/drug effects
5.
Fitoterapia ; 146: 104685, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32673639

ABSTRACT

Four new compounds, including two lovastatin analogues, terrstatins A and B (1 and 2), and a pair of butenolide derivatives, (±)-asperteretone F (3a/3b), along with eleven known compounds (4-14), were isolated from the Hypericum perforatum endophytic fungus Aspergillus terreus. Their structures and absolute configurations were determined based on extensive spectroscopic analysis, experimental and calculated electronic circular dichroism (ECD) analysis. All isolates were evaluated for cytotoxic activities against five human cancer cell lines, and compounds 3a/3b and 6 showed potential cytotoxic activities against human pancreatic cancer cells, including AsPC-1, SW1990 and PANC-1 cells, with IC50 values ranging from 1.2 to 15.6 µM.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents/pharmacology , Aspergillus/chemistry , Hypericum/microbiology , Pancreatic Neoplasms/pathology , 4-Butyrolactone/isolation & purification , 4-Butyrolactone/pharmacology , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , China , Flowers/microbiology , Humans , Lovastatin/analogs & derivatives , Pancreatic Neoplasms/drug therapy
6.
Clin Epigenetics ; 12(1): 68, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32410704

ABSTRACT

BACKGROUND: Adipogenesis, the process whereby preadipocytes differentiate into mature adipocytes, is crucial for maintaining metabolic homeostasis. Cholesterol-lowering statins increase type 2 diabetes (T2D) risk possibly by affecting adipogenesis and insulin resistance but the (epi)genetic mechanisms involved are unknown. Here, we characterised the effects of statin treatment on adipocyte differentiation using in vitro human preadipocyte cell model to identify putative effective genes. RESULTS: Statin treatment during adipocyte differentiation caused a reduction in key genes involved in adipogenesis, such as ADIPOQ, GLUT4 and ABCG1. Using Illumina's Infinium '850K' Methylation EPIC array, we found a significant hypomethylation of cg14566882, located in the promoter of the histone deacetylase 9 (HDAC9) gene, in response to two types of statins (atorvastatin and mevastatin), which correlates with an increased HDAC9 mRNA expression. We confirmed that HDAC9 is a transcriptional repressor of the cholesterol efflux ABCG1 gene expression, which is epigenetically modified in obesity and prediabetic states. Thus, we assessed the putative impact of ABCG1 knockdown in mimicking the effect of statin in adipogenesis. ABCG1 KD reduced the expression of key genes involved in adipocyte differentiation and decreased insulin signalling and glucose uptake. In human blood cells from two cohorts, ABCG1 expression was impaired in response to statins, confirming that ABCG1 is targeted in vivo by these drugs. CONCLUSIONS: We identified an epigenetic link between adipogenesis and adipose tissue insulin resistance in the context of T2D risk associated with statin use, which has important implications as HDAC9 and ABCG1 are considered potential therapeutic targets for obesity and metabolic diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Adipogenesis/drug effects , Epigenesis, Genetic , Histone Deacetylases/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Repressor Proteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/blood , ATP Binding Cassette Transporter, Subfamily G, Member 1/physiology , Adipogenesis/genetics , Atorvastatin/pharmacology , Cell Line , DNA Methylation , Histone Deacetylases/metabolism , Humans , Insulin/physiology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Promoter Regions, Genetic , Repressor Proteins/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
7.
Biomolecules ; 10(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-32121588

ABSTRACT

Mevastatin (MVS) has been previously shown to induce heme oxygenase (HO)-1 expression through Nox/ROS-dependent PDGFRα/PI3K/Akt/Nrf2/ARE axis in human pulmonary alveolar epithelial cells (HPAEpiCs). However, alternative signaling pathways might involve in MVS-induced HO-1 expression. We found that tumor necrosis factor α (TNFα) induced vascular cell adhesion protein 1 (VCAM-1) expression and NF-κB p65 phosphorylation which were attenuated by pretreatment with MVS via up-regulation of HO-1, determined by Western blot and real-time qPCR. TNFα-induced VCAM-1 expression was attenuated by an NF-κB inhibitor, Bay117082. The inhibitory effects of MVS were reversed by tin protoporphyrin (SnPP)IX (an inhibitor of HO-1 activity). In addition, pretreatment with the inhibitor of pan-Protein kinase C (PKC) (GF109203X), PKCα (Gö6983), Pyk2 (PF431396), p38α MAPK (SB202190), JNK1/2 (SP600125), or AP-1 (Tanshinone IIA), and transfection with their respective siRNAs abolished MVS-induced HO-1 expression in HPAEpiCs. c-Jun (one of AP-1 subunits) was activated by PKCα, Pyk2, p38α MAPK, and JNK1/2, which turned on the transcription of the homx1 gene. The interaction between c-Jun and HO-1 promoter was confirmed by a chromatin immunoprecipitation (ChIP) assay, which was attenuated by these pharmacological inhibitors. These results suggested that MVS induces AP-1/HO-1 expression via PKCα/Pyk2/p38α MAPK- or JNK1/2-dependent c-Jun activation, which further binds with AP-1-binding site on HO-1 promoter and suppresses the TNFα-mediated inflammatory responses in HPAEpiCs. Thus, upregulation of the AP-1/HO-1 system by MVS exerts a potentially therapeutic strategy to protect against pulmonary inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Heme Oxygenase-1/biosynthesis , Lovastatin/analogs & derivatives , Monocytes/metabolism , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Cell Adhesion/drug effects , Cell Line , Humans , Lovastatin/pharmacology
8.
Pharmazie ; 74(11): 658-660, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31739831

ABSTRACT

A novel HPLC method with UV detection was developed and validated in skin penetration (in vitro) studies to identify and quantify lovastatin, mevastatin, rosuvastatin and simvastatin. A Venusil XBP C18 (2), 150 x 4.6 mm, 5 µm column (Agela Technologies, Newark, DE) was used with gradient elution (start at 45 % acetonitrile and increase linearly to 90 % after 1 min; hold at 90 % until 6 min and then re-equilibrate at start conditions) and the mobile phase consisted of (A) Milli-Q ® water and 0.1% orthophosphoric acid, and (B) HPLC grade acetonitrile. The flow rate was set at 1 ml/min, 240 nm UV detection and an injection volume of 10 µl. Linearity was obtained over a range of 0.50-200.00 µg/ml and correlation coefficients ranging from 0.998-1.000 were obtained. Average recovery ranged from 95.9-100.6 %. The LOD and LOQ values obtained from the slope of a calibration curve and the standard deviation of the response ranged from 0.0138-0.0860 µg/ml and 0.0419-0.2615 µg/ml, respectively, where lovastatin and simvastatin could be detected at a concentration similar to the other statins, but could only be quantified at a higher concentration than the remaining statins. The specificity of the method was proved as accurate and quantification of statins was found, even within the incorporation of other compounds.


Subject(s)
Lovastatin/analogs & derivatives , Lovastatin/analysis , Rosuvastatin Calcium/analysis , Simvastatin/analysis , Chromatography, High Pressure Liquid/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/analysis , In Vitro Techniques , Skin Absorption
9.
JCI Insight ; 4(23)2019 12 05.
Article in English | MEDLINE | ID: mdl-31661463

ABSTRACT

Diabetic foot ulcers (DFUs) are a life-threatening disease that often results in lower limb amputations and a shortened life span. Current treatment options are limited and often not efficacious, raising the need for new therapies. To investigate the therapeutic potential of topical statins to restore healing in patients with DFUs, we performed next-generation sequencing on mevastatin-treated primary human keratinocytes. We found that mevastatin activated and modulated the EGF signaling to trigger an antiproliferative and promigratory phenotype, suggesting that statins may shift DFUs from a hyperproliferative phenotype to a promigratory phenotype in order to stimulate healing. Furthermore, mevastatin induced a migratory phenotype in primary human keratinocytes through EGF-mediated activation of Rac1, resulting in actin cytoskeletal reorganization and lamellipodia formation. Interestingly, the EGF receptor is downregulated in tissue biopsies from patients with DFUs. Mevastatin restored EGF signaling in DFUs through disruption of caveolae to promote keratinocyte migration, which was confirmed by caveolin-1 (Cav1) overexpression studies. We conclude that topical statins may have considerable therapeutic potential as a treatment option for patients with DFUs and offer an effective treatment for chronic wounds that can be rapidly translated to clinical use.


Subject(s)
Caveolin 1/metabolism , ErbB Receptors/metabolism , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Signal Transduction/drug effects , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Diabetic Foot , Disease Models, Animal , Female , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Phenotype , Skin/pathology , Swine , Wound Healing/physiology
10.
Atherosclerosis ; 276: 28-38, 2018 09.
Article in English | MEDLINE | ID: mdl-30025252

ABSTRACT

BACKGROUND AND AIMS: Induction of low-density lipoprotein receptor (LDLR) plays a significant role in reduction of plasma LDL-cholesterol (LDL-C) levels. Therefore, strategies that enhance the protein level of LDLR provide an attractive therapeutic target for the treatment of hypercholesterolemia. With this aim in mind, we concentrated our effort on studying the role of AKT kinase in regulation of LDLR levels and proceeded to examine the effect of MK-2206, an allosteric and highly selective AKT inhibitor, on LDLR expression. METHODS: Cultured human hepatoma cells were used to examine the effect of MK-2206 on the proteolytic processing of sterol regulatory element-binding protein-2 (SREBP-2), the expression of LDLR and cellular internalization of LDL. We also examined the effect of MK-2206 on LDLR levels in primary human hepatocytes. RESULTS: MK-2206 induced the proteolytic processing of SREBP-2, upregulated LDLR expression and stimulated LDL uptake. In contrast to statins, induction of LDLR levels by MK-2206 did not rely on 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibition. As a result, cotreatment of cells with MK-2206 and mevastatin potentiated the impact of mevastatin on LDLR. Importantly, MK-2206 stimulated the expression of LDLR by primary human hepatocytes. CONCLUSIONS: MK-2206 is a novel LDLR-inducing agent that, either alone or in combination with statins, exerts a stimulating effect on cellular LDL uptake.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol, LDL/metabolism , Hepatocytes/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Hypercholesterolemia/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, LDL/metabolism , Biological Transport , HeLa Cells , Hep G2 Cells , Hepatocytes/enzymology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypercholesterolemia/enzymology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
11.
Acta Pharmacol Sin ; 39(11): 1804-1815, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29921884

ABSTRACT

Xuezhikang capsule (XZK) is a traditional Chinese medicine that contains lovastatin (Lv) for hyperlipidemia treatment, although it has fewer side effects than Lv. However, the pharmacokinetic mechanisms contributing to its distinct efficacy and low side effects are unclear. Mice were fed a high-fat diet (HFD) for 6 weeks to induce hyperlipidemia. We first conducted the pharmacokinetic studies in HFD mice following oral administration of Lv (10 mg/kg, i.g.) and found that HFD remarkably decreased the active form of Lv (the lovastatin acid, LvA) exposure in the circulation system, especially in the targeting organ liver, with a declined conversion from Lv to LvA, whereas the Lv (responsible for myotoxicity) exposure in muscle markedly increased. Then we compared the pharmacokinetic profiles of Lv in HFD mice after the oral administration of XZK (1200 mg/kg, i.g.) or an equivalent dose of Lv (10 mg/kg, i.g.). A higher exposure of LvA and lower exposure of Lv were observed after XZK administration, suggesting a pharmacokinetic interaction of some ingredients in XZK. Further studies revealed that HFD promoted the inflammation and inhibited carboxylesterase (CES) activities in the intestine and the liver, thus contributing to the lower transformation of Lv into LvA. In contrast, XZK inhibited the inflammation and upregulated CES in the intestine and the liver. Finally, we evaluated the effects of monacolins and phytosterols, the fractional extracts of isoflavones, on inflammatory LS174T or HepG2 cells, which showed that isoflavones inhibited inflammation, upregulated CES, and markedly enhanced the conversion of Lv into LvA. For the first time, we provide evidence that isoflavones and Lv in XZK act in concert to enhance the efficacy and reduce the side effects of Lv.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Hyperlipidemias/drug therapy , Isoflavones/pharmacology , Lovastatin/analogs & derivatives , Lovastatin/therapeutic use , Administration, Oral , Animals , Carboxylesterase/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/metabolism , Drugs, Chinese Herbal/pharmacokinetics , Humans , Inflammation/drug therapy , Lovastatin/administration & dosage , Lovastatin/metabolism , Lovastatin/pharmacokinetics , Male , Mice, Inbred C57BL , Pregnane X Receptor/genetics , Up-Regulation/drug effects
12.
FASEB J ; 32(5): 2841-2854, 2018 05.
Article in English | MEDLINE | ID: mdl-29401610

ABSTRACT

Vimentin is a cytoskeletal intermediate filament protein that is expressed in mesenchymal cells and cancer cells during the epithelial-mesenchymal transition. The goal of this study was to identify vimentin-targeting small molecules by using the Tocriscreen library of 1120 biochemically active compounds. We monitored vimentin filament reorganization and bundling in adrenal carcinoma SW13 vimentin-positive (SW13-vim+) cells via indirect immunofluorescence. The screen identified 18 pharmacologically diverse hits that included 2 statins-simvastatin and mevastatin. Simvastatin induced vimentin reorganization within 15-30 min and significant perinuclear bundling within 60 min (IC50 = 6.7 nM). Early filament reorganization coincided with increased vimentin solubility. Mevastatin produced similar effects at >1 µM, whereas the structurally related pravastatin and lovastatin did not affect vimentin. In vitro vimentin filament assembly assays revealed a direct targeting mechanism, as determined biochemically and by electron microscopy. In SW13-vim+ cells, simvastatin, but not pravastatin, reduced total cell numbers (IC50 = 48.1 nM) and promoted apoptosis after 24 h. In contrast, SW13-vim- cell viability was unaffected by simvastatin, unless vimentin was ectopically expressed. Simvastatin similarly targeted vimentin filaments and induced cell death in MDA-MB-231 (vim+), but lacked effect in MCF7 (vim-) breast cancer cells. In conclusion, this study identified vimentin as a direct molecular target that mediates simvastatin-induced cell death in 2 different cancer cell lines.-Trogden, K. P., Battaglia, R. A., Kabiraj, P., Madden, V. J., Herrmann, H., Snider, N. T. An image-based small-molecule screen identifies vimentin as a pharmacologically relevant target of simvastatin in cancer cells.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Simvastatin/pharmacology , Vimentin/metabolism , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/ultrastructure , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Death , Female , Humans , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , MCF-7 Cells , Microscopy, Fluorescence , Neoplasm Proteins/antagonists & inhibitors , Vimentin/antagonists & inhibitors
13.
Arterioscler Thromb Vasc Biol ; 38(3): 592-598, 2018 03.
Article in English | MEDLINE | ID: mdl-29284604

ABSTRACT

OBJECTIVE: Evolocumab, a PCSK9 (proprotein convertase subtilisin kexin type 9)-neutralizing antibody, lowers low-density lipoprotein cholesterol (LDL-C) in homozygous familial hypercholesterolemic (HoFH) patients with reduced LDLR (low-density lipoprotein receptor) function. However, their individual responses are highly variable, even among carriers of identical LDLR genetic defects. We aimed to elucidate why HoFH patients variably respond to PCSK9 inhibition. APPROACH AND RESULTS: Lymphocytes were isolated from 22 HoFH patients enrolled in the TAUSSIG trial (Trial Assessing Long Term Use of PCSK9 Inhibition in Subjects With Genetic LDL Disorders). Ten patients were true homozygotes (FH1/FH1) and 5 identical compound heterozygotes (FH1/FH2). Lymphocytes were plated with or without mevastatin, recombinant PCSK9 (rPCSK9), or a PCSK9-neutralizing antibody. Cell surface LDLR expression was analyzed by flow cytometry. All HoFH lymphocytes had reduced cell surface LDLR expression compared with non-FH lymphocytes, for each treatment modality. Lymphocytes from FH1/FH2 patients (LDLR defective/negative) displayed the lowest LDLR expression levels followed by lymphocytes from FH1/FH1 patients (defective/defective). Mevastatin increased, whereas rPCSK9 reduced LDLR expression. The PCSK9-neutralizing antibody restored LDLR expression. Lymphocytes displaying higher LDLR expression levels were those isolated from patients presenting with lowest levels of LDL-C and apolipoprotein B, before and after 24 weeks of evolocumab treatment. These negative correlations remained significant in FH1/FH1 patients and appeared more pronounced when patients with apolipoprotein E3/E3 genotypes were analyzed separately. Significant positive correlations were found between the levels of LDLR expression and the percentage reduction in LDL-C on evolocumab treatment. CONCLUSIONS: Residual LDLR expression in HoFH is a major determinant of LDL-C levels and seems to drive their individual response to evolocumab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Anticholesteremic Agents/therapeutic use , Homozygote , Hyperlipoproteinemia Type II/drug therapy , Lymphocytes/drug effects , Mutation , PCSK9 Inhibitors , Receptors, LDL/genetics , Serine Proteinase Inhibitors/therapeutic use , Adolescent , Adult , Antibodies, Monoclonal, Humanized , Apolipoprotein B-100/blood , Cells, Cultured , Cholesterol, LDL/blood , Drug Therapy, Combination , Ezetimibe/therapeutic use , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/genetics , Lovastatin/analogs & derivatives , Lovastatin/therapeutic use , Lymphocytes/enzymology , Male , Middle Aged , Phenotype , Receptors, LDL/metabolism , Treatment Outcome , Young Adult
14.
J Biol Chem ; 293(4): 1439-1449, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29158265

ABSTRACT

Diabetic foot ulcers (DFUs), a life-threatening complication of diabetes mellitus, have limited treatment options, often resulting in amputations. HMG-CoA reductase inhibitors such as statins are cholesterol-reducing agents that may provide a new therapeutic option. Statins target the cholesterol pathway and block the synthesis of the wound-healing inhibitors farnesyl pyrophosphate (FPP) and cortisol, ligands for the glucocorticoid receptor (GR). Here we demonstrate that the naturally occurring statin mevastatin reverses FPP's effects and promotes healing by using in vitro wound healing assays, human ex vivo and porcine in vivo wound models, and DFU tissue. Moreover, we measured cortisol levels by ELISA and found that mevastatin inhibited cortisol synthesis in keratinocytes and biopsies from patients with DFU. Of note, topical mevastatin stimulated epithelialization and angiogenesis in vivo Mevastatin also reversed FPP-mediated induction of the GR target, the transcription factor c-Myc (a biomarker of non-healing wounds), in porcine and human wound models. Importantly, mevastatin reversed c-Myc overexpression in DFUs. It induced expression of the long noncoding RNA Gas5 that blocks c-Myc expression, which was confirmed by overexpression studies. We conclude that topical mevastatin accelerates wound closure by promoting epithelialization via multiple mechanisms: modulation of GR ligands and induction of the long noncoding RNA Gas5, leading to c-Myc inhibition. In light of these findings, we propose that repurposing statin drugs for topical treatment of DFUs may offer another option for managing this serious condition.


Subject(s)
Gene Expression Regulation/drug effects , Keratinocytes/metabolism , Lovastatin/analogs & derivatives , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Long Noncoding/metabolism , Receptors, Glucocorticoid/metabolism , Wound Healing/drug effects , Administration, Topical , Diabetic Foot/drug therapy , Diabetic Foot/genetics , Diabetic Foot/metabolism , Diabetic Foot/pathology , Humans , Keratinocytes/pathology , Lovastatin/pharmacology , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics
15.
Biochim Biophys Acta Proteins Proteom ; 1866(1): 23-31, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28583351

ABSTRACT

Cytochrome P450 enzymes belonging to the CYP105 family are predominantly found in bacteria belonging to the phylum Actinobacteria and the order Actinomycetales. In this review, we focused on the protein engineering of P450s belonging to the CYP105 family for industrial use. Two Arg substitutions to Ala of CYP105A1 enhanced its vitamin D3 25- and 1α-hydroxylation activities by 400 and 100-fold, respectively. The coupling efficiency between product formation and NADPH oxidation was largely improved by the R84A mutation. The quintuple mutant Q87W/T115A/H132L/R194W/G294D of CYP105AB3 showed a 20-fold higher activity than the wild-type enzyme. Amino acids at positions 87 and 191 were located at the substrate entrance channel, and that at position 294 was located close to the heme group. Semi-rational engineering of CYP105A3 selected the best performing mutant, T85F/T119S/V194N/N363Y, for producing pravastatin. The T119S and N363Y mutations synergistically had remarkable effects on the interaction between CYP105A3 and putidaredoxin. Although wild-type CYP105AS1 hydroxylated compactin to 6-epi-pravastatin, the quintuple mutant I95T/Q127R/A180V/L236I/A265N converted almost all compactin to pravastatin. Five amino acid substitutions by two rounds of mutagenesis almost completely changed the stereo-selectivity of CYP105AS1. These results strongly suggest that the protein engineering of CYP105 enzymes greatly increase their industrial utility. This article is part of a Special Issue entitled: Cytochrome P450 biodiversity and biotechnology, edited by Erika Plettner, Gianfranco Gilardi, Luet Wong, Vlada Urlacher, Jared Goldstone.


Subject(s)
Actinobacteria/genetics , Amino Acid Substitution , Bacterial Proteins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Mutation , Protein Engineering/methods , Actinobacteria/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholecalciferol/metabolism , Conserved Sequence , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Ferredoxins/metabolism , Gene Expression , Hydroxylation , Isoenzymes , Lovastatin/analogs & derivatives , Lovastatin/metabolism , Molecular Docking Simulation , Pravastatin/biosynthesis , Streptomyces/enzymology , Streptomyces/genetics , Substrate Specificity
16.
Phytochemistry ; 145: 146-152, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29132077

ABSTRACT

Eight previously undescribed metabolites including of lovastatin analogues, a pair of diastereoisomers, a cyclopentenone dimer, and three polyketides were isolated from the culture of Aspergillus terreus YIM PH30711. Two types of unprecedented skeletons, benzene-cyclopentanone complex and linear polyketide, and an unusual dimer structure were determined by spectral analysis. Compound, 3α-hydroxy-3,5-dihydromonacolin L showed moderate activity against HMG-CoA reductase, with an inhibition ratio of 34% at the concentration of 50 µM, while lovastatin and dihydromonacolin K ethyl ester presented much stronger activity against HMGR with inhibition rates of 85% and 90% at the concentration of 50 µM, respectively. Aspereusin A was active against AChE with a ratio of 62% at the concentration of 50 µM, while its stereomers did not showed obvious inhibition (<10%). The configuration at C-4 of these three diastereoisomers was crucial in the inhibition against AChE, and the ß-orientation of substituted methoxyl acrylic acid should be beneficial to the combining with AChE.


Subject(s)
Acyl Coenzyme A/antagonists & inhibitors , Aspergillus/chemistry , Enzyme Inhibitors/pharmacology , Lovastatin/pharmacology , Acetylcholinesterase/metabolism , Acyl Coenzyme A/metabolism , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Erythrocytes/enzymology , Humans , Lovastatin/analogs & derivatives , Lovastatin/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Stereoisomerism , Structure-Activity Relationship
17.
Metab Brain Dis ; 32(6): 1999-2007, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28840430

ABSTRACT

Statins or HMG-CoA reductase inhibitors have been shown to be effective at lowering cholesterol levels, and the application of these molecules has gradually emerged as an attractive therapeutic strategy for neurodegenerative diseases. Epidemiological studies suggest that statin use is associated with a decreased incidence of Alzheimer's disease (AD). Thus, statins may play a beneficial role in reducing amyloid ß (Aß) toxicity, the most relevant pathological feature and pathogenesis of AD. However, the precise mechanisms involved in statin-inhibited Aß toxicity remain unclear. In the present study, we report that mevastatin significantly protects against Aß-induced neurotoxicity in SK-N-MC neuronal cells by restoring impaired insulin signaling. This protection appears to be associated with the activation of AMP-activated protein kinase (AMPK), which has long been known to increase insulin sensitivity. Our results also indicate that high levels of cholesterol likely underlie Aß-induced neurotoxicity and that activation of AMPK by mevastatin alleviates insulin resistance. Signaling through the insulin receptor substrate-1/Akt pathway appears to lead to cell survival. These findings demonstrate that mevastatin plays a potential therapeutic role in targeting Aß-mediated neurotoxicity. The molecule presents a novel therapeutic strategy for further studies in AD prevention and therapeutics.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Amyloid beta-Peptides/pharmacology , Cell Survival/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/analogs & derivatives , Neurons/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lovastatin/pharmacology , Neurons/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
18.
J Biotechnol ; 258: 171-180, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28751276

ABSTRACT

The synthesis and enzymatic reduction of several 6-substituted dioxohexanoates are presented. Two-step syntheses of tert-butyl 6-bromo-3,5-dioxohexanoate and the corresponding 6-hydroxy compound have been achieved in 89% and 59% yield, respectively. Regio- and enantioselective reduction of these diketones and of the 6-chloro derivative with alcohol dehydrogenase from Lactobacillus brevis (LBADH) gave the (5S)-5-hydroxy-3-oxo products with enantiomeric excesses of 91%, 98.4%, and >99.5%, respectively. Chain elongation of the reduction products by one carbon via cyanide addition, and by more than one carbon by Julia-Kocienski olefination, gave access to well-established statine side-chain building blocks. Application in the synthesis of the cholesterol-lowering natural compound solistatin is given.


Subject(s)
Amino Acids/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Lovastatin/analogs & derivatives , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/metabolism , Amino Acids/chemical synthesis , Amino Acids/metabolism , Caproates/chemical synthesis , Caproates/chemistry , Caproates/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Levilactobacillus brevis/enzymology , Lovastatin/chemical synthesis , Lovastatin/chemistry , Lovastatin/metabolism , Models, Molecular , NADP/chemistry , NADP/metabolism , Oxidoreductases/metabolism
19.
J Microbiol Biotechnol ; 27(5): 956-964, 2017 May 28.
Article in English | MEDLINE | ID: mdl-28274099

ABSTRACT

Compactin and pravastatin are competitive cholesterol biosynthesis inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase and belong to the statin drugs; however, the latter shows superior pharmacokinetic characteristics. Previously, we reported that the bacterial P450, CYP105D7, from Streptomyces avermitilis can catalyze the hydroxylation of 1-deoxypentalenic acid, diclofenac, and naringenin. Here, we demonstrate that CYP105D7 could also catalyze compactin hydroxylation in vitro. In the presence of both bacterial and cyanobacterial redox partner systems with an NADPH regeneration system, the reaction produced two hydroxylated products, including pravastatin (hydroxylated at the C6 position). The steady-state kinetic parameters were measured using the redox partners of putidaredoxin and its reductase. The Km and kcat values for compactin were 39.1 ± 8.8 µM and 1.12 ± 0.09 min-1, respectively. The kcat/Km value for compactin (0.029 min-1·µM-1) was lower than that for diclofenac (0.114 min-1·µM-1). Spectroscopic analysis showed that CYP105D7 binds to compactin with a Kd value of 17.5 ± 3.6 µM. Molecular docking analysis was performed to build a possible binding model of compactin. Comparisons of different substrates with CYP105D7 were conclusively illustrated for the first time.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Lovastatin/analogs & derivatives , Streptomyces/metabolism , Biotechnology/methods , Catalytic Domain , Cytochrome P-450 Enzyme System/chemistry , Ferredoxins/metabolism , Hydroxylation , Kinetics , Lovastatin/metabolism , Molecular Docking Simulation , Oxidation-Reduction , Pravastatin/chemistry , Pravastatin/metabolism , Spectrophotometry, Ultraviolet , Streptomyces/enzymology
20.
Oncotarget ; 8(11): 17833-17848, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147319

ABSTRACT

Histone deacetylase inhibitors (HDACi) are promising anti-cancer agents, and combining a HDACi with other agents is an attractive therapeutic strategy in solid tumors. We report here that mevastatin increases HDACi LBH589-induced cell death in triple-negative breast cancer (TNBC) cells. Combination treatment inhibited autophagic flux by preventing Vps34/Beclin 1 complex formation and downregulating prenylated Rab7, an active form of the small GTPase necessary for autophagosome-lysosome fusion. This means that co-treatment with mevastatin and LBH589 activated LKB1/AMPK signaling and subsequently inhibited mTOR. Co-treatment also led to cell cycle arrest in G2/M phase and induced corresponding expression changes of proteins regulating the cell cycle. Co-treatment also increased apoptosis both in vitro and in vivo, and reduced tumor volumes in xenografted mice. Our results indicate that disruption of autophagosome-lysosome fusion likely underlies mevastatin-LBH589 synergistic anticancer effects. This study confirms the synergistic efficacy of, and demonstrates a potential therapeutic role for mevastatin plus LBH589 in targeting aggressive TNBC, and presents a novel therapeutic strategy for further clinical study. Further screening for novel autophagy modulators could be an efficient approach to enhance HDACi-induced cell death in solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Lovastatin/analogs & derivatives , Lysosomes/metabolism , Triple Negative Breast Neoplasms/drug therapy , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Beclin-1/metabolism , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/metabolism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Lovastatin/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Panobinostat , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , rab GTP-Binding Proteins/biosynthesis , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...