Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Xenobiotica ; 53(4): 223-230, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37194558

ABSTRACT

Glutathione S-transferases (GSTs) are conjugating enzymes involved in drug metabolism, antioxidant defence, and cell signalling. Herein, we investigated hepatic GST conjugation in several mouse and rat strains, including both sexes, with a direct comparison to humans.Using general and isoform-selective substrates, all mouse strains had significantly greater activities than humans for total cytosolic GST, GST-M, GST-T, and microsomal GST activities. Some strains had significantly greater GST-P activities compared to humans. Sex differences between males and females were evident in all strains for total cytosolic GST, GST-M, and GST-P, and sex differences in GST-T and microsomal GST activities within strains were noted.All rats had significantly greater activities than humans for GST-M and GST-T; only some strains were significantly greater than humans for GST-P, total cytosolic GST, and microsomal GST. Sex differences within strains showed significantly greater GST-M and GST-T activities in males compared to females. Select strains showed sex differences for total cytosolic and microsomal GST activities; there were no sex differences in GST-P activities.Significant differences in glutathione conjugation between humans and rodents exist, including sex differences. This highlights the need for careful animal selection in pre-clinical studies where GSTs are the primary metabolic pathway.


Subject(s)
Glutathione Transferase , Rodentia , Male , Female , Humans , Rats , Mice , Animals , Rodentia/metabolism , Species Specificity , Glutathione Transferase/metabolism , Liver/metabolism , Glutathione
2.
J Assist Reprod Genet ; 39(1): 227-238, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34988769

ABSTRACT

PURPOSE: Assisted reproduction technologies (ART) are associated with increased risks of pregnancy complications and obstetric interventions. Here, we aimed to determine if ART affects placental inflammation and oxidative stress as a mechanism for unfavorable pregnancy outcomes. METHODS: The levels of six cytokines (IFN-γ, IL-1ß, IL-6, IL-8, IL-10, TNFα) were measured using multiplex ELISA. The activity of four antioxidant enzymes (glutathione S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase, superoxide dismutase) and levels of two antioxidants (GSH, vitamin E) were measured using commercial/in-house assays. Markers were compared between ART and unassisted pregnancies, and then groups were stratified using ICD9/10 codes to determine differences in specific clinical contexts. RESULTS: In unassisted twin pregnancies, there was a trend of decreased cytokine levels (IL-1ß, IL-6, IL-8, TNFα, p < 0.05), but cytokines in ART twins were the same or higher. Additionally, GST and GPx activities were lower in unassisted twins, and vitamin E levels were higher in ART twins (p < 0.05). In pregnancies complicated by chorioamnionitis, there was a trend of increased cytokine levels in unassisted pregnancies (IL-1ß, IL-6, and IL-8, p < 0.05). No increase was observed in ART, and IFN-γ and TNFα were decreased (p < 0.05). Placental GST and GPx activities were higher in unassisted pregnancies with chorioamnionitis compared to ART (p < 0.05). CONCLUSION: Attenuation of protective placental inflammatory and oxidative stress responses may play a role in the underlying pathogenesis of negative birth outcomes in ART, expanding our understanding of adverse pregnancy outcomes when ART is used to conceive.


Subject(s)
Inflammation/therapy , Oxidative Stress/physiology , Pregnancy, Twin/metabolism , Adult , Chorioamnionitis/physiopathology , Female , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Placenta/metabolism , Pregnancy , Pregnancy, Twin/physiology , Reproductive Techniques, Assisted/instrumentation , Reproductive Techniques, Assisted/statistics & numerical data
3.
Eur J Drug Metab Pharmacokinet ; 46(2): 173-183, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33340340

ABSTRACT

Mathematical models that can predict the kinetics of compounds have been increasingly adopted for drug development and risk assessment. Data for these models may be generated from in vitro experimental systems containing enzymes contributing to metabolic clearance, such as subcellular tissue fractions including microsomes and cytosol. Extrapolation from these systems is facilitated by common scaling factors, known as microsomal protein per gram (MPPG) and cytosolic protein per gram (CPPG). Historically, parameterization of MPPG and CPPG has employed the use of recovery factors, commonly benchmarked to cytochromes P450 which work well in some contexts, but could be problematic for other enzymes. Here, we propose absolute quantification of protein content and supplementary assays to evaluate microsomal/cytosolic purity that should be employed. Examples include calculation of microsomal latency by mannose-6-phosphatase activity and immunoblotting of subcellular fractions with fraction-specific markers. Further considerations include tissue source, as disease states can affect enzyme expression and activity, and the methodology used for scalar parameterization. Regional- and organ-specific expression of enzymes, in addition to differences in organ physiology, is another important consideration. Because most efforts have focused on the liver that is, for the most part, homogeneous, derived scalars may not capture the heterogeneity of other major tissues contributing to xenobiotic metabolism including the kidneys and small intestine. Better understanding of these scalars, and how to appropriately derive them from extrahepatic tissues can provide support to the inferences made with physiologically based pharmacokinetic modeling, increase its accuracy in characterizing in vivo drug pharmacokinetics, and improve confidence in go-no-go decisions for clinical trials.


Subject(s)
Cytosol/metabolism , Microsomes/metabolism , Models, Theoretical , Proteins/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Drug Development/methods , Humans , Pharmacokinetics , Risk Assessment/methods , Subcellular Fractions/metabolism , Xenobiotics/metabolism
4.
Toxicol Rep ; 7: 1311-1318, 2020.
Article in English | MEDLINE | ID: mdl-33072523

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs are contraindicated in the third trimester of pregnancy due to negative effects including alteration of uteroplacental blood flow, premature ductus arteriosus closure, and adverse effects on the fetal kidney. However, many women are unaware of these risks, and commonly report their use in pregnancy. We aimed to determine if umbilical cord was a reliable matrix for detecting NSAID use, determine incidence of use close to labour, and uncover associations with obstetric/neonatal outcomes. METHODS: We developed a UHPLC-MS/MS method to simultaneously detect diclofenac, ibuprofen, indomethacin, naproxen, and salicylic acid in plasma and umbilical cord lysate. Using this method, we screened 380 lysates to determine the prevalence of NSAID use. Results were compared to the clinical outcomes in pregnancy using ICD9/10 chart codes (n = 21). RESULTS: The UHPLC-MS/MS method has excellent linearity, accuracy, and precision in solvent and plasma, but lower sensitivity in umbilical cord lysate. We report a 3 % rate of NSAID ingestion within days of labour - the pharmacokinetically-determined window for active ingestion. There were no significant differences observed for maternal, obstetric, or neonatal outcomes between the NSAID positive group (n = 11) and NSAID negative group (n = 369). CONCLUSIONS: Because NSAID use in third trimester is contraindicated, even a 3% usage rate is alarmingly high. Based on UHPLC-MS/MS performance of umbilical cord lysate, 3% is likely a conservative estimate. Recent adoption of NSAIDs under clinical supervision to support in vitro fertilisation and prevent pre-eclampsia indicates future work should focus on determining safe dosages of NSAIDs and the correct therapeutic window in pregnancy.

5.
ACS Omega ; 5(12): 6872-6887, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32258923

ABSTRACT

The elimination of numerous endogenous compounds and xenobiotics via glucuronidation by uridine-5'-diphosphate glycosyltransferase enzymes (UGTs) is an essential process of the body's chemical defense system. UGTs have distinct but overlapping substrate preferences, but the molecular basis for their substrate specificity remains poorly understood. Three-dimensional protein structures can greatly enhance our understanding of the interactions between enzymes and their substrates, but because of the inherent difficulties in purifying and crystallizing integral endoplasmic reticulum membrane proteins, no complete mammalian UGT structure has yet been produced. To address this problem, we have created a homology model of UGT1A6 using I-TASSER to explore, in detail, the interactions of human UGT1A6 with its substrates. Ligands were docked into our model in the presence of the cosubstrate uridine-5'-diphosphate-glucuronic acid, interacting residues were examined, and poses were compared to those cocrystallized with various plant and bacterial glycosyltransferases (GTs). Our model structurally resembles other GTs, and docking experiments replicated many of the expected UGT-substrate interactions. Some bias toward the template structures' protein-substrate interactions and binding preferences was evident.

6.
J Clin Pharmacol ; 60(6): 722-733, 2020 06.
Article in English | MEDLINE | ID: mdl-31879975

ABSTRACT

Uridine diphosphate glucuronosyltransferases (UGTs) catalyze glucuronidation to facilitate systemic and local clearance of numerous chemicals and drugs. To investigate whether UGT expression is coregulated in human liver, we analyzed the protein expression of UGTs 1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 3A1, and 3A2 using western blots from 164 healthy human liver samples, comparing expression with age and sex. UGT1A6 levels were significantly higher in children than adults, and UGT3A1 and 3A2 expression significantly increased with age from childhood to age >65 yearas. In children aged <18 years, UGT1A4/1A9 protein expression was significantly correlated, but not for adults aged >18 years. UGT1A3 expression was always significantly correlated with other UGT1A isoforms in all adults aged >18 years. In individuals aged ≥12 years, expression of UGT1A1/1A4, UGT1A1/1A6, UGT1A1/1A9, and UGT1A4/1A6 significantly correlated, which was not observed in children aged <12 years. In contrast, UGT1A4/2B7 showed significant correlation in children aged <12 years, but not in individuals aged ≥12 years, and this was observed in female but not male individuals. Expression of UGT1A6/1A9 and UGT3A1/3A2 correlated in the entire sample population, but UGT3As did not correlate with other UGTs. These correlations were sex dependent, as UGT1A3/1A1, UGT1A4/2B7 and UGT3A1/3A2 correlated more highly in male than female individuals, while UGT1A4/1A6 protein correlated more significantly in female than male individuals. This is the first report on the ontogeny of UGT3A isoforms, showing maximal expression in the elderly, and is the first demonstration that UGT isoforms commonly coexpress in vivo, in both age-dependent and sex-dependent manners.


Subject(s)
Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/enzymology , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Female , Gene Expression Regulation , Glucuronides/metabolism , Humans , Infant , Infant, Newborn , Male , Microsomes, Liver/enzymology , Middle Aged , Sex Characteristics , Young Adult
7.
Xenobiotica ; 48(9): 882-890, 2018 Sep.
Article in English | MEDLINE | ID: mdl-28868965

ABSTRACT

1. The UDP-glucuronosyltransferase (UGT) enzymes are important in the metabolism, elimination and detoxification of many xenobiotics and endogenous compounds. As extrapolation of in vitro kinetics of drug metabolizing enzymes to predict in vivo clearance rates becomes more sophisticated, it is important to ensure proper optimization of enzyme assays. The luminal location of the enzyme active site (i.e. latency), and the complexity of UGT kinetics, results in consistent under-prediction of clearance of drugs metabolized by glucuronidation. 2. We examined inhibition of UGT activity in alamethicin-disrupted human liver microsomes (HLM) by uridine diphosphate (UDP), a UGT reaction product, and its reversal by Mg2+ ions. We also determined whether UDP-sugars other than the co-substrate UDP-glucuronic acid (UDP-GlcA) affected glucuronidation. 3. We show that other UDP-sugars do not significantly influence glucuronidation. We also demonstrate that UDP inhibits HLM UGT activity and that this is reversed by including Mg2+ in the assay. The Mg2+ effect can be offset using EDTA, and is dependent on the concentration of UDP-GlcA in the assay. 4. We propose that formation of a Mg2+-UDP complex prevents UDP from affecting the enzyme. Our results suggest that 5 mM UDP-GlcA and 10 mM Mg2+ be used for UGT assays in fully disrupted HLM.


Subject(s)
Glucuronosyltransferase/metabolism , Magnesium/pharmacology , Microsomes, Liver/drug effects , Uridine Diphosphate Sugars/pharmacology , Uridine Diphosphate/pharmacology , Alamethicin/pharmacology , Humans , Microsomes, Liver/metabolism
8.
Pharmaceutics ; 9(3)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28867809

ABSTRACT

Uridine diphosphate-glucuronosyltransferases (UGTs) are phase 2 conjugation enzymes mainly located in the endoplasmic reticulum (ER) of the liver and many other tissues, and can be recovered in artificial ER membrane preparations (microsomes). They catalyze glucuronidation reactions in various aglycone substrates, contributing significantly to the body's chemical defense mechanism. There has been controversy over the last 50 years in the UGT field with respect to the explanation for the phenomenon of latency: full UGT activity revealed by chemical or physical disruption of the microsomal membrane. Because latency can lead to inaccurate measurements of UGT activity in vitro, and subsequent underprediction of drug clearance in vivo, it is important to understand the mechanisms behind this phenomenon. Three major hypotheses have been advanced to explain UGT latency: compartmentation, conformation, and adenine nucleotide inhibition. In this review, we discuss the evidence behind each hypothesis in depth, and suggest some additional studies that may reveal more information on this intriguing phenomenon.

9.
Chem Biol Interact ; 259(Pt A): 2-7, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27174136

ABSTRACT

The sulfuryl transfer reaction is of fundamental biological importance. One of the most important manifestations of this process are the reactions catalyzed by members of the cytosolic sulfotransferase (SULT) superfamily. These enzymes transfer the sulfuryl moiety from the universal donor PAPS (3'-phosphoadenosine 5'-phosphosulfate) to a wide variety of substrates with hydroxyl- or amino-groups. Normally a detoxification reaction this facilitates the elimination of a multitude of xenobiotics, although for some molecules sulfation is a bioactivation step. In addition, sulfation plays a key role in endocrine and other signalling pathways since many steroids, sterols, thyroid hormones and catecholamines exist primarily as sulfate conjugates in humans. This article summarizes much of our current knowledge of the organization and function of the human cytosolic sulfotransferases and highlights some of the important interspecies differences that have implications for, among other things, drug development and chemical safety analysis.


Subject(s)
Phosphoadenosine Phosphosulfate/metabolism , Sulfotransferases/metabolism , Humans , Inactivation, Metabolic , Substrate Specificity , Sulfates/metabolism
10.
Drug Metab Lett ; 9(2): 99-108, 2015.
Article in English | MEDLINE | ID: mdl-26031463

ABSTRACT

Understanding the detailed ontogeny of xenobiotic metabolizing enzymes is important if we are to be able to predict the risk of toxicity to the developing fetus or the fate of drugs in neonates and children. This review summarizes current knowledge of the development of the major families of conjugating enzymes in humans: the UDP-glucuronosyltransferases, sulfotransferases, glutathione S-transferases, arylamine N-acetyltransferases and methyltransferases; little is known of the last three. Based on the available information, sulfation appears to be the most highly developed pathway during fetal development where glucuronidation in particular is lacking. Following birth, glucuronidation capacity develops rapidly and for many of the enzymes adult capacity occurs in mid-late childhood. The importance of developing pharmacokinetic and physiology-based pharmacokinetic models to support the more informed use of drugs in children is highlighted.


Subject(s)
Liver/enzymology , Metabolic Detoxication, Phase II , Xenobiotics/pharmacokinetics , Age Factors , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Liver/growth & development , Methyltransferases/genetics , Methyltransferases/metabolism , Morphogenesis , Substrate Specificity , Sulfotransferases/genetics , Sulfotransferases/metabolism , Xenobiotics/administration & dosage
11.
Xenobiotica ; 45(6): 495-502, 2015.
Article in English | MEDLINE | ID: mdl-25539458

ABSTRACT

1. Cattle are an important component of the human food chain. Drugs used either legally or illegally in cattle may therefore enter the food chain and it is thus important to understand pathways of drug metabolism in this species, including sulfation catalyzed by the sulfotransferases (SULTs). 2. In this study, we have analyzed the sulfation of 4-nitrophenol and other compounds in male and female bovine liver and characterized recombinant bovine SULT isoforms 1A1 and 1B1 expressed in Escherichia coli. 3. We found that, in contrast to most other mammalian species, the major phenol sulfotransferase SULT1A1 is not expressed in bovine liver. Rather SULT1B1 seems to be a major form in both male and female bovine liver. 4. We also identified kinetic differences between bovine and human SULT1A1 and, using the human SULT1A1 crystal structure, identified two amino acid positions in the active site of bovine SULT1A1 (Ile89Val and Phe247Val) that may be responsible for these differences.


Subject(s)
Liver/enzymology , Sulfotransferases/chemistry , Sulfotransferases/metabolism , Animals , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Cattle , Crystallography, X-Ray , Female , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Nitrophenols/pharmacokinetics , Nitrophenols/pharmacology , Sulfotransferases/genetics
12.
Biofactors ; 39(3): 271-8, 2013.
Article in English | MEDLINE | ID: mdl-23281118

ABSTRACT

Morphine is converted to morphine 3-ß-D-glucuronide (M3G) by the UDP-glucuronosyltransferase Ugt2b1 in the endoplasmic reticulum (ER) of rat liver. Because of its luminal localization, UGT activity requires UDP-glucuronate import and glucuronide export across the ER membrane. The former transport is generally considered to be rate limiting and to explain the latency of UGT activities in intact microsomal vesicles. However, some observations indicate that the release of bulky glucuronides, such as M3G, might also be rate limiting for glucuronidation. This assumption was tested by characterizing the transport of M3G and its distribution between the intra- and extravesicular spaces during synthesis in rat liver microsomes. The amount of vesicle-associated M3G was measured using rapid filtration and LC-MS measurement. Our results reveal a remarkable accumulation of newly synthesized M3G in the microsomal lumen above the equilibrium. The transport showed a linear concentration-dependence in a wide range (5-200 µM). Therefore, the build-up of high (about 20 µM) luminal M3G concentration could adjust the rate of release to that of synthesis (44.85 ± 4.08 pmol/min/mg protein) during the conjugation of 100 µM morphine. These data can explain earlier findings indicative of separate intracellular pools of M3G in rat liver. Accumulation of bulky glucuronides in the ER lumen might also play an important role in their targeting and in the control of biliary excretion.


Subject(s)
Microsomes, Liver/metabolism , Morphine Derivatives/metabolism , Animals , Biological Transport/physiology , Chromatography, Liquid , Endoplasmic Reticulum/metabolism , In Vitro Techniques , Male , Mass Spectrometry , Rats , Rats, Wistar
13.
Drug Metab Dispos ; 40(7): 1321-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22492655

ABSTRACT

This article reports on the development of UDP-glucuronosyltransferase 1A9 (UGT1A9) in neonatal and pediatric liver. The substrate 4-methylumbelliferone (4MU) with specific inhibition by niflumic acid was used to define specific UGT1A9 activity. Subsequently, in silico pharmacokinetic (PK) and physiology-based pharmacokinetic (PBPK) modeling was used to determine UGT1A9 maturation and hepatic clearance. Modeled maximal enzyme activity was 27.9 nmol · min(-1) · mg protein(-1) at 4 months of age, which had high concordance with the average V(max) in 45 individual adult (>20 years) livers of 29.0 nmol · min(-1) · mg protein(-1). The activity of UGT1A9 ranged 7.5-fold in the adult population (4.1-54.5 nmol · min(-1) · mg protein(-1)). Expression of UGT1A9 correlated with age only in children younger than 1 year (Spearman r = 0.70). Activity correlated with expression up to 18 years of age (Spearman r = 0.76). Furthermore, scaling intrinsic hepatic clearance of 4MU with an allometric PK model yielded a high clearance at birth and then fell to adult levels (1.3 l · h(-1) · kg(-1) at 18.1 years for well stirred or 1.4 l · h(-1) · kg(-1) at 18.7 years for parallel tube). The Simcyp PBPK models did not converge but showed an increase in clearance at under 1 year of age and then decreased to adult levels at approximately 20 years of age. Allometric scaling may be more accurate in cases of high-extraction drugs. Enzyme activities or hepatic clearances did not differ with gender or ethnicity. The UGT1A9 isoform has higher normalized clearance for 4MU at young ages, which may explain how other UGT1A9 substrates, such as propofol, have higher clearances in children than in adults.


Subject(s)
Glucuronosyltransferase/metabolism , Liver/enzymology , Liver/growth & development , Microsomes, Liver/enzymology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Enzyme Inhibitors/pharmacology , Female , Humans , Hymecromone/analogs & derivatives , Hymecromone/metabolism , Infant , Infant, Newborn , Male , Microsomes, Liver/drug effects , Middle Aged , Niflumic Acid/pharmacology , UDP-Glucuronosyltransferase 1A9 , Young Adult
14.
Glycobiology ; 22(4): 561-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22156920

ABSTRACT

Glycosaminoglycan (GAG) assembly initiates through the formation of a linkage tetrasaccharide region serving as a primer for both chondroitin sulfate (CS) and heparan sulfate (HS) chain polymerization. A possible role for sulfation of the linkage structure and of the constitutive disaccharide unit of CS chains in the regulation of CS-GAG chain synthesis has been suggested. To investigate this, we determined whether sulfate substitution of galactose (Gal) residues of the linkage region or of N-acetylgalactosamine (GalNAc) of the disaccharide unit influences activity and specificity of chondroitin sulfate N-acetylgalactosaminyltransferase-1 (CSGalNAcT-1), a key glycosyltransferase of CS biosynthesis. We synthesized a series of sulfated and unsulfated analogs of the linkage oligosaccharide and of the constitutive unit of CS and tested these molecules as potential acceptor substrates for the recombinant human CSGalNAcT-1. We show here that sulfation at C4 or C6 of the Gal residues markedly influences CSGalNAcT-1 initiation activity and catalytic efficiency. Kinetic analysis indicates that CSGalNAcT-1 exhibited 3.6-, 1.6-, and 2.2-fold higher enzymatic efficiency due to lower K(m) values toward monosulfated trisaccharides substituted at C4 or C6 position of Gal1, and at C6 of Gal2, respectively, compared with the unsulfated oligosaccharide. This highlights the critical influence of Gal substitution on both CSGalNAcT-1 activity and specifity. No GalNAcT activity was detected toward sulfated and unsulfated analogs of the CS constitutive disaccharide (GlcA-ß1,3-GalNAc), indicating that CSGalNAcT-1 was involved in initiation but not in elongation of CS chains. Our results strongly suggest that sulfation of the linkage region acts as a regulatory signal in CS chain initiation.


Subject(s)
Chondroitin Sulfates/chemistry , N-Acetylgalactosaminyltransferases/chemistry , Acetylglucosamine/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Galactans/chemistry , Galactose/chemistry , Glycosylation , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Oligosaccharides/chemistry , Recombinant Proteins/chemistry , Substrate Specificity
15.
Biochem Pharmacol ; 83(2): 279-85, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22062654

ABSTRACT

The ATP-binding cassette (ABC) transporters breast cancer resistance protein (BCRP), multidrug resistance-associated protein 2 (MRP2), and P-glycoprotein (Pgp) are important in the distribution and elimination of many drugs and endogenous metabolites. Due to their membrane location and hydrophobicity it is difficult to generate purified protein standards to quantify these transporters in human tissues. The present study generated transporter proteins fused with the S-peptide of ribonuclease for use as standards in immunoquantification in human liver and small intestine. Quantification of the S•tag™, a 15 amino acid peptide, is based on the formation of a functional ribonuclease activity upon its high affinity reconstitution with ribonuclease S-protein. S-tagged transporters were used as full-length protein standards in the immunoquantification of endogenous BCRP, MRP2, and Pgp levels in 14 duodenum and 13 liver human tissue samples. Expression levels in the duodenum were 305±248 (BCRP), 66±70 (MRP2), and 275±205 (Pgp) fmoles per cm(2). Hepatic levels were 2.6±0.9 (BCRP), 19.8±10.5 (MRP2), and 26.1±10.1 (total Pgp) pmoles per g of liver. The mean hepatic scaling factor was 35.8mg crude membrane per g of liver, and the mean duodenal scaling factor was 1.3mg crude membrane per cm(2) mucosal lining. Interindividual variability was greater in duodenal samples than liver samples. It is hoped that this innovative method of quantifying these transporters (and other membrane proteins) will improve in vivo-in vitro extrapolation and in silico prediction of drug absorption and elimination, thus supporting drug development.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/chemistry , Duodenum/metabolism , Gene Expression Regulation , Liver/metabolism , Peptide Fragments/standards , Ribonuclease, Pancreatic/standards , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/standards , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/standards , Duodenum/chemistry , HEK293 Cells , Humans , Immunoblotting/methods , Immunoblotting/standards , Liver/chemistry , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/biosynthesis , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/standards , Neoplasm Proteins/biosynthesis , Peptide Fragments/chemistry , Predictive Value of Tests , Reproducibility of Results , Ribonuclease, Pancreatic/chemistry
16.
Drug Metab Dispos ; 39(12): 2258-63, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21880828

ABSTRACT

Glucuronidation is a major pathway of drug and xenobiotic metabolism that is catalyzed by members of the UDP-glucuronosyltransferase (UGT) family. Predicting the contribution of individual UGTs to drug metabolism would be of considerable value in drug development and would be greatly aided by the availability of detailed absolute expression levels of these proteins; this is hampered by the lack of purified protein standards because of the hydrophobic membrane-associated nature of UGTs and the consequential difficulties in expression and purification. Here we describe a novel solution to this problem by expressing UGTs in Escherichia coli as fusion proteins with ribonuclease S-peptide, targeted to the periplasm with the pelB leader sequence. After addition of ribonuclease S-protein to membrane extracts, a functional ribonuclease is reconstituted that provides a direct and absolute quantification of the amount of UGT fusion protein; this is subsequently used to generate standard curves for immunoquantification by immunoblotting. To illustrate the value of the method, we have quantified the expression of UGT1A1 and UGT1A6 in human liver and kidney microsomes using new isoform-specific antibodies developed against peptides from these proteins. Expression levels of both proteins in liver were highly variable (28- and 20-fold, respectively) and correlated strongly with UGT enzyme activity toward the probe substrates bilirubin and 1-naphthol, respectively. The method is broadly applicable and provides a straightforward means of determining the absolute, as opposed to relative, quantities of UGT proteins present in human tissues.


Subject(s)
Glucuronosyltransferase/metabolism , Isoenzymes/metabolism , Base Sequence , Blotting, Western , DNA Primers , Humans , Polymerase Chain Reaction
17.
J Biol Chem ; 285(48): 37342-58, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20843813

ABSTRACT

Glycosaminoglycans (GAGs) play a central role in many pathophysiological events, and exogenous xyloside substrates of ß1,4-galactosyltransferase 7 (ß4GalT7), a major enzyme of GAG biosynthesis, have interesting biomedical applications. To predict functional peptide regions important for substrate binding and activity of human ß4GalT7, we conducted a phylogenetic analysis of the ß1,4-galactosyltransferase family and generated a molecular model using the x-ray structure of Drosophila ß4GalT7-UDP as template. Two evolutionary conserved motifs, (163)DVD(165) and (221)FWGWGREDDE(230), are central in the organization of the enzyme active site. This model was challenged by systematic engineering of point mutations, combined with in vitro and ex vivo functional assays. Investigation of the kinetic properties of purified recombinant wild-type ß4GalT7 and selected mutants identified Trp(224) as a key residue governing both donor and acceptor substrate binding. Our results also suggested the involvement of the canonical carboxylate residue Asp(228) acting as general base in the reaction catalyzed by human ß4GalT7. Importantly, ex vivo functional tests demonstrated that regulation of GAG synthesis is highly responsive to modification of these key active site amino acids. Interestingly, engineering mutants at position 224 allowed us to modify the affinity and to modulate the specificity of human ß4GalT7 toward UDP-sugars and xyloside acceptors. Furthermore, the W224H mutant was able to sustain decorin GAG chain substitution but not GAG synthesis from exogenously added xyloside. Altogether, this study provides novel insight into human ß4GalT7 active site functional domains, allowing manipulation of this enzyme critical for the regulation of GAG synthesis. A better understanding of the mechanism underlying GAG assembly paves the way toward GAG-based therapeutics.


Subject(s)
Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Glycosaminoglycans/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalytic Domain , Galactosyltransferases/genetics , Humans , Invertebrates/chemistry , Invertebrates/classification , Invertebrates/enzymology , Invertebrates/genetics , Models, Molecular , Molecular Sequence Data , Phylogeny , Sequence Alignment , Substrate Specificity , Vertebrates/classification , Vertebrates/genetics , Vertebrates/metabolism
18.
FEBS Lett ; 584(18): 3962-8, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20691685

ABSTRACT

ß1,4-Galactosyltransferase 7 (ß4GalT7) is a key enzyme initiating glycosaminoglycan (GAG) synthesis. Based on in vitro and ex vivo kinetics studies and structure-based modelling, we molecularly characterized ß4GalT7 mutants linked to the progeroid form of Ehlers-Danlos syndrome (EDS), a severe connective tissue disorder. Our results revealed that loss of activity upon L206P substitution due to altered protein folding is the primary cause for the GAG synthesis defect in patients carrying the compound A186D and L206P mutations. We showed that R270C substitution strongly reduced ß4GalT7 affinity towards xyloside acceptor, thus affecting GAG chains formation. This study establishes the molecular basis for ß4GalT7 defects associated with altered GAG synthesis in EDS.


Subject(s)
Ehlers-Danlos Syndrome/enzymology , Galactosyltransferases/metabolism , Glycosaminoglycans/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Ehlers-Danlos Syndrome/genetics , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Humans , Models, Molecular , Mutation
19.
Methods Mol Biol ; 640: 309-26, 2010.
Article in English | MEDLINE | ID: mdl-20645060

ABSTRACT

Since phase II reactions quantitatively represent the most important pathways involved in drug biotransformation, the development and the use of in vitro approaches to predict glucuronidation and sulfation are currently attracting intense interest to assist in the selection of new drug candidates and for the optimization of dosage regimens for established drugs. At present, primary cultures of human hepatocytes represent the most suitable in vitro model for drug metabolism studies. This system theoretically expresses the full complement of drug-metabolizing enzymes associated with the endoplasmic reticulum (CYP and UDP-glucuronosyltransferases) or located in the cytosolic compartment (sulfotransferases), and relevant accessory proteins required for drug transport and excretion. Primary hepatocytes also represent a unique in vitro model for global examination of inductive potential of drugs on conjugation reactions (monitored as increases in mRNA content or activity). The progress in cryopreservation over the last decade has made available preserved hepatocytes to address key issues such as the (i) establishment of phase II metabolic profile and rate, (ii) identification of conjugation enzymes involved, and (iii) evaluation of drug-drug interactions. These advances allow a better assessment of phase II reactions during drug discovery and development.


Subject(s)
Glucuronosyltransferase/metabolism , Hepatocytes/enzymology , Pharmaceutical Preparations/metabolism , Sulfotransferases/metabolism , Cells, Cultured , Drug Interactions , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/enzymology , Liver/metabolism , Models, Biological
20.
FASEB J ; 24(2): 436-50, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19812376

ABSTRACT

Heparan sulfate proteoglycans (HSPGs), strategically located at the cell-tissue-organ interface, regulate major biological processes, including cell proliferation, migration, and adhesion. These vital functions are compromised in tumors, due, in part, to alterations in heparan sulfate (HS) expression and structure. How these modifications occur is largely unknown. Here, we investigated whether epigenetic abnormalities involving aberrant DNA methylation affect HS biosynthetic enzymes in cancer cells. Analysis of the methylation status of glycosyltransferase and sulfotransferase genes in H-HEMC-SS chondrosarcoma cells showed a typical hypermethylation profile of 3-OST sulfotransferase genes. Exposure of chondrosarcoma cells to 5-aza-2'-deoxycytidine (5-Aza-dc), a DNA-methyltransferase inhibitor, up-regulated expression of 3-OST1, 3-OST2, and 3-OST3A mRNAs, indicating that aberrant methylation affects transcription of these genes. Furthermore, HS expression was restored on 5-Aza-dc treatment or reintroduction of 3-OST expression, as shown by indirect immunofluorescence microscopy and/or analysis of HS chains by anion-exchange and gel-filtration chromatography. Notably, 5-Aza-dc treatment of HEMC cells or expression of 3-OST3A cDNA reduced their proliferative and invading properties and augmented adhesion of chondrosarcoma cells. These results provide the first evidence for specific epigenetic regulation of 3-OST genes resulting in altered HSPG sulfation and point to a defect of HS-3-O-sulfation as a factor in cancer progression.


Subject(s)
DNA Methylation , Heparan Sulfate Proteoglycans/biosynthesis , Sulfotransferases/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chondrosarcoma/genetics , CpG Islands/physiology , DNA Methylation/drug effects , Decitabine , Enzyme Repression , HL-60 Cells , HeLa Cells , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Neoplasm Invasiveness/genetics , Sulfotransferases/drug effects , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...