Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Metab Dispos ; 49(3): 202-211, 2021 03.
Article in English | MEDLINE | ID: mdl-33355213

ABSTRACT

All-trans-retinoic acid (atRA) is a critical endogenous signaling molecule. atRA is predominantly synthesized from retinaldehyde by aldehyde dehydrogenase 1A1 (ALDH1A1), but aldehyde oxidase (AOX) may also contribute to atRA biosynthesis. The goal of this study was to test the hypothesis that AOX contributes significantly to atRA formation in human liver. Human recombinant AOX formed atRA from retinaldehyde (Km ∼1.5 ± 0.4 µM; kcat ∼3.6 ± 2.0 minute-1). In human liver S9 fractions (HLS9), atRA formation was observed in the absence of NAD+, suggesting AOX contribution to atRA formation. In the presence of NAD+, Eadie-Hofstee plots of atRA formation in HLS9 indicated that two enzymes contributed to atRA formation. The two enzymes were identified as AOX and ALDH1A1 based on inhibition of atRA formation by AOX inhibitor hydralazine (20%-50% inhibition) and ALDH1A1 inhibitor WIN18,446 (50%-80%inhibition). The expression of AOX in HLS9 was 9.4-24 pmol mg-1 S9 protein, whereas ALDH1A1 expression was 156-285 pmol mg-1 S9 protein measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) quantification of signature peptides. The formation velocity of atRA in the presence of NAD+ correlated significantly with the expression of ALDH1A1 and AOX protein. Taken together, the data show that both AOX and ALDH1A1 contribute to atRA biosynthesis in the human liver, with ALDH1A1 being the high-affinity, low-capacity enzyme and AOX being the low-affinity, high-capacity enzyme. The results suggest that in the case of ALDH1A dysfunction or excess vitamin A, AOX may play an important role in regulating hepatic vitamin A homeostasis and that inhibition of AOX may alter atRA biosynthesis and signaling. SIGNIFICANCE STATEMENT: This study provides direct evidence to show that human AOX converts retinaldehyde to atRA and contributes to hepatic atRA biosynthesis. The finding that AOX may be responsible for 20%-50% of overall hepatic atRA formation suggests that alterations in AOX activity via drug-drug interactions, genetic polymorphisms, or disease states may impact hepatic atRA concentrations and signaling and alter vitamin A homeostasis.


Subject(s)
Aldehyde Oxidase/biosynthesis , Liver/metabolism , Tretinoin/metabolism , Adult , Amino Acid Sequence , Animals , Child , Dose-Response Relationship, Drug , Female , Humans , Liver/drug effects , Male , Mice , Middle Aged , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology
2.
Cell Tissue Res ; 379(1): 45-62, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31858241

ABSTRACT

Lipid droplet (LD) binding proteins in mammary glands and in adipocytes were previously compared and striking similar sets of these specific proteins demonstrated. Xanthine oxidoreductase (XOR) together with perilipins and the lactating mammary gland protein butyrophilin play an important role in the secretion process of LDs into milk ducts. In contrast, in adipose tissue and in adipocytes, mainly perilipins have been described. Moreover, XOR was reported in mouse adipose tissue and adipocyte culture cells as "novel regulator of adipogenesis". This obvious coincidence of protein sets prompted us to revisit the formation of LDs in human-cultured adipocytes in more detail with special emphasis on the possibility of a LD association of XOR. We demonstrate by electron and immunoelectron microscopy new structural details on LD formation in adipocytes. Surprisingly, by immunological and proteomic analysis, we identify in contrast to previous data showing the enzyme XOR, predominantly the expression of aldehyde oxidase (AOX). AOX could be detected tightly linked to LDs when adipocytes were treated with starvation medium. In addition, the majority of cells show an enormous interconnected, tubulated mitochondria network. Here, we discuss that (1) XOR is involved-together with perilipins-in the secretion of LDs in alveolar epithelial cells of the lactating mammary gland and is important in the transcytosis pathway of capillary endothelial cells. (2) In cells, where LDs are not secreted, XOR cannot be detected at the protein level, whereas in contrast in these cases, AOX is often present. We detect AOX in adipocytes together with perilipins and find evidence that these proteins might direct LDs to mitochondria. Finally, we here report for the first time the exclusive and complementary localization of XOR and AOX in diverse cell types.


Subject(s)
Adipocytes/metabolism , Aldehyde Oxidase/biosynthesis , Lipid Droplets/metabolism , Adipocytes/enzymology , Adipocytes/ultrastructure , Animals , Cells, Cultured , Culture Media , Humans , Perilipins/metabolism , Xanthine Dehydrogenase/metabolism
3.
Development ; 145(18)2018 09 17.
Article in English | MEDLINE | ID: mdl-30111657

ABSTRACT

The developing vertebrate embryo is exquisitely sensitive to retinoic acid (RA) concentration, particularly during anteroposterior patterning. In contrast to Nodal and Wnt signaling, RA was not previously considered to be an instructive signal in mesoderm formation during gastrulation. Here, we show in Xenopus that RARγ is indispensable for the expression of early mesoderm markers and is, therefore, an obligatory factor in mesodermal competence and/or maintenance. We identified several novel targets upregulated by RA receptor signaling in the early gastrula that are expressed in the circumblastoporal ring and linked to mesodermal development. Despite overlapping expression patterns of the genes encoding the RA-synthesizing enzyme Aldh1a2 and the RA-degrading enzyme Cyp26a1, RARγ1 functions as a transcriptional activator in early mesoderm development, suggesting that RA ligand is available to the embryo earlier than previously appreciated. RARγ1 is required for cellular adhesion, as revealed by spontaneous dissociation and depletion of ncam1 mRNA in animal caps harvested from RARγ1 knockdown embryos. RARγ1 knockdown obliterates somite boundaries, and causes loss of Myod protein in the presomitic mesoderm, but ectopic, persistent expression of Myod protein in the trunk. Thus, RARγ1 is required for stabilizing the mesodermal fate, myogenic commitment, somite boundary formation, and terminal skeletal muscle differentiation.


Subject(s)
Body Patterning/genetics , Mesoderm/embryology , Muscle, Skeletal/embryology , Receptors, Retinoic Acid/genetics , Xenopus laevis/embryology , Aldehyde Dehydrogenase 1 Family , Aldehyde Oxidase/biosynthesis , Aldehyde Oxidase/genetics , Animals , CD56 Antigen/metabolism , Cell Adhesion/genetics , Gastrulation/genetics , MyoD Protein/metabolism , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase/biosynthesis , Retinoic Acid 4-Hydroxylase/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics , Tretinoin/metabolism , Xenopus Proteins/biosynthesis , Xenopus Proteins/genetics , Xenopus laevis/genetics , Retinoic Acid Receptor gamma
4.
Drug Metab Dispos ; 44(8): 1277-85, 2016 08.
Article in English | MEDLINE | ID: mdl-26842593

ABSTRACT

Aldehyde oxidase (AOX1) is an enzyme with broad substrate specificity, catalyzing the oxidation of a wide range of endogenous and exogenous aldehydes as well as N-heterocyclic aromatic compounds. In humans, the enzyme's role in phase I drug metabolism has been established and its importance is now emerging. However, the true physiologic function of AOX1 in mammals is still unknown. Further, numerous single-nucleotide polymorphisms (SNPs) have been identified in human AOX1. SNPs are a major source of interindividual variability in the human population, and SNP-based amino acid exchanges in AOX1 reportedly modulate the catalytic function of the enzyme in either a positive or negative fashion. For the reliable analysis of the effect of amino acid exchanges in human proteins, the existence of reproducible expression systems for the production of active protein in ample amounts for kinetic, spectroscopic, and crystallographic studies is required. In our study we report an optimized expression system for hAOX1 in Escherichia coli using a codon-optimized construct. The codon-optimization resulted in an up to 15-fold increase of protein production and a simplified purification procedure. The optimized expression system was used to study three SNPs that result in amino acid changes C44W, G1269R, and S1271L. In addition, the crystal structure of the S1271L SNP was solved. We demonstrate that the recombinant enzyme can be used for future studies to exploit the role of AOX in drug metabolism, and for the identification and synthesis of new drugs targeting AOX when combined with crystallographic and modeling studies.


Subject(s)
Aldehyde Oxidase/biosynthesis , Aldehyde Oxidase/genetics , Polymorphism, Single Nucleotide , Protein Engineering/methods , Aldehyde Oxidase/chemistry , Codon , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression Regulation, Enzymologic , Genotype , Humans , Kinetics , Models, Molecular , Phenotype , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Structure-Activity Relationship , Substrate Specificity
5.
J Ind Microbiol Biotechnol ; 43(4): 517-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26790417

ABSTRACT

High Pichia pastoris biomass density could be obtained using high co-feeding rate of methanol and sorbitol in a fed-batch or continuous culture, while further higher feeding rate finally leads to oxygen limitation in bioreactor. In the literature, there is lack of report about AOX1 promoter regulation with regard to dissolved oxygen level (DO). Therefore, in this work, chemostat cultures were performed to investigate the cell growth, metabolism and regulation of the AOX1 promoter (pAOX1) regarding co-feeding rate of optimized methanol/sorbitol mixture (methanol fraction 0.60 C-mol/C-mol) using a P. pastoris Mut+/pAOX1-lacZ strain. The oxygen transfer rates (OTR) in bioreactor were kept in the range of typical values of large bioreactor, i.e., 4-8 g/(L h) if DO equals 30 % saturation or 5-10 g/(L h) if DO nears zero. For DO >0, an increase of the carbon fed led to an increase of pAOX1 induction. By contrast, when dissolved oxygen was completely depleted, methanol accumulated, causing a 30 % decrease of pAOX1 induction. However, this decrease is more likely to be lined to methanol accumulation than to low level of dissolved oxygen (<4 % DO). Methanol/sorbitol co-feeding allowed cells to adapt to oxygen transient limitations that often occur at industrial scale with reduced effect on pAOX1 induction. The optimal feeding rate tested here was 6.6 mmol C (DCW h)(-1) at an OTR of 8.28 g O2(L h)(-1) with over fivefold pAOX1 induction (probably directly associated with target protein productivity) compared with previous work.


Subject(s)
Aldehyde Oxidase/biosynthesis , Bioreactors , Methanol/metabolism , Mutation , Oxygen/metabolism , Pichia/metabolism , Sorbitol/metabolism , Aldehyde Oxidase/genetics , Biomass , Carbon/metabolism , Kinetics , Methanol/pharmacology , Oxygen Consumption , Pichia/drug effects , Pichia/genetics , Pichia/growth & development , Promoter Regions, Genetic/genetics , Sorbitol/pharmacology
6.
Pharmacogenomics J ; 11(1): 15-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20177421

ABSTRACT

The clinical development of a candidate p38 kinase inhibitor was terminated because of its unexpectedly rapid clearance in human subjects. Its short half-life and metabolic profile in human beings were vastly different from that in rats, dogs, and monkeys characterized during routine pre-clinical studies. Mice generated the predominant drug (4-hydroxylated) metabolite produced in human beings, which was not found in other species. The data from a murine in vitro drug biotransformation assay that used liver extracts from 14 inbred mouse strains were analyzed by haplotype-based computational genetic analysis. This led to the identification of aldehyde oxidase-1 (AOX1) as the enzyme responsible for the rapid metabolism of this drug. Specific enzyme inhibitors and expressed recombinant enzymes were used to confirm that AOX catalyzed the formation of the 4-hydroxylated drug metabolite in mouse and man. Genetic variation within Aox1 regulated the level of hepatic Aox1 mRNA, AOX1 protein, and enzyme activity among the inbred strains. Thus, computational murine pharmacogenetic analysis can facilitate the identification and characterization of drug metabolism pathways that are differentially utilized by humans and other species.


Subject(s)
Aldehyde Oxidase/metabolism , Pharmacogenetics/methods , Protein Kinase Inhibitors/pharmacokinetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adolescent , Adult , Aldehyde Oxidase/biosynthesis , Aldehyde Oxidase/genetics , Animals , Arthritis, Rheumatoid/drug therapy , Dogs , Female , Half-Life , Haplorhini , Humans , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidinones/pharmacology , Rats , Rats, Inbred BB , Single-Blind Method , Species Specificity , Young Adult , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Drug Metab Dispos ; 37(12): 2393-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741035

ABSTRACT

Human aldehyde oxidase 1 (AOX1) has been subcloned into a vector suitable for expression in Escherichia coli, and the protein has been expressed. The resulting protein is active, with sulfur being incorporated in the molybdopterin cofactor. Expression levels are modest, but 1 liter of cells supplies enough protein for both biochemical and kinetic characterization. Partial purification is achieved by nickel affinity chromatography through the addition of six histidines to the amino-terminal end of the protein. Kinetic analysis, including kinetic isotope effects and comparison with xanthine oxidase, reveal similar mechanisms, with some subtle differences. This expression system will allow for the interrogation of human aldehyde oxidase structure/function relationships by site-directed mutagenesis and provide protein for characterizing the role of AOX1 in drug metabolism.


Subject(s)
Aldehyde Oxidase/metabolism , Cloning, Molecular , Escherichia coli/enzymology , Aldehyde Oxidase/biosynthesis , Aldehyde Oxidase/genetics , Aldehyde Oxidase/isolation & purification , Animals , Cattle , Chromatography, Affinity , Escherichia coli/genetics , Histidine , Humans , Kinetics , Mutagenesis, Site-Directed , Mutation , Oligopeptides , Recombinant Proteins/metabolism , Structure-Activity Relationship , Xanthine Oxidase/metabolism
8.
Plant J ; 42(6): 862-76, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15941399

ABSTRACT

The plant molybdenum-cofactor (Moco) and flavin-containing enzymes, xanthine dehydrogenase (XDH; EC 1.2.1.37) and aldehyde oxidase (AO; EC 1.2.3.1) are thought to play important metabolic roles in purine metabolism and hormone biosynthesis, respectively. Their animal counterparts contribute to reactive oxygen species (ROS) production in numerous pathologies and here we examined these enzymes as potential sources of ROS in plants. Novel in-gel assay techniques and Moco sulfurase mutants, lacking a sulfur ligand in their Moco active center, were employed to demonstrate that the native tomato and Arabidopsis XDHs are capable of producing O, but not H2O2, while the animal counterpart was shown to produce both, O and H2O2. Superoxide production was dependent on Moco sulfuration when using hypoxanthine/xanthine but not NADH as substrates. The activity was inhibited by diphenylene iodonium (DPI), a suicide inhibitor of FAD containing enzymes. Analysis of XDH in an Arabidopsis Atxdh1 T-DNA insertion mutant and RNA interference lines revealed loss of O activity, providing direct molecular evidence that plant XDH generates superoxides. Contrary to XDH, AO activity produced only H2O2 dissimilar to native animal AO, that can produce O as well. Surprisingly, H2O2 accumulation was not sensitive to DPI. Plant ROS production and transcript levels of AO and XDH were rapidly upregulated by application of abscisic acid and in water-stressed leaves and roots. These results, supported by in vivo measurement of ROS accumulation, indicate that plant AO and XDH are possible novel sources for ROS increase during water stress.


Subject(s)
Abscisic Acid/physiology , Aldehyde Oxidase/biosynthesis , Plants/enzymology , Reactive Oxygen Species/metabolism , Water/metabolism , Xanthine Dehydrogenase/biosynthesis , Aldehyde Oxidase/genetics , Arabidopsis/enzymology , Arabidopsis/genetics , Enzyme Induction , Solanum lycopersicum/enzymology , Solanum lycopersicum/genetics , Molecular Sequence Data , Molybdenum , Xanthine Dehydrogenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...