Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 606
Filter
1.
Biosci Rep ; 41(6)2021 05 28.
Article in English | MEDLINE | ID: mdl-33969376

ABSTRACT

It has been demonstrated that trimethylamine N-oxide (TMAO) serves as a driver of atherosclerosis, suggesting that reduction of TMAO level might be a potent method to prevent the progression of atherosclerosis. Herein, we explored the role of TMAO in the stability of carotid atherosclerotic plaques and disclosed the underlying mechanisms. The unstable carotid artery plaque models were established in C57/BL6 mice. L-carnitine (LCA) and methimazole (MMI) administration were applied to increase and reduce TMAO levels. Hematoxylin and eosin (H&E) staining, Sirius red, Perl's staining, Masson trichrome staining and immunohistochemical staining with CD68 staining were used for histopathology analysis of the carotid artery plaque. M1 and M2 macrophagocyte markers were assessed by RT-PCR to determine the polarization of RAW264.7 cells. MMI administration for 2 weeks significantly decreased the plaque area, increased the thickness of the fibrous cap and reduced the size of the necrotic lipid cores, whereas 5-week of administration of MMI induced intraplate hemorrhage. LCA treatment further deteriorated the carotid atherosclerotic plaque but with no significant difference. In mechanism, we found that TMAO treatment impaired the M2 polarization and efferocytosis of RAW264.7 cells with no obvious effect on the M1 polarization. In conclusion, the present study demonstrated that TMAO reduction enhanced the stability of carotid atherosclerotic plaque through promoting macrophage M2 polarization and efferocytosis.


Subject(s)
Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Enzyme Inhibitors/pharmacology , Macrophages/drug effects , Methimazole/pharmacology , Methylamines/metabolism , Phagocytosis/drug effects , Plaque, Atherosclerotic , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Disease Models, Animal , Down-Regulation , Fibrosis , Humans , Jurkat Cells , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Oxygenases/antagonists & inhibitors , Oxygenases/metabolism , Phenotype , RAW 264.7 Cells
2.
Aging (Albany NY) ; 12(1): 931-944, 2020 01 11.
Article in English | MEDLINE | ID: mdl-31927537

ABSTRACT

Flavin-containing monooxygenase 3 (FMO3) gene expression is often upregulated in long-lived murine models. However, the specific relationship between FMO3 and aging remains unknown. Here, we show that 40% calorie restriction (CR), which is considered to be one of the most robust interventions to delay aging progression, markedly upregulates FMO3. Most importantly, upregulation of hepatocyte FMO3 in murine models prevented or reversed hepatic aging. Accordingly, the upregulation of FMO3 mimicked the effects of CR: reduced serum levels of pro-inflammatory cytokine interleukin-6 and fasting insulin; relief of oxidative stress, with lower hepatic malondialdehyde levels and higher superoxide dismutase activity; reduced serum and hepatic levels of total cholesterol and triglyceride, as well as reduced lipid deposition in the liver; and diminished levels of aging-related markers ß-gal and p16. There were also synergistic effects between FMO3 upregulation and CR. Inhibition of autophagy blocked the anti-aging effects of upregulation of hepatocyte FMO3, including reversing the amelioration of the serum and hepatic parameters related to inflammation, oxidative stress, lipid metabolism, liver function, and hepatocyte senescence. Our results suggest that the upregulation of FMO3 mimics CR to prevent or reverse hepatic aging by promoting autophagy.


Subject(s)
Aging/genetics , Aging/metabolism , Autophagy/genetics , Caloric Restriction , Gene Expression Regulation , Liver/metabolism , Oxygenases/genetics , Biomarkers , Gene Expression , Hepatocytes/metabolism , Humans , Immunohistochemistry , Male , Oxidative Stress , Oxygenases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
3.
J Chem Ecol ; 46(2): 217-231, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31879865

ABSTRACT

Despite active research, antiherbivore activity of specific plant phenolics remains largely unresolved. We constructed silver birch (Betula pendula) lines with modified phenolic metabolism to study the effects of foliar flavonoids and condensed tannins on consumption and growth of larvae of a generalist herbivore, the autumnal moth (Epirrita autumnata). We conducted a feeding experiment using birch lines in which expression of dihydroflavonol reductase (DFR), anthocyanidin synthase (ANS) or anthocyanidin reductase (ANR) had been decreased by RNA interference. Modification-specific effects on plant phenolics, nutrients and phenotype, and on larval consumption and growth were analyzed using uni- and multivariate methods. Inhibiting DFR expression increased the concentration of flavonoids at the expense of condensed tannins, and silencing DFR and ANR decreased leaf and plant size. E. autumnata larvae consumed on average 82% less of DFRi plants than of unmodified controls, suggesting that flavonoids or glandular trichomes deter larval feeding. However, larval growth efficiency was highest on low-tannin DFRi plants, indicating that condensed tannins (or their monomers) are physiologically more harmful than non-tannin flavonoids for E. autumnata larvae. Our results show that genetic manipulation of the flavonoid pathway in plants can effectively be used to produce altered phenolic profiles required for elucidating the roles of low-molecular weight phenolics and condensed tannins in plant-herbivore relationships, and suggest that phenolic secondary metabolites participate in regulation of plant growth.


Subject(s)
Betula/chemistry , Flavonoids/metabolism , Moths/physiology , Plants, Genetically Modified/chemistry , Tannins/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Betula/enzymology , Betula/parasitology , Flavonoids/pharmacology , Herbivory/drug effects , Host-Parasite Interactions , Larva/growth & development , Larva/physiology , Moths/growth & development , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Oxygenases/metabolism , Plant Proteins/antagonists & inhibitors , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified/metabolism , Plants, Genetically Modified/parasitology , RNA Interference , Tannins/pharmacology
4.
J Biol Chem ; 294(52): 19923-19933, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31732559

ABSTRACT

Plant development is regulated by both synergistic and antagonistic interactions of different phytohormones, including a complex crosstalk between ethylene and auxin. For instance, auxin and ethylene synergistically control primary root elongation and root hair formation. However, a lack of chemical agents that specifically modulate ethylene or auxin production has precluded precise delineation of the contribution of each hormone to root development. Here, we performed a chemical genetic screen based on the recovery of root growth in ethylene-related Arabidopsis mutants with constitutive "short root" phenotypes (eto1-2 and ctr1-1). We found that ponalrestat exposure recovers root elongation in these mutants in an ethylene signal-independent manner. Genetic and pharmacological investigations revealed that ponalrestat inhibits the enzymatic activity of the flavin-containing monooxygenase YUCCA, which catalyzes the rate-limiting step of the indole-3-pyruvic acid branch of the auxin biosynthesis pathway. In summary, our findings have identified a YUCCA inhibitor that may be useful as a chemical tool to dissect the distinct steps in auxin biosynthesis and in the regulation of root development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Indoleacetic Acids/metabolism , Oxygenases/metabolism , Phthalazines/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ethylenes/metabolism , Indoles/chemistry , Indoles/metabolism , Molecular Docking Simulation , Mutagenesis , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Phenotype , Phthalazines/metabolism , Phthalazines/pharmacology , Plant Roots/drug effects , Plant Roots/growth & development , Plant Roots/metabolism , Protein Structure, Tertiary , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell Metab ; 30(6): 1141-1151.e5, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31543404

ABSTRACT

The gut-microbe-derived metabolite trimethylamine N-oxide (TMAO) is increased by insulin resistance and associated with several sequelae of metabolic syndrome in humans, including cardiovascular, renal, and neurodegenerative disease. The mechanism by which TMAO promotes disease is unclear. We now reveal the endoplasmic reticulum stress kinase PERK (EIF2AK3) as a receptor for TMAO: TMAO binds to PERK at physiologically relevant concentrations; selectively activates the PERK branch of the unfolded protein response; and induces the transcription factor FoxO1, a key driver of metabolic disease, in a PERK-dependent manner. Furthermore, interventions to reduce TMAO, either by manipulation of the gut microbiota or by inhibition of the TMAO synthesizing enzyme, flavin-containing monooxygenase 3, can reduce PERK activation and FoxO1 levels in the liver. Taken together, these data suggest TMAO and PERK may be central to the pathogenesis of the metabolic syndrome.


Subject(s)
Metabolic Syndrome/metabolism , Methylamines/metabolism , eIF-2 Kinase/metabolism , Animals , Gastrointestinal Microbiome/physiology , HEK293 Cells , Hep G2 Cells , Humans , Indoles/pharmacology , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Oxygenases/antagonists & inhibitors
6.
Chem Rec ; 18(12): 1760-1781, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30151867

ABSTRACT

The 2-oxoglutarate (2OG) dependent oxygenases were first identified as having roles in the post-translational modification of procollagen in animals. Subsequently in plants and microbes, they were shown to have roles in the biosynthesis of many secondary metabolites, including signalling molecules and the penicillin/cephalosporin antibiotics. Crystallographic studies of microbial 2OG oxygenases and related enzymes, coupled to DNA sequence analyses, led to the prediction that 2OG oxygenases are widely distributed in aerobic biology. This personal account begins with examples of the roles of 2OG oxygenases in antibiotic biosynthesis, and then describes efforts to assign functions to other predicted 2OG oxygenases. In humans, 2OG oxygenases have been found to have roles in small molecule metabolism, as well as in the epigenetic regulation of protein and nucleic acid biosynthesis and function. The roles and functions of human 2OG oxygenases are compared, focussing on discussion of their substrate and product selectivities. The account aims to emphasize how scoping the substrate selectivity of, sometimes promiscuous, enzymes can provide insights into their functions and so enable therapeutic work.


Subject(s)
Oxygenases/metabolism , Animals , Epigenomics , Histone Demethylases/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Ketoglutaric Acids/chemistry , Ketoglutaric Acids/metabolism , Oxygenases/antagonists & inhibitors , Oxygenases/chemistry , Prolyl Hydroxylases/chemistry , Prolyl Hydroxylases/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational
7.
J Thromb Haemost ; 16(9): 1857-1872, 2018 09.
Article in English | MEDLINE | ID: mdl-29981269

ABSTRACT

Essentials Microbe-dependent production of trimethylamine N-oxide (TMAO) contributes to thrombosis risk. The impact of host flavin monooxygenase 3 (FMO3) modulation on platelet function is unknown. Genetic manipulation of FMO3 in mice alters systemic TMAO levels and thrombosis potential. Genetic manipulation of FMO3 is associated with alteration of gut microbial community structure. SUMMARY: Background Gut microbes play a critical role in the production of trimethylamine N-oxide (TMAO), an atherogenic metabolite that impacts platelet responsiveness and thrombosis potential. Involving both microbe and host enzymatic machinery, TMAO generation utilizes a metaorganismal pathway, beginning with ingestion of trimethylamine (TMA)-containing dietary nutrients such as choline, phosphatidylcholine and carnitine, which are abundant in a Western diet. Gut microbial TMA lyases use these nutrients as substrates to produce TMA, which upon delivery to the liver via the portal circulation, is converted into TMAO by host hepatic flavin monooxygenases (FMOs). Gut microbial production of TMA is rate limiting in the metaorganismal TMAO pathway because hepatic FMO activity is typically in excess. Objectives FMO3 is the major FMO responsible for host generation of TMAO; however, a role for FMO3 in altering platelet responsiveness and thrombosis potential in vivo has not yet been explored. Methods The impact of FMO3 suppression (antisense oligonucleotide-targeting) and overexpression (as transgene) on plasma TMAO levels, platelet responsiveness and thrombosis potential was examined using a murine FeCl3 -induced carotid artery injury model. Cecal microbial composition was examined using 16S analyses. Results Modulation of FMO3 directly impacts systemic TMAO levels, platelet responsiveness and rate of thrombus formation in vivo. Microbial composition analyses reveal taxa whose proportions are associated with both plasma TMAO levels and in vivo thrombosis potential. Conclusions The present studies demonstrate that host hepatic FMO3, the terminal step in the metaorganismal TMAO pathway, participates in diet-dependent and gut microbiota-dependent changes in both platelet responsiveness and thrombosis potential in vivo.


Subject(s)
Blood Platelets/physiology , Gastrointestinal Microbiome/physiology , Liver/enzymology , Methylamines/metabolism , Oxygenases/physiology , Thrombophilia/enzymology , Animals , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/chemically induced , Carotid Artery, Common , Chlorides/toxicity , Ferric Compounds/toxicity , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/pharmacology , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Platelet-Rich Plasma , Ribotyping , Risk , Thrombophilia/microbiology , Transgenes
8.
Int J Biol Macromol ; 118(Pt A): 460-468, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-29959003

ABSTRACT

Human flavin-containing monooxygenase isoform 3 (hFMO3) is an important hepatic drug-metabolizing enzyme, catalyzing the monooxygenation of nucleophilic heteroatom-containing xenobiotics. Based on the structure of bacterial FMO, it is proposed that a conserved asparagine is involved in both NADP(H) and substrate binding. In order to explore the role of this amino acid in hFMO3, two mutants were constructed. In the case of N61Q, increasing the steric hindrance above the flavin N5-C4a causes poor NADP(H) binding, destabilizing the catalytic FAD intermediate, whereas the introduction of a negatively charged residue, N61D, interferes mainly with catalytic intermediate formation and its stability. To better understand the substrate-enzyme interaction, in vitro as well as in silico experiments were carried out with methimazole as substrate. Methimazole is a high-affinity substrate of hFMO3 and can competitively suppress the metabolism of other compounds. Our results demonstrate that methimazole Pi-stacks above the isoalloxazine ring of FAD in hFMO3, in a similar way to indole binding to the bacterial FMO. However, for hFMO3 indole is found to act as a non-substrate competitive inhibitor. Finally, understanding the binding mode of methimazole and indole could be advantageous for development of hFMO3 inhibitors, currently investigated as a possible treatment strategy for atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , NADP/chemistry , Oxygenases/chemistry , Protein Binding , Amino Acids/chemistry , Atherosclerosis/genetics , Catalysis , Computer Simulation , Flavins/chemistry , Flavins/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Methimazole/chemistry , Methimazole/pharmacology , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Substrate Specificity
9.
PLoS One ; 12(11): e0187294, 2017.
Article in English | MEDLINE | ID: mdl-29121650

ABSTRACT

Busulphan (Bu) is an alkylating agent used in the conditioning regimen prior to hematopoietic stem cell transplantation (HSCT). Bu is extensively metabolized in the liver via conjugations with glutathione to form the intermediate metabolite (sulfonium ion) which subsequently is degraded to tetrahydrothiophene (THT). THT was reported to be oxidized forming THT-1-oxide that is further oxidized to sulfolane and finally 3-hydroxysulfolane. However, the underlying mechanisms for the formation of these metabolites remain poorly understood. In the present study, we performed in vitro and in vivo investigations to elucidate the involvement of flavin-containing monooxygenase-3 (FMO3) and cytochrome P450 enzymes (CYPs) in Bu metabolic pathway. Rapid clearance of THT was observed when incubated with human liver microsomes. Furthermore, among different recombinant microsomal enzymes, the highest intrinsic clearance for THT was obtained via FMO3 followed by several CYPs including 2B6, 2C8, 2C9, 2C19, 2E1 and 3A4. In Bu- or THT-treated mice, inhibition of FMO3 by phenylthiourea significantly suppressed the clearance of both Bu and THT. Moreover, the simultaneous administration of a high dose of THT (200µmol/kg) to Bu-treated mice reduced the clearance of Bu. Consistently, in patients undergoing HSCT, repeated administration of Bu resulted in a significant up-regulation of FMO3 and glutathione-S-transfrase -1 (GSTA1) genes. Finally, in a Bu-treated patient, additional treatment with voriconazole (an antimycotic drug known as an FMO3-substrate) significantly altered the Bu clearance. In conclusion, we demonstrate for the first time that FMO3 along with CYPs contribute a major part in busulphan metabolic pathway and certainly can affect its kinetics. The present results have high clinical impact. Furthermore, these findings might be important for reducing the treatment-related toxicity of Bu, through avoiding interaction with other concomitant used drugs during conditioning and hence improving the clinical outcomes of HSCT.


Subject(s)
Busulfan/metabolism , Metabolic Networks and Pathways , Oxygenases/metabolism , Adolescent , Adult , Animals , Child , Child, Preschool , Gene Expression Regulation/drug effects , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Kinetics , Male , Metabolic Networks and Pathways/drug effects , Metabolome/drug effects , Mice, Inbred C57BL , Microsomes/enzymology , Middle Aged , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity/drug effects , Thiophenes/metabolism , Time Factors , Transplantation Conditioning , Voriconazole/pharmacology
10.
Drug Metab Lett ; 10(4): 295-305, 2017.
Article in English | MEDLINE | ID: mdl-28137210

ABSTRACT

OBJECTIVE: Drug therapy is occasionally accompanied by an idiosyncratic severe toxicity, which occurs very rarely, but can lead to patient mortality. Methazolamide, an anti-glaucomatous agent, could cause severe skin eruptions called Stevens-Johnson syndrome/toxic epidermal necrolyis (SJS/TEN). Its precise etiology is still uncertain. In this study, the metabolism of methazolamide was investigated in immortalized human keratinocytes to reveal the possible mechanism which causes SJS/TEN. METHODS: The metabolism of methazolamide was studied using immortalized human keratinocytes, HaCaT cells. HPLC was used to isolate a metabolite from the culture medium. Mass spectrometry (LCMS/ MS) was employed for its characterization. Three typical chemical inducers were assessed for the inducibility of cytochrome P450, and methimazole was used as the inhibitor of flavin-containing monooxygenase (FMO). RESULTS: A sulfonic acid, N-[3-methyl-5-sulfo-1,3,4-thiadiazol-2(3H)-ylidene]acetamide (MSO) was identified as the final metabolite. Dexamethasone and ß-naphthoflavone behaved as an inducer of cytochrome P450 in the metabolism, but isoniazid did not. The effect of methimazole was not consistent. We did not detect any glucuronide nor any mercapturic acid (N-acetylcysteine conjugate). CONCLUSION: N-[3-methyl-5-sulfo-1,3,4-thiadiazol-2(3H)-ylidene]acetamide (MSO) is not considered to be a direct product of an enzymatic reaction, but rather an auto-oxidation product of N-[3-methyl-5- sulfe-1,3,4-thiadiazol-2(3H)-ylidene]acetamide, a chemically unstable sulfenic acid, which is produced by cytochrome P450 from the ß-lyase product of cysteine conjugate of methazolamide. MSO is considered to be susceptible to glutathione and to return to glutathione conjugate of methazolamide, forming a futile cycle. A hypothetical scenario is presented as to the onset of the disease.


Subject(s)
Carbonic Anhydrase Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Methazolamide/metabolism , Stevens-Johnson Syndrome/etiology , Sulfonic Acids/toxicity , Acetylcysteine/metabolism , Carbonic Anhydrase Inhibitors/therapeutic use , Carbonic Anhydrase Inhibitors/toxicity , Cell Line , Chromatography, High Pressure Liquid/methods , Cysteine/metabolism , Dexamethasone/pharmacology , Glaucoma/drug therapy , Glucuronides/metabolism , Humans , Isoniazid/pharmacology , Keratinocytes , Lyases/metabolism , Methazolamide/therapeutic use , Methazolamide/toxicity , Methimazole/pharmacology , Oxidation-Reduction , Oxygenases/antagonists & inhibitors , Sulfenic Acids/metabolism , Sulfonic Acids/metabolism , Tandem Mass Spectrometry/methods , beta-Naphthoflavone/pharmacology
11.
PLoS One ; 12(1): e0169910, 2017.
Article in English | MEDLINE | ID: mdl-28072829

ABSTRACT

As an important enzyme in Phase I drug metabolism, the flavin-containing monooxygenase (FMO) also metabolizes some xenobiotics with soft nucleophiles. The site of metabolism (SOM) on a molecule is the site where the metabolic reaction is exerted by an enzyme. Accurate prediction of SOMs on drug molecules will assist the search for drug leads during the optimization process. Here, some quantum mechanics features such as the condensed Fukui function and attributes from circular fingerprints (called Molprint2D) are computed and classified using the support vector machine (SVM) for predicting some potential SOMs on a series of drugs that can be metabolized by FMO enzymes. The condensed Fukui function fA- representing the nucleophilicity of central atom A and the attributes from circular fingerprints accounting the influence of neighbors on the central atom. The total number of FMO substrates and non-substrates collected in the study is 85 and they are equally divided into the training and test sets with each carrying roughly the same number of potential SOMs. However, only N-oxidation and S-oxidation features were considered in the prediction since the available C-oxidation data was scarce. In the training process, the LibSVM package of WEKA package and the option of 10-fold cross validation are employed. The prediction performance on the test set evaluated by accuracy, Matthews correlation coefficient and area under ROC curve computed are 0.829, 0.659, and 0.877 respectively. This work reveals that the SVM model built can accurately predict the potential SOMs for drug molecules that are metabolizable by the FMO enzymes.


Subject(s)
Enzyme Inhibitors/chemistry , Oxygenases/antagonists & inhibitors , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , Humans , Oxygenases/chemistry , Oxygenases/metabolism , Protein Binding , Quantitative Structure-Activity Relationship , Support Vector Machine
12.
Biochem Pharmacol ; 116: 153-61, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27422753

ABSTRACT

Ketoconazole (KC), an antifungal agent, rarely causes severe liver injury when orally administered. It has been reported that KC is mainly hydrolyzed to N-deacetyl ketoconazole (DAK), followed by the N-hydroxylation of DAK by flavin-containing monooxygenase (FMO). Although the metabolism of KC has been considered to be associated with hepatotoxicity, the responsible enzyme(s) remain unknown. The purpose of this study was to identify the responsible enzyme(s) for KC hydrolysis in humans and to clarify their relevance to KC-induced toxicity. Kinetic analysis and inhibition studies using human liver microsomes (HLM) and recombinant enzymes revealed that human arylacetamide deacetylase (AADAC) is responsible for KC hydrolysis to form DAK, and confirmed that FMO3 is the enzyme responsible for DAK N-hydroxylation. In HLM, the clearance of KC hydrolysis occurred to the same extent as DAK N-hydroxylation, which indicates that both processes are not rate-limiting pathways. Cytotoxicity of KC and DAK was evaluated using HepaRG cells and human primary hepatocytes. Treatment of HepaRG cells with DAK for 24h showed cytotoxicity in a dose-dependent manner, whereas treatment with KC did not show due to the low expression of AADAC. Overexpression of AADAC in HepaRG cells with an adenovirus expression system elicited the cytotoxicity of KC. Cytotoxicity of KC in human primary hepatocytes was attenuated by diisopropylfluorophosphate, an AADAC inhibitor. In conclusion, the present study demonstrated that human AADAC hydrolyzes KC to trigger hepatocellular toxicity.


Subject(s)
Antifungal Agents/metabolism , Carboxylic Ester Hydrolases/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Cytochrome P-450 CYP3A Inhibitors/metabolism , Hepatocytes/metabolism , Ketoconazole/metabolism , Microsomes, Liver/enzymology , Activation, Metabolic/drug effects , Antifungal Agents/adverse effects , Biocatalysis/drug effects , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Cell Line, Tumor , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hydrolysis/drug effects , Hydroxylation/drug effects , Isoflurophate/pharmacology , Ketoconazole/adverse effects , Ketoconazole/analogs & derivatives , Ketoconazole/toxicity , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Middle Aged , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Oxygenases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
13.
Toxicol Lett ; 258: 55-70, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27320963

ABSTRACT

Little is known about the role of flavin-containing monooxygenases (FMOs) in the metabolism of xenobiotics. FMO3 is the isoform in adult human liver with the highest impact on drug metabolism. The aim of the presented study was to elucidate the contribution of human FMO3 to the N-oxygenation of selected therapeutic drugs and drugs of abuse (DOAs). Its contribution to the in vivo hepatic net clearance of the N-oxygenation products was calculated by application of an extended relative activity factor (RAF) approach to differentiate from contribution of cytochrome P450 (CYP) isoforms. FMO3 and CYP substrates were identified using pooled human liver microsomes after heat inactivation and chemical inhibition, or single enzyme incubations. Kinetic parameters were subsequently determined using recombinant human enzymes and mass spectrometric analysis via authentic reference standards or simple peak areas of the products divided by those of the internal standard. FMO3 was identified as enzyme mainly responsible for the formation of N,N-diallyltryptamine N-oxide and methamphetamine hydroxylamine (>80% contribution for both). A contribution of 50 and 30% was calculated for the formation of N,N-dimethyltryptamine N-oxide and methoxypiperamide N-oxide, respectively. However, FMO3 contributed with less than 5% to the formation of 3-bromomethcathinone hydroxylamine, amitriptyline N-oxide, and clozapine N-oxide. There was no significant difference in the contributions when using calibrations with reference metabolite standards or peak area ratio calculations. The successful application of a modified RAF approach including FMO3 proved the importance of FMO3 in the N-oxygenation of DOAs in human metabolism.


Subject(s)
Antitussive Agents/metabolism , Central Nervous System Agents/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Models, Biological , Oxygenases/metabolism , Animals , Antitussive Agents/chemistry , Biotransformation/drug effects , Calibration , Cell Line , Central Nervous System Agents/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Enzyme Inhibitors/pharmacology , Hot Temperature , Humans , Insecta , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oxidation-Reduction , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substance-Related Disorders/enzymology , Substance-Related Disorders/etiology , Substance-Related Disorders/metabolism
14.
Drug Metab Dispos ; 44(8): 1262-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27079250

ABSTRACT

N1-Substituted-6-arylthiouracils, represented by compound 1 [6-(2,4-dimethoxyphenyl)-1-(2-hydroxyethyl)-2-thioxo-2,3-dihydropyrimidin-4(1H)-one], are a novel class of selective irreversible inhibitors of human myeloperoxidase. The present account is a summary of our in vitro studies on the facile oxidative desulfurization in compound 1 to a cyclic ether metabolite M1 [5-(2,4-dimethoxyphenyl)-2,3-dihydro-7H-oxazolo[3,2-a]pyrimidin-7-one] in NADPH-supplemented rats (t1/2 [half-life = mean ± S.D.] = 8.6 ± 0.4 minutes) and dog liver microsomes (t1/2 = 11.2 ± 0.4 minutes), but not in human liver microsomes (t1/2 > 120 minutes). The in vitro metabolic instability also manifested in moderate-to-high plasma clearances of the parent compound in rats and dogs with significant concentrations of M1 detected in circulation. Mild heat deactivation of liver microsomes or coincubation with the flavin-containing monooxygenase (FMO) inhibitor imipramine significantly diminished M1 formation. In contrast, oxidative metabolism of compound 1 to M1 was not inhibited by the pan cytochrome P450 inactivator 1-aminobenzotriazole. Incubations with recombinant FMO isoforms (FMO1, FMO3, and FMO5) revealed that FMO1 principally catalyzed the conversion of compound 1 to M1. FMO1 is not expressed in adult human liver, which rationalizes the species difference in oxidative desulfurization. Oxidation by FMO1 followed Michaelis-Menten kinetics with Michaelis-Menten constant, maximum rate of oxidative desulfurization, and intrinsic clearance values of 209 µM, 20.4 nmol/min/mg protein, and 82.7 µl/min/mg protein, respectively. Addition of excess glutathione essentially eliminated the conversion of compound 1 to M1 in NADPH-supplemented rat and dog liver microsomes, which suggests that the initial FMO1-mediated S-oxygenation of compound 1 yields a sulfenic acid intermediate capable of redox cycling to the parent compound in a glutathione-dependent fashion or undergoing further oxidation to a more electrophilic sulfinic acid species that is trapped intramolecularly by the pendant alcohol motif in compound 1.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Liver/enzymology , Oxygenases/metabolism , Peroxidase/antagonists & inhibitors , Thiouracil/pharmacokinetics , Administration, Intravenous , Animals , Biotransformation , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Half-Life , Humans , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Models, Biological , Oxidation-Reduction , Oxygenases/antagonists & inhibitors , Peroxidase/metabolism , Rats, Wistar , Species Specificity , Thiouracil/administration & dosage , Thiouracil/analogs & derivatives , Thiouracil/blood
15.
J Biol Chem ; 291(3): 1348-67, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26578517

ABSTRACT

The kidney is one of the target organs for various metabolic diseases, including diabetes, metabolic syndrome, and obesity. Most of the metabolic studies underscore glomerular pathobiology, although the tubulo-interstitial compartment has been underemphasized. This study highlights mechanisms concerning the pathobiology of tubular injury in the context of myo-inositol oxygenase (Miox), a tubular enzyme. The kidneys of mice fed a high fat diet (HFD) had increased Miox expression and activity, and the latter was related to phosphorylation of serine/threonine residues. Also, expression of sterol regulatory element-binding protein1 (Srebp1) and markers of cellular/nuclear damage was increased along with accentuated apoptosis and loss of tubular brush border. Similar results were observed in cells treated with palmitate/BSA. Multiple sterol-response elements and E-box motifs were found in the miox promoter, and its activity was modulated by palmitate/BSA. Electrophoretic mobility and ChIP assays confirmed binding of Srebp to consensus sequences of the miox promoter. Exposure of palmitate/BSA-treated cells to rapamycin normalized Miox expression and prevented Srebp1 nuclear translocation. In addition, rapamycin treatment reduced p53 expression and apoptosis. Like rapamycin, srebp siRNA reduced Miox expression. Increased expression of Miox was associated with the generation of reactive oxygen species (ROS) in kidney tubules of mice fed an HFD and cell exposed to palmitate/BSA. Both miox and srebp1 siRNAs reduced generation of ROS. Collectively, these findings suggest that HFD or fatty acids modulate transcriptional, translational, and post-translational regulation of Miox expression/activity and underscore Miox being a novel target of the transcription factor Srebp1. Conceivably, activation of the mTORC1/Srebp1/Miox pathway leads to the generation of ROS culminating into tubulo-interstitial injury in states of obesity.


Subject(s)
Diabetic Nephropathies/metabolism , Inositol Oxygenase/metabolism , Kidney Tubules/enzymology , Obesity/metabolism , Oxidative Stress , Protein Processing, Post-Translational , Up-Regulation , Animals , Apoptosis , Cell Line , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Diet, High-Fat/adverse effects , Humans , Inositol Oxygenase/antagonists & inhibitors , Inositol Oxygenase/genetics , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Obesity/etiology , Obesity/pathology , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Oxygenases/metabolism , Phosphorylation , Promoter Regions, Genetic , Proteins/antagonists & inhibitors , Proteins/genetics , Proteins/metabolism , RNA Interference , Rats , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sus scrofa
16.
Chem Commun (Camb) ; 51(84): 15458-61, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26345662

ABSTRACT

There is interest in developing potent, selective, and cell-permeable inhibitors of human ferrous iron and 2-oxoglutarate (2OG) oxygenases for use in functional and target validation studies. The 3-component Betti reaction enables efficient one-step C-7 functionalisation of modified 8-hydroxyquinolines (8HQs) to produce cell-active inhibitors of KDM4 histone demethylases and other 2OG oxygenases; the work exemplifies how a template-based metallo-enzyme inhibitor approach can be used to give biologically active compounds.


Subject(s)
Enzyme Inhibitors/pharmacology , Oxygenases/antagonists & inhibitors , Oxyquinoline/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Oxygenases/metabolism , Oxyquinoline/chemical synthesis , Oxyquinoline/chemistry , Structure-Activity Relationship
17.
Plant J ; 84(4): 827-37, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26402640

ABSTRACT

Auxin is essential for plant growth and development, this makes it difficult to study the biological function of auxin using auxin-deficient mutants. Chemical genetics have the potential to overcome this difficulty by temporally reducing the auxin function using inhibitors. Recently, the indole-3-pyruvate (IPyA) pathway was suggested to be a major biosynthesis pathway in Arabidopsis thaliana L. for indole-3-acetic acid (IAA), the most common member of the auxin family. In this pathway, YUCCA, a flavin-containing monooxygenase (YUC), catalyzes the last step of conversion from IPyA to IAA. In this study, we screened effective inhibitors, 4-biphenylboronic acid (BBo) and 4-phenoxyphenylboronic acid (PPBo), which target YUC. These compounds inhibited the activity of recombinant YUC in vitro, reduced endogenous IAA content, and inhibited primary root elongation and lateral root formation in wild-type Arabidopsis seedlings. Co-treatment with IAA reduced the inhibitory effects. Kinetic studies of BBo and PPBo showed that they are competitive inhibitors of the substrate IPyA. Inhibition constants (Ki ) of BBo and PPBo were 67 and 56 nm, respectively. In addition, PPBo did not interfere with the auxin response of auxin-marker genes when it was co-treated with IAA, suggesting that PPBo is not an inhibitor of auxin sensing or signaling. We propose that these compounds are a class of auxin biosynthesis inhibitors that target YUC. These small molecules are powerful tools for the chemical genetic analysis of auxin function.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oxygenases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Boronic Acids/chemistry , Boronic Acids/pharmacology , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Plant/drug effects , Indoleacetic Acids/metabolism , Indoles/metabolism , Indoles/pharmacology , Molecular Structure , Mutation , Oxygenases/genetics , Oxygenases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Seedlings/genetics , Seedlings/metabolism , Small Molecule Libraries/chemistry
18.
Phytochemistry ; 117: 456-461, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26196940

ABSTRACT

2-Oxoglutarate (2OG) and ferrous iron dependent oxygenases are involved in many biological processes in organisms ranging from humans (where some are therapeutic targets) to plants. These enzymes are of significant biomedicinal interest because of their roles in hypoxic signaling and epigenetic regulation. Synthetic N-oxalylglycine (NOG) has been identified as a broad-spectrum 2OG oxygenase inhibitor and is currently widely used in studies on the hypoxic response and chromatin modifications in animals. We report the identification of NOG as a natural product present in Rheum rhabarbarum (rhubarb) and Spinach oleracea (spinach) leaves; NOG was not observed in Escherchia coli or human embryonic kidney cells (HEK 293T). The finding presents the possibility that NOG plays a natural role in regulating gene expression by inhibiting 2OG dependent oxygenases. This has significance because tricarboxylic acid cycle (TCA) intermediate inhibition of 2OG dependent oxygenases has attracted major interest in cancer research.


Subject(s)
Amino Acids, Dicarboxylic/isolation & purification , Plant Leaves/chemistry , Rheum/chemistry , Spinacia oleracea/chemistry , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/isolation & purification , Amino Acids, Dicarboxylic/chemistry , Amino Acids, Dicarboxylic/pharmacology , Brassica/chemistry , Chromatography, Liquid/methods , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Escherichia coli/chemistry , HEK293 Cells/chemistry , Humans , Ketoglutaric Acids/metabolism , Magnetic Resonance Spectroscopy , Oxygenases/antagonists & inhibitors , Tandem Mass Spectrometry
19.
J Biol Inorg Chem ; 20(5): 757-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25911498

ABSTRACT

Staphylococcus aureus IsdG catalyzes the final step of staphylococcal iron acquisition from host hemoglobin, whereby host-derived heme is converted to iron and organic products. The Asn7 distal pocket residue is known to be critical for enzyme activity, but the influence of this residue on the substrate electronic structure was unknown prior to this work. Here, an optical spectroscopic and density functional theory characterization of azide- and cyanide-inhibited wild type and N7A IsdG is presented. Magnetic circular dichroism data demonstrate that Asn7 perturbs the electronic structure of azide-inhibited, but not cyanide-inhibited, IsdG. As the iron-ligating α-atom of azide, but not cyanide, can act as a hydrogen bond acceptor, these data indicate that the terminal amide of Asn7 is a hydrogen bond donor to the α-atom of a distal ligand to heme in IsdG. Circular dichroism characterization of azide- and cyanide-inhibited forms of WT and N7A IsdG strongly suggests that the Asn7···N3 hydrogen bond influences the orientation of a distal azide ligand with respect to the heme substrate. Specifically, density functional theory calculations suggest that Asn7···N3 hydrogen bond donation causes the azide ligand to rotate about an axis perpendicular to the porphyrin plane and weakens the π-donor strength of the azide ligand. This lowers the energies of the Fe 3d xz and 3d yz orbitals, mixes Fe 3d xy and porphyrin a 2u character into the singly-occupied molecular orbital, and results in spin delocalization onto the heme meso carbons. These discoveries have important implications for the mechanism of heme oxygenation catalyzed by IsdG.


Subject(s)
Azides/pharmacology , Cyanides/pharmacology , Enzyme Inhibitors/pharmacology , Heme/chemistry , Oxygenases/antagonists & inhibitors , Azides/chemistry , Cyanides/chemistry , Dose-Response Relationship, Drug , Electrons , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Ligands , Molecular Structure , Oxygenases/chemistry , Oxygenases/metabolism , Quantum Theory , Staphylococcus aureus , Structure-Activity Relationship
20.
Int J Mol Sci ; 15(11): 20900-12, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25402648

ABSTRACT

Chloroanilines are widely used in the manufacture of drugs, pesticides and industrial intermediates. Among the trichloroanilines, 3,4,5-trichloroaniline (TCA) is the most potent nephrotoxicant in vivo. The purpose of this study was to examine the nephrotoxic potential of TCA in vitro and to determine if renal biotransformation and/or free radicals contributed to TCA cytotoxicity using isolated renal cortical cells (IRCC) from male Fischer 344 rats as the animal model. IRCC (~4 million cells/mL; 3 mL) were incubated with TCA (0, 0.1, 0.25, 0.5 or 1.0 mM) for 60-120 min. In some experiments, IRCC were pretreated with an antioxidant or a cytochrome P450 (CYP), flavin monooxygenase (FMO), cyclooxygenase or peroxidase inhibitor prior to incubation with dimethyl sulfoxide (control) or TCA (0.5 mM) for 120 min. At 60 min, TCA did not induce cytotoxicity, but induced cytotoxicity as early as 90 min with 0.5 mM or higher TCA and at 120 min with 0.1 mM or higher TCA, as evidenced by increased lactate dehydrogenase (LDH) release. Pretreatment with the CYP inhibitor piperonyl butoxide, the cyclooxygenase inhibitor indomethacin or the peroxidase inhibitor mercaptosuccinate attenuated TCA cytotoxicity, while pretreatment with FMO inhibitors or the CYP inhibitor metyrapone had no effect on TCA nephrotoxicity. Pretreatment with an antioxidant (α-tocopherol, glutathione, ascorbate or N-acetyl-L-cysteine) also reduced or completely blocked TCA cytotoxicity. These results indicate that TCA is directly nephrotoxic to IRCC in a time and concentration dependent manner. Bioactivation of TCA to toxic metabolites by CYP, cyclooxygenase and/or peroxidase contributes to the mechanism of TCA nephrotoxicity. Lastly, free radicals play a role in TCA cytotoxicity, although the exact nature of the origin of these radicals remains to be determined.


Subject(s)
Aniline Compounds/toxicity , Cytotoxins/toxicity , Free Radicals/metabolism , Kidney/drug effects , Aniline Compounds/metabolism , Animals , Antioxidants/pharmacology , Biotransformation , Cells, Cultured , Cyclooxygenase Inhibitors/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Cytotoxins/metabolism , Kidney/cytology , Kidney/metabolism , Male , Oxygenases/antagonists & inhibitors , Oxygenases/metabolism , Peroxidases/antagonists & inhibitors , Peroxidases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...