Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 239
Filter
1.
Drug Metab Dispos ; 49(8): 706-717, 2021 08.
Article in English | MEDLINE | ID: mdl-34011532

ABSTRACT

The squalene synthase inhibitor squalestatin 1 (Squal1) is a potent and efficacious inducer of CYP2B expression in primary cultured rat hepatocytes and rat liver. To determine whether Squal1 is also an inducer of human CYP2B, the effects of Squal1 treatment were evaluated in primary cultured human hepatocytes, differentiated HepaRG cells, and humanized mouse livers. Squal1 treatment did not increase CYP2B6 mRNA levels in human hepatocytes or HepaRG cells and only slightly and inconsistently increased CYP2B6 mRNA content in humanized mouse liver. However, treatment with farnesol, which mediates Squal1's effect on rat CYP2B expression, increased CYP2B6 mRNA levels in HepaRG cells expressing the constitutive androstane receptor (CAR), but not in cells with knocked-down CAR. To determine the impact of cholesterol biosynthesis inhibition on CAR activation, the effects of pravastatin (Prava) were determined on CITCO-mediated gene expression in primary cultured human hepatocytes. Prava treatment abolished CITCO-inducible CYP2B6 expression, but had less effect on rifampicin-mediated CYP3A4 induction, and CITCO treatment did not affect Prava-inducible HMG-CoA reductase (HMGCR) expression. Treatment with inhibitors of different steps of cholesterol biosynthesis attenuated CITCO-mediated CYP2B6 induction in HepaRG cells, and Prava treatment increased HMGCR expression and inhibited CYP2B6 induction with comparable potency. Transfection of HepG2 cells with transcriptionally active sterol regulatory element binding proteins (SREBPs) reduced CAR-mediated transactivation, and inducible expression of transcriptionally active SREBP2 attenuated CITCO-inducible CYP2B6 expression in HepaRG cells. These findings suggest that Squal1 does not induce CYP2B6 in human hepatocytes because Squal1's inhibitory effect on cholesterol biosynthesis interferes with CAR activation. SIGNIFICANCE STATEMENT: The cholesterol biosynthesis inhibitor squalestatin 1 induces rat hepatic CYP2B expression indirectly by causing accumulation of an endogenous isoprenoid that activates the constitutive androstane receptor (CAR). This study demonstrates that squalestatin 1 does not similarly induce CYP2B6 expression in human hepatocytes. Rather, inhibition of cholesterol biosynthesis interferes with CAR activity, likely by activating sterol regulatory element binding proteins. These findings increase our understanding of the endogenous processes that modulate human drug-metabolizing gene expression.


Subject(s)
Anticholesteremic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholesterol/biosynthesis , Constitutive Androstane Receptor/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Tricarboxylic Acids/pharmacology , Animals , Cell Line , Cytochrome P-450 CYP2B6/biosynthesis , Cytochrome P-450 CYP2D6/biosynthesis , Cytochrome P-450 CYP2D6/genetics , Farnesol/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/enzymology , Mice , Pravastatin/pharmacology , Rats
2.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884533

ABSTRACT

This study aimed to evaluate the in vitro effect of the novel bioactive adhesive monomer CMET, a calcium salt of 4-methacryloxyethyl trimellitate acid (4-MET), on human dental pulp stem cells (hDPSCs) and its capacity to induce tertiary dentin formation in a rat pulp injury model. Aqueous solutions of four tested materials [4-MET, CMET, Ca(OH)2, and mineral trioxide aggregate (MTA)] were added to the culture medium upon confluence, and solvent (dH2O) was used as a control. Cell proliferation was assessed using the Cell Counting Kit-8 assay, and cell differentiation was evaluated by real-time quantitative reverse transcription-polymerase chain reaction. The mineralization-inducing capacity was evaluated using alizarin red S staining and an alkaline phosphatase activity assay. For an in vivo experiment, a mechanical pulp exposure model was prepared on Wistar rats; damaged pulp was capped with Ca(OH)2 or CMET. Cavities were sealed with composite resin, and specimens were assessed after 14 and 28 days. The in vitro results showed that CMET exhibited the lowest cytotoxicity and highest odontogenic differentiation capacity among all tested materials. The favorable outcome on cell mineralization after treatment with CMET involved p38 and c-Jun N-terminal kinases signaling. The nuclear factor kappa B pathway was involved in the CMET-induced mRNA expression of odontogenic markers. Similar to Ca(OH)2, CMET produced a continuous hard tissue bridge at the pulp exposure site, but treatment with only CMET produced a regular dentinal tubule pattern. The findings suggest that (1) the evaluated novel bioactive adhesive monomer provides favorable biocompatibility and odontogenic induction capacity and that (2) CMET might be a very promising adjunctive for pulp-capping materials.


Subject(s)
Dental Pulp/cytology , Dentin/cytology , Methacrylates/pharmacology , Odontoblasts/cytology , Odontogenesis , Regeneration , Stem Cells/cytology , Tricarboxylic Acids/pharmacology , Adhesives , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dental Pulp/drug effects , Dental Pulp/metabolism , Dentin/drug effects , Dentin/metabolism , Male , Odontoblasts/drug effects , Odontoblasts/metabolism , Rats , Rats, Wistar , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism
3.
Bull Exp Biol Med ; 169(4): 508-511, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910380

ABSTRACT

A stable preparation of agaricinic acid nanoparticles was obtained. The mean hydrodynamic size of nanoparticles according to photon correlation spectroscopy was 200 nm and zeta potential was -57 mV. Cytotoxic activity of agaricinic acid nanoparticles against human HepG2 hepatoma cells was evaluated. Nanoparticles with a low concentration of agaricinic acid stimulated and with high concentration - suppressed metabolic activity and viability of hepatoma cells. The EC50 for the stimulating effect was 32.8 µg/ml, and the IC50=602.1 mg/ml. The preparation of agaricinic acid nanoparticles can be used in medicine as a potential antitumor agent.


Subject(s)
Alkanes/pharmacology , Antineoplastic Agents/pharmacology , Coriolaceae/chemistry , Nanoparticles/chemistry , Tricarboxylic Acids/pharmacology , Alkanes/isolation & purification , Antineoplastic Agents/isolation & purification , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fruiting Bodies, Fungal/chemistry , Hep G2 Cells , Humans , Particle Size , Tricarboxylic Acids/isolation & purification
4.
J Lipid Res ; 60(3): 579-593, 2019 03.
Article in English | MEDLINE | ID: mdl-30622150

ABSTRACT

Geranylgeranoic acid (GGA) has been reported to induce autophagic cell death via upregulation of lipid-induced unfolded protein response in several human hepatoma-derived cell lines, and its 4,5-didehydro derivative has been developed as a preventive agent against second primary hepatoma in clinical trials. We have previously reported that GGA is a natural diterpenoid synthesized in several medicinal herbs. Here, we provide unequivocal evidence for de novo GGA biosynthesis in mammals. First, with normal male Wistar rats, the levels of GGA in liver were found to be far greater than those in other organs analyzed. Second, we demonstrated the metabolic GGA labeling from the 13C-labeled mevalonolactone in the human hepatoma-derived cell line, HuH-7. Isotopomer spectral analysis revealed that approximately 80% of the cellular GGA was newly synthesized from mevalonate (MVA) in 12 h and the acid picked up preexisting farnesyl diphosphate (FPP) and geranylgeranyl diphosphate (GGPP), suggesting that GGA is derived from FPP and GGPP through the MVA pathway. Third, zaragozic acid A, a squalene synthase inhibitor, induced dose-dependent upregulation of endogenous GGA content in HuH-7 cells and their concomitant cell death. These results strongly suggest that a cancer-preventive GGA is biosynthesized via the MVA pathway in mammals.


Subject(s)
Diterpenes/metabolism , Mevalonic Acid/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Male , Rats , Rats, Wistar , Tricarboxylic Acids/pharmacology
5.
Biochem Biophys Res Commun ; 512(3): 517-523, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30904161

ABSTRACT

Invasive aspergillosis (IA) is a life-threatening disease impacting immunocompromised individuals. Standard treatments of IA, including polyenes and azoles, suffer from high toxicity and emerging resistance, leading to the need to develop new antifungal agents with novel mechanisms of action. Ergosterol biosynthesis is a classic target for antifungals, and squalene synthase (SQS) catalyzes the first committed step in ergosterol biosynthesis in Aspergillus spp. making SQS of interest in the context of antifungal development. Here, we cloned, expressed, purified and characterized SQS from the pathogen Aspergillus flavus (AfSQS), confirming that it produced squalene. To identify potential leads targeting AfSQS, we tested known squalene synthase inhibitors, zaragozic acid and the phosphonosulfonate BPH-652, finding that they were potent inhibitors. We then screened a library of 744 compounds from the National Cancer Institute (NCI) Diversity Set V for inhibition activity. 20 hits were identified and IC50 values were determined using dose-response curves. 14 compounds that interfered with the assay were excluded and the remaining 6 compounds were analyzed for drug-likeness, resulting in one compound, celastrol, which had an AfSQS IC50 value of 830 nM. Enzyme inhibition kinetics revealed that celastrol binds to AfSQS in a noncompetitive manner, but did not bind covalently. Since celastrol is also known to inhibit growth of the highly virulent Aspergillus fumigatus by inhibiting flavin-dependent monooxygenase siderophore A (SidA, under iron starvation conditions), it may be a promising multi-target lead for antifungal development.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus flavus/enzymology , Enzyme Inhibitors/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus flavus/genetics , Aspergillus flavus/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cloning, Molecular , Farnesyl-Diphosphate Farnesyltransferase/genetics , Humans , Models, Molecular , Molecular Targeted Therapy , Pentacyclic Triterpenes , Tricarboxylic Acids/pharmacology , Triterpenes/pharmacology
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 649-659, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29247837

ABSTRACT

The production of amyloid-ß (Aß) is the key factor driving pathogenesis in Alzheimer's disease (AD). Increasing concentrations of Aß within the brain cause synapse degeneration and the dementia that is characteristic of AD. Here the factors that affect the release of disease-relevant forms Aß were studied in a cell model. 7PA2 cells expressing the human amyloid precursor protein released soluble Aß oligomers that caused synapse damage in cultured neurons. Supernatants from 7PA2 cells treated with the cholesterol synthesis inhibitor squalestatin contained similar concentrations of Aß42 to control cells but did not cause synapse damage in neuronal cultures. These supernatants contained reduced concentrations of Aß42 oligomers and increased concentrations of Aß42 monomers. Treatment of 7PA2 cells with platelet-activating factor (PAF) antagonists had similar effects; it reduced concentrations of Aß42 oligomers and increased concentrations of Aß42 monomers in cell supernatants. PAF activated cholesterol ester hydrolases (CEH), enzymes that released cholesterol from stores of cholesterol esters. Inhibition of CEH also reduced concentrations of Aß42 oligomers and increased concentrations of Aß42 monomers in cell supernatants. The Aß monomers produced by treated cells protected neurons against Aß oligomer-induced synapse damage. These studies indicate that pharmacological manipulation of cells can alter the ratio of Aß monomer:oligomer released and consequently their effects on synapses.


Subject(s)
Amyloid beta-Peptides/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/pharmacology , Sterol Esterase/antagonists & inhibitors , Synapses/drug effects , Tricarboxylic Acids/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Down-Regulation/drug effects , Embryo, Mammalian , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Polymers/metabolism , Synapses/metabolism
7.
Mol Cell Biochem ; 441(1-2): 109-124, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28887754

ABSTRACT

Lichens are a source of secondary metabolites which possess important biological activities, including antioxidant, antibacterial, anti-inflammatory, and cytotoxic effects. The anticancer activity of lichens was shown in many types of tumors, including colorectal cancers (CRC). Several studies revealed that the application of lichen extracts diminished the proliferation of CRC cells and induced apoptosis. Colon carcinogenesis is associated with aberrations in Wnt signaling. Elevated transcriptional activity of ß-catenin induces cell survival, proliferation, and migration. Thus, the inhibition of Wnt signaling is a promising therapeutic strategy in colorectal cancer. The aim of this study was the evaluation of the effects of lichen-derived depsides (atranorin, lecanoric acid, squamatic acid) and depsidones (physodic acid, salazinic acid) and a poly-carboxylic fatty acid-caperatic acid, on Wnt signaling in HCT116 and DLD-1 colorectal cancer cell lines. HCT116 cells were more sensitive to the modulatory effects of the compounds. PKF118-310, which was used as a reference ß-catenin inhibitor, dose-dependently reduced the expression of the classical ß-catenin target gene-Axin2 in both cell lines. Lecanoric acid slightly reduced Axin2 expression in HCT116 cells while caperatic acid tended to reduce Axin2 expression in both cell lines. Physodic acid much more potently decreased Axin2 expression in HCT116 cells than in DLD-1 cells. Physodic acid and caperatic acid also diminished the expression of survivin and MMP7 in a cell line and time-dependent manner. None of the compounds affected the nuclear translocation of ß-catenin. This is the first report showing the ability of caperatic acid and physodic acid to modulate ß-catenin-dependent transcription.


Subject(s)
Colorectal Neoplasms/metabolism , Dibenzoxepins/pharmacology , Lichens/chemistry , Tricarboxylic Acids/pharmacology , Wnt Signaling Pathway/drug effects , Axin Protein/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dibenzoxepins/chemistry , Humans , Neoplasm Proteins/metabolism , Tricarboxylic Acids/chemistry
8.
J Cell Physiol ; 231(11): 2452-63, 2016 11.
Article in English | MEDLINE | ID: mdl-26916741

ABSTRACT

Skin produces cholesterol and a wide array of sterols and non-sterol mevalonate metabolites, including isoprenoid derivative farnesyl pyrophosphate (FPP). To characterize FPP action in epidermis, we generated transcriptional profiles of primary human keratinocytes treated with zaragozic acid (ZGA), a squalene synthase inhibitor that blocks conversion of FPP to squalene resulting in endogenous accumulation of FPP. The elevated levels of intracellular FPP resulted in regulation of epidermal differentiation and adherens junction signaling, insulin growth factor (IGF) signaling, oxidative stress response and interferon (IFN) signaling. Immunosuppressive properties of FPP were evidenced by STAT-1 downregulation and prominent suppression of its nuclear translocation by IFNγ. Furthermore, FPP profoundly downregulated genes involved in epidermal differentiation of keratinocytes in vitro and in human skin ex vivo. Elevated levels of FPP resulted in induction of cytoprotective transcriptional factor Nrf2 and its target genes. We have previously shown that FPP functions as ligand for the glucocorticoid receptor (GR), one of the major regulator of epidermal homeostasis. Comparative microarray analyses show significant but not complete overlap between FPP and glucocorticoid regulated genes, suggesting that FPP may have wider transcriptional impact. This was further supported by co-transfection and chromatin immunoprecipitation experiments where we show that upon binding to GR, FPP recruits ß-catenin and, unlike glucocorticoids, recruits co-repressor GRIP1 to suppress keratin 6 gene. These findings have many clinical implications related to epidermal lipid metabolism, response to glucocorticoid therapy as well as pleiotropic effects of cholesterol lowering therapeutics, statins. J. Cell. Physiol. 231: 2452-2463, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Movement/drug effects , Epidermis/pathology , Inflammation/pathology , Oxidative Stress/drug effects , Polyisoprenyl Phosphates/pharmacology , Sesquiterpenes/pharmacology , Skin/metabolism , Adherens Junctions/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cell Movement/genetics , Cells, Cultured , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Insulin-Like Growth Factor I/metabolism , Interferons/metabolism , Keratin-6/genetics , Keratin-6/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Models, Biological , NF-E2-Related Factor 2/metabolism , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oxidative Stress/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tricarboxylic Acids/pharmacology , Wound Healing/drug effects , beta Catenin/metabolism
9.
J Pharmacol Exp Ther ; 358(2): 216-29, 2016 08.
Article in English | MEDLINE | ID: mdl-27225895

ABSTRACT

Squalene synthase inhibitors (SSIs), such as squalestatin 1 (SQ1), reduce cholesterol biosynthesis but cause the accumulation of isoprenoids derived from farnesyl pyrophosphate (FPP), which can modulate the activity of nuclear receptors, including the constitutive androstane receptor (CAR), farnesoid X receptor, and peroxisome proliferator-activated receptors (PPARs). In comparison, 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (e.g., pravastatin) inhibit production of both cholesterol and nonsterol isoprenoids. To characterize the effects of isoprenoids on hepatocellular physiology, microarrays were used to compare orthologous gene expression from primary cultured mouse and rat hepatocytes that were treated with either SQ1 or pravastatin. Compared with controls, 47 orthologs were affected by both inhibitors, 90 were affected only by SQ1, and 51 were unique to pravastatin treatment (P < 0.05, ≥1.5-fold change). When the effects of SQ1 and pravastatin were compared directly, 162 orthologs were found to be differentially coregulated between the two treatments. Genes involved in cholesterol and unsaturated fatty acid biosynthesis were up-regulated by both inhibitors, consistent with cholesterol depletion; however, the extent of induction was greater in rat than in mouse hepatocytes. SQ1 induced several orthologs associated with microsomal, peroxisomal, and mitochondrial fatty acid oxidation and repressed orthologs involved in cell cycle regulation. By comparison, pravastatin repressed the expression of orthologs involved in retinol and xenobiotic metabolism. Several of the metabolic genes altered by isoprenoids were inducible by a PPARα agonist, whereas cytochrome P450 isoform 2B was inducible by activators of CAR. Our findings indicate that SSIs uniquely influence cellular lipid metabolism and cell cycle regulation, probably due to FPP catabolism through the farnesol pathway.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholesterol/biosynthesis , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Pravastatin/pharmacology , Terpenes/metabolism , Tricarboxylic Acids/pharmacology , Animals , Drug Synergism , Female , Male , Mice , Rats , Sequence Homology, Nucleic Acid
10.
Drug Metab Dispos ; 44(3): 352-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700959

ABSTRACT

Farnesyl pyrophosphate (FPP) is a branch-point intermediate in the mevalonate pathway that is normally converted mainly to squalene by squalene synthase in the first committed step of sterol biosynthesis. Treatment with the squalene synthase inhibitor squalestatin 1 (SQ1) causes accumulation of FPP, its dephosphorylated metabolite farnesol, and several oxidized farnesol-derived metabolites. In addition, SQ1 treatment of primary cultured rat hepatocytes increases CYP2B expression through a mechanism that requires FPP synthesis and activation of the constitutive androstane receptor (CAR). Because direct farnesol treatment also increases CYP2B expression, it seems likely that SQ1-mediated CAR activation requires FPP dephosphorylation to farnesol. The lipid phosphatase, phosphatidic acid phosphatase domain containing 2 (PPAPDC2), was recently reported to catalyze FPP dephosphorylation. We therefore determined the effect of overexpressing or knocking down PPAPDC2 on SQ1-mediated CAR activation in primary cultured rat hepatocytes. Cotransfection of rat hepatocytes with a plasmid expressing rat or human PPAPDC2 enhanced SQ1-mediated activation of a CAR-responsive reporter by 1.7- or 2.4-fold over the SQ1-mediated activation that was produced when hepatocytes were cotransfected with empty expression plasmid. Similarly, transduction of rat hepatocytes with a recombinant adenovirus expressing PPAPDC2 enhanced SQ1-mediated CYP2B1 mRNA induction by 1.4-fold over the induction that was seen in hepatocytes transduced with control adenovirus. Cotransfection with a short hairpin RNA targeting PPAPDC2 reduced SQ1-mediated CAR activation by approximately 80% relative to the activation that occurred in hepatocytes transfected with nontargeting short hairpin RNA. These results indicate that PPAPDC2 plays an important role in SQ1-mediated CAR activation, most likely by catalyzing the conversion of FPP to farnesol.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Phosphatidate Phosphatase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tricarboxylic Acids/pharmacology , Animals , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B1/metabolism , Humans , Male , Polyisoprenyl Phosphates/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sesquiterpenes/metabolism , Transfection/methods
11.
Biochim Biophys Acta ; 1841(7): 977-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24747199

ABSTRACT

2,3-Oxidosqualene is an intermediate in cholesterol biosynthesis and 2,3:22,23-dioxidosqualene act as the substrate for an alternative pathway that produces 24(S),25-epoxycholesterol which effects cholesterol homeostasis. In light of our previous findings concerning the biological effects of certain epoxidated all-trans-polyisoprenes, the effects of squalene carrying epoxy moieties on the second and third isoprene residues were investigated here. In cultures of HepG2 cells both monoepoxides of squalene and one of their hydrolytic products inhibited cholesterol synthesis and stimulated the synthesis of coenzyme Q (CoQ). Upon prolonged treatment the cholesterol content of these cells and its labeling with [(3)H]mevalonate were reduced, while the amount and labeling of CoQ increased. Injection of the squalene monoepoxides into mice once daily for 6days elevated the level of CoQ in their blood, but did not change the cholesterol level. The same effects were observed upon treatment of apoE-deficient mice and diabetic GK-rats. This treatment increased the hepatic level of CoQ10 in mice, but the amount of CoQ9, which is the major form, was unaffected. The presence of the active compounds in the blood was supported by the finding that cholesterol synthesis in the white blood cells was inhibited. Since the ratio of CoQ9/CoQ10 varies depending on the experimental conditions, the cells were titrated with substrate and inhibitors, leading to the conclusion that the intracellular isopentenyl-PP pool is a regulator of this ratio. Our present findings indicate that oxidosqualenes may be useful for stimulating both the synthesis and level of CoQ both in vitro and in vivo.


Subject(s)
Cholesterol/analogs & derivatives , Cholesterol/biosynthesis , Hemiterpenes/metabolism , Organophosphorus Compounds/metabolism , Squalene/analogs & derivatives , Ubiquinone/analogs & derivatives , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Etidronic Acid/analogs & derivatives , Etidronic Acid/pharmacology , Hep G2 Cells , Humans , Lovastatin/pharmacology , Male , Mevalonic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Wistar , Risedronic Acid , Squalene/metabolism , Squalene/pharmacology , Tricarboxylic Acids/pharmacology , Ubiquinone/biosynthesis
12.
Antimicrob Agents Chemother ; 59(8): 5084-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26055383

ABSTRACT

Previous studies have shown that fosmidomycin, risedronate, and nerolidol exert antimalarial activity in vitro. We included squalestatin, an inhibitor of the isoprenoid metabolism in Erwinia uredovora, and found that combinations of compounds which act on different targets of the plasmodial isoprenoid pathway possess important supra-additivity effects.


Subject(s)
Antimalarials/pharmacology , Biosynthetic Pathways/drug effects , Plasmodium falciparum/drug effects , Terpenes/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Interactions , Fosfomycin/analogs & derivatives , Fosfomycin/pharmacology , Malaria/drug therapy , Parasitic Sensitivity Tests , Plasmodium falciparum/metabolism , Risedronic Acid/pharmacology , Sesquiterpenes/pharmacology , Tricarboxylic Acids/pharmacology
13.
J Pharmacol Exp Ther ; 355(3): 429-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26427720

ABSTRACT

Cytosolic sulfotransferase 1C2 (SULT1C2) is expressed in the kidney, stomach, and liver of rats; however, the mechanisms regulating expression of this enzyme are not known. We evaluated transcriptional regulation of SULT1C2 by mevalonate (MVA)-derived intermediates in primary cultured rat hepatocytes using several cholesterol synthesis inhibitors. Blocking production of mevalonate with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin (30 µM), reduced SULT1C2 mRNA content by ∼40% whereas the squalene synthase inhibitor squalestatin (SQ1, 0.1 µM), which causes accumulation of nonsterol isoprenoids, increased mRNA content by 4-fold. Treatment with MVA (10 mM) strongly induced SULT1C2 mRNA by 12-fold, and this effect was blocked by inhibiting squalene epoxidase but not by more distal cholesterol inhibitors, indicating the effects of MVA are mediated by postsqualene metabolites. Using rapid amplification of cDNA ends (RACE), we characterized the 5' end of SULT1C2 mRNA and used this information to generate constructs for promoter analysis. SQ1 and MVA increased reporter activity by ∼1.6- and 3-fold, respectively, from a construct beginning 49 base pairs (bp) upstream from the longest 5'-RACE product (-3140:-49). Sequence deletions from this construct revealed a hepatocyte nuclear factor 1 (HNF1) element (-2558), and mutation of this element reduced basal (75%) and MVA-induced (30%) reporter activity and attenuated promoter activation following overexpression of HNF1α or 1ß. However, the effects of SQ1 were localized to a more proximal promoter region (-281:-49). Collectively, our findings demonstrate that cholesterol biosynthetic intermediates influence SULT1C2 expression in rat primary hepatocytes. Further, HNF1 appears to play an important role in mediating basal and MVA-induced SULT1C2 transcription.


Subject(s)
Cholesterol/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/enzymology , Sulfotransferases/biosynthesis , Sulfotransferases/genetics , Animals , Anticholesteremic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Genes, Reporter , Hepatocyte Nuclear Factor 1/genetics , Hepatocyte Nuclear Factor 1/metabolism , Hepatocytes/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , Mevalonic Acid/pharmacology , Primary Cell Culture , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Squalene Monooxygenase/antagonists & inhibitors , Sulfotransferases/drug effects , Transfection , Tricarboxylic Acids/pharmacology
14.
Parasitol Res ; 113(11): 4165-76, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199551

ABSTRACT

Eimeria bovis macromeront formation in bovine endothelial host cells is an energy- and nutrient-demanding process. Obligate intracellular replicating coccidians are generally considered as auxotrophic for cholesterol synthesis and scavenge cholesterol from the host cell by either enhancing the uptake of extracellular cholesterol sources or by upregulating the host cellular de novo biosynthesis. We here focused on the latter mechanism and analyzed the effects of several inhibitors targeting the host cellular mevalonate biosynthesis pathway and cholesterol processing. The following inhibitors were used: lovastatin, squalestatin, CI976 and C75 targeting HMG-CoA reductase, squalene synthase, acyl-CoA:cholesterol acyltransferase, and fatty acid synthase, respectively. In summary, all inhibitors significantly interfered with E. bovis meront formation and merozoite production in a dose-dependent manner. Dose effect responses identified lovastatin as the most effective compound, followed by CI976, C75, and squalestatin, respectively. Overall, merozoite production was inhibited by 99.6, 99.7, 84.6, and 70.2% via lovastatin (1 µM), CI976, C75, and squalestatin (all 5 µM) treatments, respectively. Concerning macromeront formation, both the rate and size of developing meronts were affected by inhibitor treatments. The effects were characterized by developmental arrest and meront degradation. In the case of CI976 treatment, we additionally observed detrimental effects on host cellular lipid droplet formation leading to meront developmental arrest irrespective of the time point of treatment onset. These analyses clearly indicate that successful E. bovis intracellular development strictly depends on the host cellular de novo biosynthesis of cholesterol and on the adequate subsequent processing thereof.


Subject(s)
Cholesterol/biosynthesis , Eimeria/growth & development , Endothelial Cells/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cattle , Cells, Cultured , Endothelial Cells/parasitology , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Fatty Acid Synthases/antagonists & inhibitors , Lovastatin/pharmacology , Merozoites/growth & development , Sterol O-Acyltransferase/antagonists & inhibitors , Tricarboxylic Acids/pharmacology
15.
J Lipid Res ; 54(11): 3052-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966666

ABSTRACT

The beneficial effects of statin therapy in the reduction of cardiovascular pathogenesis, atherosclerosis, and diabetic complications are well known. The receptor for advanced glycation end products (RAGE) plays an important role in the progression of these diseases. In contrast, soluble forms of RAGE act as decoys for RAGE ligands and may prevent the development of RAGE-mediated disorders. Soluble forms of RAGE are either produced by alternative splicing [endogenous secretory RAGE (esRAGE)] or by proteolytic shedding mediated by metalloproteinases [shed RAGE (sRAGE)]. Therefore we analyzed whether statins influence the production of soluble RAGE. Lovastatin treatment of either mouse alveolar epithelial cells endogenously expressing RAGE or HEK cells overexpressing RAGE caused induction of RAGE shedding, but did not influence secretion of esRAGE from HEK cells overexpressing esRAGE. Lovastatin-induced secretion of sRAGE was also evident after restoration of the isoprenylation pathway, demonstrating a correlation of sterol biosynthesis and activation of RAGE shedding. Lovastatin-stimulated induction of RAGE shedding was completely abolished by a metalloproteinase ADAM10 inhibitor. We also demonstrate that statins stimulate RAGE shedding at low physiologically relevant concentrations. Our results show that statins, due to their cholesterol-lowering effects, increase the soluble RAGE level by inducing RAGE shedding, and by doing this, might prevent the development of RAGE-mediated pathogenesis.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/chemistry , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cholesterol/metabolism , Dose-Response Relationship, Drug , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Humans , Lovastatin/pharmacology , Mice , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Solubility , Tricarboxylic Acids/pharmacology , beta-Cyclodextrins/pharmacology
16.
J Biol Chem ; 286(8): 6085-91, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21183681

ABSTRACT

The majority of congenital disorders of glycosylation (CDG) are caused by defects of dolichol (Dol)-linked oligosaccharide assembly, which lead to under-occupancy of N-glycosylation sites. Most mutations encountered in CDG are hypomorphic, thus leaving residual activity to the affected biosynthetic enzymes. We hypothesized that increased cellular levels of Dol-linked substrates might compensate for the low biosynthetic activity and thereby improve the output of protein N-glycosylation in CDG. To this end, we investigated the potential of the squalene synthase inhibitor zaragozic acid A to redirect the flow of the polyisoprene pathway toward Dol by lowering cholesterol biosynthesis. The addition of zaragozic acid A to CDG fibroblasts with a Dol-P-Man synthase defect led to the formation of longer Dol-P species and to increased Dol-P-Man levels. This treatment was shown to decrease the pathologic accumulation of incomplete Dol pyrophosphate-GlcNAc(2)Man(5) in Dol-P-Man synthase-deficient fibroblasts. Zaragozic acid A treatment also decreased the amount of truncated protein N-linked oligosaccharides in these CDG fibroblasts. The increased cellular levels of Dol-P-Man and possibly the decreased cholesterol levels in zaragozic acid A-treated cells also led to increased availability of the glycosylphosphatidylinositol anchor as shown by the elevated cell-surface expression of the CD59 protein. This study shows that manipulation of the cellular Dol pool, as achieved by zaragozic acid A addition, may represent a valuable approach to improve N-linked glycosylation in CDG cells.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Congenital Disorders of Glycosylation/metabolism , Dolichols/metabolism , Enzyme Inhibitors/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Oligosaccharides/biosynthesis , Tricarboxylic Acids/pharmacology , CD59 Antigens/biosynthesis , CD59 Antigens/genetics , Cells, Cultured , Cholesterol/biosynthesis , Cholesterol/genetics , Congenital Disorders of Glycosylation/genetics , Dolichols/genetics , Farnesyl-Diphosphate Farnesyltransferase/genetics , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Gene Expression Regulation/drug effects , Glycosylation/drug effects , Humans , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Oligosaccharides/genetics
17.
Biochim Biophys Acta ; 1811(4): 227-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21237288

ABSTRACT

The isoprenoid biosynthesis pathway provides the cell with a variety of compounds which are involved in multiple cellular processes. Inhibition of this pathway with statins and bisphosphonates is widely applied in the treatment of hypercholesterolemia and metabolic bone disease, respectively. In addition, since isoprenylation of proteins is an important therapeutic target in cancer research there is interest in interfering with isoprenoid biosynthesis, for which new inhibitors to block farnesylation and geranylgeranylation of small GTPases are being developed. We recently developed a sensitive method using UPLC-MS/MS that allows the direct detection and quantification of all intermediates of the mevalonate pathway from MVA to GGPP which can be used to verify the specificity of inhibitors of the isoprenoid biosynthesis pathway. We here investigated the specificity of several inhibitors of the isoprenoid biosynthesis pathway in HepG2 cells, fibroblasts and lymphoblasts. The nitrogen-containing bisphosphonates pamidronate and zoledronate specifically inhibit farnesyl pyrophosphate synthase indicated by the accumulation of IPP/DMAPP. However, zaragozic acid A, a squalene synthase inhibitor, causes an increase of MVA in addition to the expected increase of FPP. Analysis of isoprenoid intermediate profiles after incubation with 6-fluoromevalonate showed a very nonspecific result with an increase in MVA, MVAP, MVAPP and IPP/DMAPP. These results show that inhibitors of a particular enzyme of the isoprenoid biosynthesis pathway can have additional effects on other enzymes of the pathway either direct or indirect through accumulation of isoprenoid intermediates. Our method can be used to test new inhibitors and their effect on overall isoprenoid biosynthesis.


Subject(s)
Chromatography, Liquid , Mevalonic Acid/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry , Terpenes/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Bone Density Conservation Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Farnesyltranstransferase/antagonists & inhibitors , Fibroblasts/metabolism , Hep G2 Cells/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Lymphocytes/metabolism , Mevalonic Acid/analogs & derivatives , Pamidronate , Prenylation , Tricarboxylic Acids/pharmacology , Zoledronic Acid
18.
Neurobiol Dis ; 46(1): 69-77, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245662

ABSTRACT

Activation of group III metabotropic glutamate (mGlu) receptors has been recently highlighted as a potential approach in the treatment of Parkinson's disease (PD). This study evaluates the antiparkinsonian action of systemic administration of the broad-spectrum agonist of group III mGlu receptors, 1-aminocyclopentane-1,3,4-tricarboxylic acid (ACPT-I), and investigates its site of action within the basal ganglia circuitry. Acute injection of ACPT-I reverses haloperidol-induced catalepsy, an index of akinesia in rodents. In a rat model of early PD based on partial bilateral nigrostriatal lesions, chronic (2weeks) administration of ACPT-I is required to efficiently alleviate the akinetic deficit evidenced in a reaction time task. This treatment counteracts the post-lesional increases in the gene expression of cytochrome oxidase subunit I, a metabolic marker of neuronal activity, in the overall subthalamic nucleus and in the lateral motor part of the substantia nigra pars reticulata (SNr) but has no effect in the globus pallidus. Paradoxically, ACPT-I administration in sham animals impairs performance and induces overexpression of cytochrome oxidase subunit I mRNA in the lateral SNr, and has no effect in the subthalamic nucleus or globus pallidus. Altogether, our results provide new evidence for the antiparkinsonian efficiency of group III mGlu receptor agonism, point to the regulation of the overactive subthalamo-nigral connection as a main site of action in an early stage of PD and underline the complex interplay between these receptors and the dopaminergic system to regulate basal ganglia function in control and PD conditions.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Parkinsonian Disorders/drug therapy , Receptors, Metabotropic Glutamate/agonists , Substantia Nigra/drug effects , Subthalamic Nucleus/drug effects , Animals , Cyclopentanes/pharmacology , Disease Models, Animal , Male , Neural Pathways/drug effects , Neural Pathways/metabolism , Parkinsonian Disorders/metabolism , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Stereoisomerism , Substantia Nigra/metabolism , Subthalamic Nucleus/metabolism , Tricarboxylic Acids/pharmacology
19.
J Cell Sci ; 123(Pt 4): 595-605, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20103534

ABSTRACT

Cholesterol and sphingolipids are abundant in neuronal membranes, where they help the organisation of the membrane microdomains involved in major roles such as axonal and dendritic growth, and synapse and spine stability. The aim of this study was to analyse their roles in presynaptic physiology. We first confirmed the presence of proteins of the exocytic machinery (SNARES and Ca(v)2.1 channels) in the lipid microdomains of cultured neurons, and then incubated the neurons with fumonisin B (an inhibitor of sphingolipid synthesis), or with mevastatin or zaragozic acid (two compounds that affect the synthesis of cholesterol by inhibiting HMG-CoA reductase or squalene synthase). The results demonstrate that fumonisin B and zaragozic acid efficiently decrease sphingolipid and cholesterol levels without greatly affecting the viability of neurons or the expression of synaptic proteins. Electron microscopy showed that the morphology and number of synaptic vesicles in the presynaptic boutons of cholesterol-depleted neurons were similar to those observed in control neurons. Zaragozic acid (but not fumonisin B) treatment impaired synaptic vesicle uptake of the lipophilic dye FM1-43 and an antibody directed against the luminal epitope of synaptotagmin-1, effects that depended on the reduction in cholesterol because they were reversed by cholesterol reloading. The time-lapse confocal imaging of neurons transfected with ecliptic SynaptopHluorin showed that cholesterol depletion affects the post-depolarisation increase in fluorescence intensity. Taken together, these findings show that reduced cholesterol levels impair synaptic vesicle exocytosis in cultured neurons.


Subject(s)
Cholesterol/metabolism , Exocytosis/physiology , Synaptic Vesicles/physiology , Animals , Anticholesteremic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium Channels, N-Type/metabolism , Cells, Cultured , Exocytosis/drug effects , Fumonisins/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunoglobulin G/metabolism , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Membrane Microdomains/metabolism , Microscopy, Electron, Transmission , Models, Neurological , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , SNARE Proteins/metabolism , Sphingolipids/metabolism , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Synaptotagmin I/antagonists & inhibitors , Synaptotagmin I/immunology , Synaptotagmin I/metabolism , Tricarboxylic Acids/pharmacology
20.
J Pharmacol Exp Ther ; 342(2): 429-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22570363

ABSTRACT

In rodents 5-hydroxytryptamine type 7 (5-HT(7)) receptor blockade has been shown to be effective in models of depression and to increase the latency to rapid eye movement (REM) sleep and decrease REM duration. In the clinic, the REM sleep reduction observed with many antidepressants may serve as a biomarker. We report here the preclinical and clinical evaluation of a 5-HT(7) receptor antagonist, (3-(4-chlorophenyl)-1,4,5,6,7,8-hexahydro-1-(phenylmethyl)pyrazolo[3,4-d]azepine 2-hydroxy-1,2,3-propanetricarboxylate) (JNJ-18038683). In rodents, JNJ-18038683 increased the latency to REM sleep and decreased REM duration, and this effect was maintained after repeated administration for 7 days. The compound was effective in the mouse tail suspension test. JNJ-18038683 enhanced serotonin transmission, antidepressant-like behavior, and REM sleep suppression induced by citalopram in rodents. In healthy human volunteers JNJ-18038683 prolonged REM latency and reduced REM sleep duration, demonstrating that the effect of 5-HT(7) blockade on REM sleep translated from rodents to humans. Like in rats, JNJ-18038683 enhanced REM sleep suppression induced by citalopram in humans, although a drug-drug interaction could not be ruled out. In a double-blind, active, and placebo-controlled clinical trial in 225 patients suffering from major depressive disorder, neither treatment with pharmacologically active doses of JNJ-18038683 or escitalopram separated from placebo, indicating a failed study lacking assay sensitivity. Post hoc analyses using an enrichment window strategy, where all the efficacy data from sites with an implausible high placebo response [placebo group Montgomery-Åsberg Depression Rating Scale (MADRS) < = 12] and from sites with no placebo response (MADRS > = 28) are removed, there was a clinically meaningful difference between JNJ-18038683 and placebo. Further clinical studies are required to characterize the potential antidepressant efficacy of JNJ-18038683.


Subject(s)
Antidepressive Agents/pharmacology , Azepines/pharmacology , Depressive Disorder, Major/drug therapy , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Sleep, REM/drug effects , Tricarboxylic Acids/pharmacology , Adolescent , Adult , Animals , Antidepressive Agents/therapeutic use , Azepines/therapeutic use , Cell Line, Transformed , Citalopram/pharmacology , Cohort Studies , Cross-Over Studies , Depressive Disorder, Major/metabolism , Double-Blind Method , Female , HEK293 Cells , Hindlimb Suspension/methods , Humans , Hypothermia/drug therapy , Hypothermia/metabolism , Hypothermia/physiopathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Antagonists/therapeutic use , Tricarboxylic Acids/therapeutic use , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL