101
|
Hebbring SJ, Moyer AM, Weinshilboum RM. Sulfotransferase gene copy number variation: pharmacogenetics and function. Cytogenet Genome Res 2009; 123:205-10. [PMID: 19287157 DOI: 10.1159/000184710] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2008] [Indexed: 11/19/2022] Open
Abstract
Pharmacogenetics is the study of the role of inheritance in variation to drug response. Drug response phenotypes can vary from adverse drug reactions at one end of the spectrum to equally serious lack of the desired effect of drug therapy at the other. Many of the current important examples of pharmacogenetics involve inherited variation in drug metabolism. Sulfate conjugation catalyzed by cytosolic sulfotransferase (SULT) enzymes, particularly SULT1A1, is a major pathway for drug metabolism in humans. Pharmacogenetic studies of SULT1A1 began over a quarter of a century ago and have advanced from biochemical genetic experiments to include cDNA and gene cloning, gene resequencing, and functional studies of the effects of single nucleotide polymorphisms (SNPs). SNP genotyping, in turn, led to the discovery of functionally important copy number variations (CNVs) in the SULT1A1 gene. This review will briefly describe the evolution of our understanding of SULT1A1 pharmacogenetics and CNV, as well as challenges involved in utilizing both SNP and CNV data in an attempt to predict SULT1A1 function. SULT1A1 represents one example of the potential importance of CNV for the evolving disciplines of pharmacogenetics and pharmacogenomics.
Collapse
Affiliation(s)
- S J Hebbring
- Division of Clinical Pharmacology, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Medical School - Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
102
|
Alnouti Y. Bile Acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci 2009; 108:225-46. [PMID: 19131563 DOI: 10.1093/toxsci/kfn268] [Citation(s) in RCA: 297] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sulfotransferase-2A1 catalyzes the formation of bile acid-sulfates (BA-sulfates). Sulfation of BAs increases their solubility, decreases their intestinal absorption, and enhances their fecal and urinary excretion. BA-sulfates are also less toxic than their unsulfated counterparts. Therefore, sulfation is an important detoxification pathway of BAs. Major species differences in BA sulfation exist. In humans, only a small proportion of BAs in bile and serum are sulfated, whereas more than 70% of BAs in urine are sulfated, indicating their efficient elimination in urine. The formation of BA-sulfates increases during cholestatic diseases. Therefore, sulfation may play an important role in maintaining BA homeostasis under pathologic conditions. Farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, and vitamin D receptor are potential nuclear receptors that may be involved in the regulation of BA sulfation. This review highlights current knowledge about the enzymes and transporters involved in the formation and elimination of BA-sulfates, the effect of sulfation on the pharmacologic and toxicologic properties of BAs, the role of BA sulfation in cholestatic diseases, and the regulation of BA sulfation.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
103
|
Caron G, Nurisso A, Ermondi G. How to Extend the Use of Grid‐Based Interaction Energy Maps from Chemistry to Biotopics. ChemMedChem 2009; 4:29-36. [DOI: 10.1002/cmdc.200800259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Giulia Caron
- CASMedChem Laboratory, Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Via P. Giuria 9, I‐10125 Torino (Italy), Fax: (+39) 011 670 7687
| | - Alessandra Nurisso
- CASMedChem Laboratory, Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Via P. Giuria 9, I‐10125 Torino (Italy), Fax: (+39) 011 670 7687
| | - Giuseppe Ermondi
- CASMedChem Laboratory, Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Via P. Giuria 9, I‐10125 Torino (Italy), Fax: (+39) 011 670 7687
| |
Collapse
|
104
|
Yasuda T, Yasuda S, Williams FE, Liu MY, Sakakibara Y, Bhuiyan S, Snow R, Carter G, Liu MC. Characterization and ontogenic study of novel steroid-sulfating SULT3 sulfotransferases from zebrafish. Mol Cell Endocrinol 2008; 294:29-36. [PMID: 18644423 DOI: 10.1016/j.mce.2008.06.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2007] [Revised: 06/16/2008] [Accepted: 06/20/2008] [Indexed: 10/21/2022]
Abstract
In vertebrates, sulfation as catalyzed by members of the cytosolic sulfotransferase (SULT) family has been suggested to be involved in the homeostasis of steroids. To establish the zebrafish as a model for investigating how sulfation functions to regulate steroid metabolism during the developmental process, we have embarked on the identification of steroid-sulfating SULTs in zebrafish. By searching the GenBank database, we identified two putative cytosolic SULT sequences from zebrafish, designated SULT3 ST1 and ST2. The recombinant proteins of these two zebrafish SULT3 STs were expressed in and purified from BL21 (DE3) cells transformed with the pGEX-2TK expression vector harboring SULT3 ST1 or ST2 cDNA. Upon enzymatic characterization, purified SULT3 ST1 displayed the strongest sulfating activity toward 17beta-estradiol among the endogenous substrates tested, while SULT3 ST2 exhibited substrate specificity toward hydroxysteroids, particularly dehydroepiandrosterone (DHEA). The pH-dependence and kinetic constants of these two enzymes with 17beta-estradiol and DHEA were determined. A developmental expression study revealed distinct patterns of the expression of SULT3 ST1 and ST2 during embryonic development and throughout the larval stage onto maturity. Collectively, these results imply that these two steroid-sulfating SULT3 STs may play differential roles in the metabolism and regulation of steroids during zebrafish development and in adulthood.
Collapse
Affiliation(s)
- Tomoko Yasuda
- Department of Pharmacology, College of Pharmacy, The University of Toledo, Toledo, OH 43606, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Tyapochkin E, Cook PF, Chen G. Isotope exchange at equilibrium indicates a steady state ordered kinetic mechanism for human sulfotransferase. Biochemistry 2008; 47:11894-9. [PMID: 18928301 DOI: 10.1021/bi801211t] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytosolic sulfotransferase (SULT)-catalyzed sulfation regulates biosignaling molecular biological activities and detoxifies hydroxyl-containing xenobiotics. The universal sulfuryl group donor for SULTcatalyzed sulfation is adenosine 3'-phosphate 5'-phosphosulfate (PAPS). The reaction products are a sulfated product and adenosine 3',5'-diphosphate (PAP). Although the kinetics has been reported since the 1980s,SULT-catalyzed reaction mechanisms remain unclear. Human SULT1A1 catalyzes the sulfation of xenobiotic phenols and has very broad substrate specificity. It has been recognized as one of the most important phase II drug-metabolizing enzymes. Understanding the kinetic mechanism of this isoform is important in understanding drug metabolism and xenobiotic detoxification. In this report, we investigated the SULT1A1-catalyzed phenol sulfation mechanism. The SULT1A1-catalyzed reaction was brought to equilibrium by varying substrate (1-naphthol) and PAPS initial concentrations. Equilibrium constants were determined. Two isotopic exchanges at equilibrium ([14C]1-naphthol <=>[14C]1-naphthyl sulfate and[35S]PAPS<=>[35S]1-naphthyl sulfate) were conducted. First-order kinetics, observed for all the is otopic exchange reactions studied over the entire time scale that was monitored, indicates that the system was truly at equilibrium prior to addition of an isotopic pulse. Complete suppression of the 35S isotopic exchange rate was observed with an increase in the levels of 1-naphthol and 1-naphthyl sulfate in a constant ratio,while no suppression of the 14C exchange rate was observed with an increase in the levels of PAPS and PAP in a constant ratio. Data are consistent with a steady state ordered kinetic mechanism with PAPS and PAP binding to the free enzyme.
Collapse
Affiliation(s)
- Eduard Tyapochkin
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | | | | |
Collapse
|
106
|
Adjei AA, Gaedigk A, Simon SD, Weinshilboum RM, Leeder JS. Interindividual variability in acetaminophen sulfation by human fetal liver: implications for pharmacogenetic investigations of drug-induced birth defects. ACTA ACUST UNITED AC 2008; 82:155-65. [PMID: 18232020 DOI: 10.1002/bdra.20535] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Acetaminophen (APAP) use in early pregnancy has been associated with the risk of gastroschisis, a rare but serious congenital defect of the abdominal wall. The purpose of this study was to characterize the variability of APAP sulfation in a panel of human fetal livers and to identify the sulfotransferases (SULT) isoform(s) responsible for catalyzing that activity. METHODS APAP sulfation was determined in a panel of human fetal (n = 73) and postnatal (n = 18) liver cytosol preparations and correlated with the catalytic activity of various SULT isoforms as determined using prototypic substrates and specific antibodies. RESULTS Of 10 heterologously expressed SULT isoforms examined, SULT1A1, SULT1A3/4, SULT1E1, and SULT2A1 all catalyzed the formation of APAP sulfate with K(m) values of 2.4, 1.5, 1.9, and 3.7 mM, respectively. Catalytic activities for these four isoforms were expressed at varying levels in human fetal liver, and APAP sulfation was positively correlated with each of the four prototypic activities. Several regression and clustering approaches revealed that SULT1A3/4 was the primary determinant of prenatal APAP sulfation but that SULT1A1 or SULT1E1 were also major contributors in subsets of samples. CONCLUSIONS The results of this study lead to the hypothesis that genetic variation in SULT1A3/4 represents a risk factor for the development of gastroschisis in the offspring of mothers exposed to APAP early in pregnancy. Interpretation of genetic association studies conducted to test this hypothesis will be complicated by the variable contributions of other SULTs toward APAP-sulfate formation in individual subjects.
Collapse
Affiliation(s)
- Araba A Adjei
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
107
|
Aspergillus niger metabolism of citrus furanocoumarin inhibitors of human cytochrome P450 3A4. Appl Microbiol Biotechnol 2008; 78:343-9. [PMID: 18183388 DOI: 10.1007/s00253-007-1314-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 11/29/2007] [Accepted: 12/03/2007] [Indexed: 10/22/2022]
Abstract
Fungi metabolize polycyclic aromatic hydrocarbons by a number of detoxification processes, including the formation of sulfated and glycosidated conjugates. A class of aromatic compounds in grapefruit is the furanocoumarins (FCs), and their metabolism in humans is centrally involved in the "grapefruit/drug interactions." Thus far, the metabolism by fungi of the major FCs in grapefruit, including 6', 7'-epoxybergamottin (EB), 6', 7'-dihydroxybergamottin (DHB), and bergamottin (BM), has received little attention. In this study, Aspergillus niger was observed to convert EB into DHB and a novel water-soluble metabolite (WSM). Bergaptol (BT) and BM were also metabolized by A. niger to the WSM, which was identified as BT-5-sulfate using mass spectrometry, UV spectroscopy, chemical hydrolysis, and (1)H and (13)C nuclear magnetic resonance spectroscopy. Similarly, the fungus had a capability of metabolizing xanthotoxol (XT), a structural isomer of BT, to a sulfated analog of BT-5-sulfate, presumably XT-8-sulfate. A possible enzyme-catalyzed pathway for the grapefruit FC metabolism involving the cleavage of the geranyl group and the addition of a sulfate group is proposed.
Collapse
|
108
|
Furimsky AM, Green CE, Sharp LEH, Catz P, Adjei AA, Parman T, Kapetanovic IM, Weinshilboum RM, Iyer LV. Effect of resveratrol on 17beta-estradiol sulfation by human hepatic and jejunal S9 and recombinant sulfotransferase 1E1. Drug Metab Dispos 2008; 36:129-36. [PMID: 17954528 DOI: 10.1124/dmd.107.016725] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The purpose of this study was to investigate the sulfation of resveratrol (3,5,4'-trihydroxystilbene) and its potential to exhibit drug-drug interactions via sulfation. The possible interaction of resveratrol with 17beta-estradiol (E2), a major estrogen hormone and prototypic substrate for sulfate conjugation, was studied. Resveratrol and E2 are both known to undergo sulfate conjugation catalyzed by human sulfotransferases (SULTs). Resveratrol is a phytoestrogen with mixed estrogen agonist/antagonist properties that is being developed as a chemopreventive agent. The sulfate conjugation of E2 and resveratrol were studied individually using S9 fractions from human liver and jejunum as well as recombinant human SULT isoforms. The sulfation of E2 (3-20 nM) was then investigated in the presence of various concentrations (0, 0.5, 1, and 2 microM) of resveratrol using the two S9 preparations as well as recombinant SULT1E1, the major isoform responsible for E2 sulfation. Resveratrol inhibited E2 sulfation with estimated K(i) values of 1.1 microM (liver), 0.6 microM (jejunum), and 2.3 microM (SULT1E1), concentrations that could be pharmacologically relevant. The results suggest that these phytoestrogens can potentially alter the homeostasis of estrogen levels. These findings also imply that resveratrol may inhibit the metabolism of other estrogen analogs or therapeutic agents such as ethinylestradiol or dietary components that are also substrates for SULT1E1.
Collapse
Affiliation(s)
- Anna M Furimsky
- Toxicology and Metabolism, Biosciences Division, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Rouprêt M. Anatomical location of urothelial carcinomas of the urinary tract leads to perspectives of specific treatment. Future Oncol 2007; 3:595-9. [DOI: 10.2217/14796694.3.6.595] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Morgan Rouprêt
- Academic Urology Department, Hopital Pitié, Assistance Publique Hôpitaux de Paris, University Paris 6, 47–83 bvd de l’Hopital, 75013 Paris, France
| |
Collapse
|
110
|
Schumacher M, Liere P, Akwa Y, Rajkowski K, Griffiths W, Bodin K, Sjövall J, Baulieu EE. Pregnenolone sulfate in the brain: a controversial neurosteroid. Neurochem Int 2007; 52:522-40. [PMID: 18068870 DOI: 10.1016/j.neuint.2007.08.022] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 07/25/2007] [Accepted: 08/31/2007] [Indexed: 12/30/2022]
Abstract
Pregnenolone sulfate (PREGS) has been shown, either at high nanomolar or at micromolar concentrations, to increase neuronal activity by inhibiting GABAergic and by stimulating glutamatergic neurotransmission. PREGS is also a potent modulator of sigma type 1 (sigma1) receptors. It has been proposed that these actions of PREGS underlie its neuropharmacological effects, and in particular its influence on memory processes. On the other hand, the PREGS-mediated increase in neuronal excitability may become dangerous under particular conditions, for example in the case of excitotoxic stress or convulsions. However, the physiopathological significance of these observations has recently been put into question by the failure to detect significant levels of PREGS within the brain and plasma of rats and mice, either by direct analytical methods based on liquid chromatography/mass spectrometry (LC/MS) or enzyme linked immunosorbent assay (ELISA) with specific antibodies against PREGS, or by indirect gas chromatography/mass spectrometry (GC/MS) analysis with improved sample workup. These recent results have not come to the attention of a large number of neurobiologists interested in steroid sulfates. However, although available direct analytical methods have failed to detect levels of PREGS above 0.1-0.3 ng/g in brain tissue, it may be premature to completely exclude the local formation of biologically active PREGS within specific and limited compartments of the nervous system. In contrast to the situation in rodents, significant levels of sulfated 3beta-hydroxysteroids have been measured in human plasma and brain. Previous indirect measures of steroid sulfates by radioimmunoassays (RIA) or GC/MS had detected elevated levels of PREGS in rodent brain. The discrepancies between the results of different assay procedures have revealed the danger of indirect analysis of steroid sulfates. Indeed, PREGS must be solvolyzed/hydrolyzed prior to RIA or GC/MS analysis, and it is the released, unconjugated PREG which is then quantified. Extreme caution needs to be exercised during the preparation of samples for RIA or GC/MS analysis, because the fraction presumed to contain only steroid sulfates can be contaminated by nonpolar components from which PREG is generated by the solvolysis/hydrolysis/derivatization reactions.
Collapse
Affiliation(s)
- Michael Schumacher
- UMR 788 Inserm, University Paris-Sud 11, 80 rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | | | | | | | | | | | | | | |
Collapse
|
111
|
Riches Z, Bloomer JC, Coughtrie MWH. Comparison of 2-aminophenol and 4-nitrophenol as in vitro probe substrates for the major human hepatic sulfotransferase, SULT1A1, demonstrates improved selectivity with 2-aminophenol. Biochem Pharmacol 2007; 74:352-8. [PMID: 17506995 DOI: 10.1016/j.bcp.2007.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/03/2007] [Accepted: 04/04/2007] [Indexed: 10/23/2022]
Abstract
Sulfation, catalysed by members of the cytosolic sulfotransferase (SULT) enzyme family, is important in xenobiotic detoxification and in the biosynthesis and homeostasis of many hormones and neurotransmitters. The major human phenol sulfotransferase SULT1A1 plays a key role in chemical defence, is widely expressed in the body and is subject to a common polymorphism that results in reduced protein levels. Study of these enzymes in vitro requires robust probe substrates, and we have previously shown measurement of activity with the widely used SULT1A1 substrate, 4-nitrophenol, does not accurately reflect protein expression. Additionally, the high degree of substrate inhibition observed with this compound further reduces its value as a probe for SULT1A1. Here we show that 2-aminophenol is a more suitable probe substrate for quantifying SULT1A1 activity in human liver. This compound is sulfated at a high rate (V(max) with purified recombinant SULT1A1=121nmol/(minmg) and shows strong affinity for the enzyme (K(m) with purified recombinant SULT1A1=9microM) and, importantly, is a very poor substrate for the other major SULT1 enzyme expressed in liver, SULT1B1 (with V(max) and K(m) values of 17nmol/(minmg) and 114microM, respectively). Experiments with purified recombinant human SULTs and a panel of 28 human liver cytosols demonstrated that 2-aminophenol shows limited substrate inhibition with SULT1A1, and V(max) values measured in liver cytosols correlated strongly with SULT1A1 enzyme protein levels measured by a quantitative immunoblot method. We therefore suggest that 2-aminophenol is a suitable substrate to use for quantifying SULT1A1 enzyme activity.
Collapse
Affiliation(s)
- Zoe Riches
- Division of Pathology & Neuroscience, University of Dundee, Ninewells Hospital & Medical School, Dundee DD1 9SY, Scotland, UK
| | | | | |
Collapse
|
112
|
Ung D, Nagar S. Variable sulfation of dietary polyphenols by recombinant human sulfotransferase (SULT) 1A1 genetic variants and SULT1E1. Drug Metab Dispos 2007; 35:740-6. [PMID: 17293380 DOI: 10.1124/dmd.106.013987] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human cytosolic sulfotransferases (SULTs) catalyze the sulfate conjugation of several important endo- and xenobiotics. Among the superfamily of SULT enzymes, SULT1A1 catalyzes the sulfation of small planar phenolic compounds, whereas SULT1E1 has a major role in estrogen conjugation. The human SULT1A1 gene has common single nucleotide polymorphisms that define three allozymes, SULT1A1*1, *2, and *3. The enzyme kinetics of SULT1A1 allozymes and SULT1E1 were characterized for the polyphenolic substrates apigenin, chrysin, epicatechin, quercetin, and resveratrol. Purified recombinant SULT proteins were generated in a baculoviral-insect cell system, and incubated in vitro with each substrate to determine catalytic activity. The effect of polyphenol sulfation was examined in mammalian cell lines stably expressing SULT1E1. For all polyphenols investigated, "normal-activity" SULT1A1*1 allozyme had significantly greater Vmax estimates than SULT1E1, and allele-specific differences in SULT1A1-mediated sulfation were observed. The polymorphic SULT1A1*2 allozyme exhibited low activity toward apigenin, epicatechin, and resveratrol. SULT1A1*1 and *3 acted as normal-activity allozymes for these substrates. Altered cellular proliferation was observed in MCF-7 cells stably expressing SULT1E1 upon treatment with chrysin, quercetin, or resveratrol, thus suggesting inactivation of these compounds by SULT1E1. These results suggest an important role for SULT isozymes and their pharmacogenetics in polyphenol disposition.
Collapse
Affiliation(s)
- Din Ung
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad St., Philadelphia, PA 19140, USA
| | | |
Collapse
|
113
|
Gershon E, Hourvitz A, Reikhav S, Maman E, Dekel N. Low expression of COX-2, reduced cumulus expansion, and impaired ovulation in SULT1E1-deficient mice. FASEB J 2007; 21:1893-901. [PMID: 17341680 DOI: 10.1096/fj.06-7688com] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The SULT1E1-encoded estrogen sulfotransferase (EST) catalyzes sulfation of estrogen, resulting in its inactivation. Reduced fertility observed in SULT1E1 knockout (KO) female mice has previously been attributed to the deleterious effect of chronic exposure to high levels of circulating estrogen on placental function. We herein suggest that, in addition to placental dysfunction, this phenotype demonstrates that an excess of estrogen impairs ovulation. The role of SULT1E1 in ovulation is suggested by the substantially low ovulatory response in hCG-treated SULT1E1 KO mice; a similar effect was observed when 17beta-estradiol was administered to wild-type (WT) females. The normal rate of ovulation in SULT1E1 KO females may be restored by PGE2. Along this line, ovaries of human Chorionic Gonadotropin (hCG)-treated SULT1E1 KO mice expressed low levels of cyclooxygenase-2 (COX-2) and its downstream TSG6; moreover, their ovaries contained a reduced number of expanded cumuli. Our results demonstrate, for the first time, that estrogen inactivation may allow the expression of COX-2 and subsequent cumulus expansion, enabling normal ovulation. Our findings may be applied to novel treatments of human ovulatory failure.
Collapse
Affiliation(s)
- Eran Gershon
- Department of Biological Regulation, Weizmann Institute of Science, P.O.B. 26, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
114
|
Hebbring SJ, Adjei AA, Baer JL, Jenkins GD, Zhang J, Cunningham JM, Schaid DJ, Weinshilboum RM, Thibodeau SN. Human SULT1A1 gene: copy number differences and functional implications. Hum Mol Genet 2007; 16:463-70. [PMID: 17189289 DOI: 10.1093/hmg/ddl468] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
SULT1A1, which catalyzes the sulfate conjugation of a wide variety of natural and synthetic compounds, is genetically polymorphic. Biochemical and pharmacogenetic studies have demonstrated that individual variation in the level of enzyme activity is inherited. Common single-nucleotide polymorphisms (SNPs) located in the open reading frame and in the 5'-flanking region (5'-FR) may account for a portion of this individual variation. In this study, we demonstrate the presence of SULT1A1 gene deletions and duplications, representing an additional source of variability in the metabolic activity of this enzyme. A quantitative multiplex PCR assay was used to measure the extent of copy number differences and the frequency of these events in different populations. An analysis of DNA from 362 Caucasian-American and 99 African-American showed the presence of 1 to approximately 5 copies of SULT1A1 in individual samples: 5% of Caucasian subjects contained a single copy of the gene and 26% had three or more copies, while 63% of African-American subjects had three or more copies. Analysis of the genomic region surrounding the SULT1A1 gene in three separate cases with a deletion demonstrated that the entire SULT1A1 gene was affected. Reporter assays, constructed for each of the various 5'-FR SNP haplotypes, suggest that these may also play a role in SULT1A1 activity. However, the variability in the level of enzyme activity among 23 human platelet and 267 human liver samples was best explained by gene copy number differences when all sources of genetic variability were considered (P < 0.0001). Overall, these observations have obvious implications for the effectiveness of SULT1A1 as a drug and hormone metabolizing enzyme and its potential role as a risk factor for disease.
Collapse
Affiliation(s)
- Scott J Hebbring
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Brandon EFA, Sparidans RW, van Ooijen RD, Meijerman I, Lazaro LL, Manzanares I, Beijnen JH, Schellens JHM. In vitro characterization of the human biotransformation pathways of aplidine, a novel marine anti-cancer drug. Invest New Drugs 2007; 25:9-19. [PMID: 16633717 DOI: 10.1007/s10637-006-7589-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Aplidine is a potent marine anti-cancer drug and is currently being investigated in phase II clinical trials. However, the enzymes involved in the biotransformation of aplidine and thus its pharmacokinetics are not known yet. To assess the biotransformation pathways of aplidine and their potential implications for human pharmacology and toxicology, the in vitro metabolism of aplidine was characterized using incubations with human plasma, liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes in combination with HPLC analysis and cytotoxicity assays with cell lines. Aplidine was metabolised by carboxyl esterases in human plasma. Using CYP supersomes and liver microsomes, it was shown that aplidine was metabolised mainly by CYP3A4 and also by CYP2A6, 2E1 and 4A11. Four metabolites were observed after incubation with human liver microsomes, one formed by CYP2A6 (C-demethylation) and three by CYP3A4 (hydroxylation and/or C-dealkylation). No conjugation was observed in human liver S9 fraction. However, the aplidine metabolites formed by CYP were further conjugated by the phase II enzymes UGT, GST and SULT. In accordance with the findings in microsomes and CYP supersomes, a significant effect of specific CYP2A6, 2E1, 3A4 and 4A11 inhibitors on the cytotoxicity of aplidine in Hep G2 and IGROV-1 cells could be observed. These results provide evidence that CYP3A4 has a major role in metabolising aplidine in vitro with additional involvement of CYP2A6, 2E1, and 4A11. Further, the metabolites formed by CYPs can be conjugated by UGT, SULT and GST. These findings could help interpret the in vivo pharmacokinetics of aplidine.
Collapse
Affiliation(s)
- Esther F A Brandon
- Department of Pharmaceutical Sciences, Section of Biomedical Analysis, Division of Drug Toxicology, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Najmanovich RJ, Allali-Hassani A, Morris RJ, Dombrovsky L, Pan PW, Vedadi M, Plotnikov AN, Edwards A, Arrowsmith C, Thornton JM. Analysis of binding site similarity, small-molecule similarity and experimental binding profiles in the human cytosolic sulfotransferase family. Bioinformatics 2007; 23:e104-9. [PMID: 17237076 DOI: 10.1093/bioinformatics/btl292] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
MOTIVATION In the present work we combine computational analysis and experimental data to explore the extent to which binding site similarities between members of the human cytosolic sulfotransferase family correlate with small-molecule binding profiles. Conversely, from a small-molecule point of view, we explore the extent to which structural similarities between small molecules correlate to protein binding profiles. RESULTS The comparison of binding site structural similarities and small-molecule binding profiles shows that proteins with similar small-molecule binding profiles tend to have a higher degree of binding site similarity but the latter is not sufficient to predict small-molecule binding patterns, highlighting the difficulty of predicting small-molecule binding patterns from sequence or structure. Likewise, from a small-molecule perspective, small molecules with similar protein binding profiles tend to be topologically similar but topological similarity is not sufficient to predict their protein binding patterns. These observations have important consequences for function prediction and drug design.
Collapse
Affiliation(s)
- Rafael J Najmanovich
- European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus Cambridge CB10 1SD, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Maiti S, Zhang J, Chen G. Redox regulation of human estrogen sulfotransferase (hSULT1E1). Biochem Pharmacol 2006; 73:1474-81. [PMID: 17266938 PMCID: PMC1950446 DOI: 10.1016/j.bcp.2006.12.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 12/19/2006] [Accepted: 12/20/2006] [Indexed: 10/23/2022]
Abstract
Sulfotransferases (SULTs) are enzymes that catalyze the sulfation of hydroxyl-containing compounds. Sulfation regulates hormone activities and detoxifies xenobiotics. Human estrogen sulfotransferase (hSULT1E1) catalyzes the sulfation of estrogens and regulates estrogen bioactivities. Oxidative regulation provides a biological mechanism for regulating enzyme activities in vivo. The oxidative regulation of human SULTs has not been reported. In this study, we used amino acid modification, manipulation of intracellular redox state, and site-directed mutagenesis to study the redox regulation of human SULTs and specifically the mechanism of hSULT1E1 inhibitory regulation by oxidized glutathione (GSSG). Of the four major human SULTs, hSULT1A1, hSULT1A3, and hSULT2A1 do not undergo redox regulation; hSULT1E1, on the other hand, can be redox regulated. GSSG inactivated hSULT1E1 activity in an efficient, time- and concentration-dependant manner. The co-enzyme adenosine 3'-phosphate 5'-phosphosulfate protected hSULT1E1 from GSSG-associated inactivation. A reduced glutathione (GSH) inducer (N-acetyl cysteine) significantly increased while a GSH depletor (buthionine sulfoxamine) significantly decreased hSULT1E1 activity, but both failed to affect the amount of hSULT1E1 protein in human hepatocyte carcinoma Hep G2 cells. Crystal structure suggested that no Cys residues exist near the active sites of hSULT1A1, hSULT1A3, and hSULT2A1, but Cys residues do exist within the active site of hSULT1E1. Site-directed mutagenesis demonstrated that Cys83 is critical for the redox regulation of hSULT1E1. This first report on the redox regulation of human SULTs suggests that the redox regulation of hSULT1E1 may interrupt the regulation and function of estrogens under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Smarajit Maiti
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Jimei Zhang
- Department of Chemical Engineering, Tianjin Polytechnic University, China
| | - Guangping Chen
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, USA
- Address correspondence to: Guangping Chen, 264 McElroy Hall, Oklahoma State University, Stillwater, OK, 74078. Phone: (405) 744-2349. Fax: (405) 744-8263. E-mail:
| |
Collapse
|
118
|
Chen X, Zhang J, Baker SM, Chen G. Human constitutive androstane receptor mediated methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1). Toxicology 2006; 231:224-33. [PMID: 17276571 PMCID: PMC1919471 DOI: 10.1016/j.tox.2006.12.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 11/15/2006] [Accepted: 12/05/2006] [Indexed: 01/01/2023]
Abstract
Sulfotransferases (SULTs) catalyzed sulfation is important in the regulation of biological activities of hormones and neurotransmitters, the metabolism of drugs, and the detoxification of xenobiotic toxicants. Sulfation also leads to the bioactivation of procarcinogens. Human dehydroepiandrosterone sulfotransferase (hSULT2A1) is a major SULT catalyzing the sulfation of hydroxysteroids and xenobiotic alcohols. Our previous studies had shown that the anti-folate drug methotrexate (MTX) can up-regulate several major isoforms of human SULTs. To determine the mechanisms controlling the regulation of hSULT2A1, the 5'-flanking region of hSULT2A1 was constructed into the pGL3-Basic luciferase reporter vector. The transcriptional regulation mechanism of hSULT2A1 promoter was studied using Caco-2 cell line based on the reporter gene assay. Nuclear receptor co-transfection results indicated that human constitutive androstane receptor (hCAR) and human retinoid X receptor alpha (hRXRalpha) were involved in the transcriptional regulation of hSULT2A1. RNA interference experiments further proved the role of hCAR in hSULT2A1 regulation. Progressive promoter deletion, DNA sequence alignment, and site directed promoter mutation results suggested that an imperfect inverted repeat DNA motif, IR2 (-186AGCTCAGATGACCC-173), within the hSULT2A1 promoter region mediated the hSULT2A1 induction by MTX. Furthermore, electrophoretic mobility shift assay and super shift assay were employed to characterize the interactions of hCAR and hRXRalpha with the IR2 element. In summary, we identified an IR2 DNA cis-element located at -186/-173 of hSULT2A1 promoter region; the IR2 element mediates the MTX induction of hSULT2A1 through interacting with hCAR and hRXRalpha.
Collapse
Affiliation(s)
- Xinrong Chen
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | |
Collapse
|
119
|
Anderson LM. Environmental genotoxicants/carcinogens and childhood cancer: Bridgeable gaps in scientific knowledge. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2006; 608:136-56. [PMID: 16829162 DOI: 10.1016/j.mrgentox.2006.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 02/13/2006] [Indexed: 10/24/2022]
Abstract
Cancer in children is a major concern in many countries. An important question is whether these childhood cancers are caused by something, or are just tragic random events. Causation of at least some children's cancers is suggested by direct and indirect evidence, including epidemiological data, and animal studies that predict early life sensitivity of humans to carcinogenic effects. Candidate risk factors include genotoxic agents (chemicals and radiation), but also diet/nutrition, and infectious agents/immune responses. With regard to likelihood of risks posed by genotoxicants, there are pros and cons. The biological properties of fetuses and infants are consistent with sensitivity to preneoplastic genotoxic damage. Recent studies of genetic polymorphisms in carcinogen-metabolizing enzymes confirm a role for chemicals. On the other hand, in numerous epidemiological studies, associations between childhood cancers and exposure to genotoxicants, including tobacco smoke, have been weak and hard to reproduce. Possibly, sensitive genetic or ontogenetic subpopulations, and/or co-exposure situations need to be discovered to allow identification of susceptible individuals and their risk factors. Among the critical knowledge gaps needing to be bridged to aid in this effort include detailed tissue and cellular ontogeny of carcinogen metabolism and DNA repair enzymes, and associations of polymorphisms in DNA repair enzymes with childhood cancers. Perinatal bioassays in animals of specific environmental candidates, for example, benzene, could help guide epidemiology. Genetically engineered animal models could be useful for identification of chemical effects on specific genes. Investigations of interactions between factors may be key to understanding risk. Finally, fathers and newborn infants should receive more attention as especially sensitive targets.
Collapse
Affiliation(s)
- Lucy M Anderson
- Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| |
Collapse
|
120
|
Martin-Skilton R, Coughtrie MWH, Porte C. Sulfotransferase activities towards xenobiotics and estradiol in two marine fish species (Mullus barbatus and Lepidorhombus boscii): characterization and inhibition by endocrine disrupters. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2006; 79:24-30. [PMID: 16806523 DOI: 10.1016/j.aquatox.2006.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 04/25/2006] [Accepted: 04/27/2006] [Indexed: 05/10/2023]
Abstract
We have characterized hepatic phenol sulfotransferase (SULT) activities in two benthic fish species, Mullus barbatus and Lepidorhombus boscii, using p-nitrophenol, dopamine, 17beta-estradiol, 4-nonylphenol, and 1-naphthol as substrates. High affinity sulfation of 17beta-estradiol was observed in both species (Km=28-75 nM), suggesting the presence of a specific estrogen sulfotransferase that catalyzes the formation of estradiol-3 sulfate. Among the tested compounds, 1-naphthol was the most effective substrate for sulfation, with Vmax/Km ratios several hundred-fold higher than the other substrates examined. Both species sulfated the tested compounds, but only M. barbatus was able to sulfate dopamine. We also tested the inhibitory effects of common marine pollutants with estrogenic (4-nonylphenol) and androgenic (tributyltin, triphenyltin) properties on p-nitrophenol and 17beta-estradiol SULT activities. 4-Nonylphenol and triphenyltin inhibited sulfation of both substrates at micromolar concentrations in both species. However, tributyltin was only effective against SULTs from L. boscii, again at micromolar concentrations. The data indicate that M. barbatus and L. boscii are able to sulfate a range of xenobiotics and endogenous compounds, and inhibition of these activities by environmental pollutants may contribute to the known toxic effects of these compounds.
Collapse
Affiliation(s)
- Rebeca Martin-Skilton
- Environmental Chemistry Department, IIQAB-CSIC, C/Jordi Girona 18, 08034 Barcelona, Spain
| | | | | |
Collapse
|
121
|
Abstract
Sulfotransferases (Sults) are phase-II conjugation enzymes that catalyze the transfer of a sulfonate group from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to target endo and xenobiotics. PAPS is formed from inorganic sulfate by the action of the enzyme PAPS synthase (PAPSs). In the present study, the tissue distribution and developmental changes in the mRNA expression of 11 Sult isozymes and 2 PAPSs isoforms in mice were quantified. Sult1a1, 1b1, 1c1, 1c2, 1d1, 1e1, 2a1/2, 2b1, 3a1, 4a1, 5a1, PAPSs1, and PAPSs2 mRNA expression was quantified in 14 tissues from male and female mice using the branched DNA signal amplification assay. Sult2a1/2 and 3a1 expression were highest in liver; Sult1b1, 2b1, and PAPSs2 in small intestine; Sult1a1 in large intestine; Sult1c2 in stomach; Sult1d1 in kidney; Sult1e1 in placenta; and Sult4a1 in brain. Sult1c1, 5a1, and PAPSs1 were ubiquitously expressed in most tissues. These enzymes demonstrated three different ontogenic expression patterns in liver. Sult1a1, 1c2, 1d1, 2a1/2, and PAPSs2 hepatic expression gradually increased from birth until about 3 weeks of age and then declined somewhat thereafter, Sult1c1 expression was highest before birth and declined after that, and Sult3a1 mRNA expression was very low in fetal livers and remained low until 30 days of age, when expression in females dramatically increased, whereas it never increased in males. The organ-specific distribution of Sults as well as the different expression of the Sults in young animals may affect the pharmacokinetic behavior and organ-specific toxicity of xenobiotics.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | |
Collapse
|
122
|
Hildebrandt MAT, Carrington DP, Thomae BA, Eckloff BW, Schaid DJ, Yee VC, Weinshilboum RM, Wieben ED. Genetic diversity and function in the human cytosolic sulfotransferases. THE PHARMACOGENOMICS JOURNAL 2006; 7:133-43. [PMID: 16801938 DOI: 10.1038/sj.tpj.6500404] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Amino-acid substitutions, which result from common nonsynonymous (NS) polymorphisms, may dramatically alter the function of the encoded protein. Gaining insight into how these substitutions alter function is a step toward acquiring predictability. In this study, we incorporated gene resequencing, functional genomics, amino-acid characterization and crystal structure analysis for the cytosolic sulfotransferases (SULTs) to attempt to gain predictability regarding the function of variant allozymes. Previously, four SULT genes were resequenced in 118 DNA samples. With additional resequencing of the remaining eight SULT family members in the same DNA samples, a total of 217 polymorphisms were revealed. Of 64 polymorphisms identified within 8785 bp of coding regions from SULT genes examined, 25 were synonymous and 39 were NS. Overall, the proportion of synonymous changes was greater than expected from a random distribution of mutations, suggesting the presence of a selective pressure against amino-acid substitutions. Functional data for common variants of five SULT genes have been previously published. These data, together with the SULT1A1 variant allozyme data presented in this paper, showed that the major mechanism by which amino acid changes altered function in a transient expression system was through decreases in immunoreactive protein rather than changes in enzyme kinetics. Additional insight with regard to mechanisms by which NS single nucleotide polymorphisms alter function was sought by analysis of evolutionary conservation, physicochemical properties of the amino-acid substitutions and crystal structure analysis. Neither individual amino-acid characteristics nor structural models were able to accurately and reliably predict the function of variant allozymes. These results suggest that common amino-acid substitutions may not dramatically alter the protein structure, but affect interactions with the cellular environment that are currently not well understood.
Collapse
Affiliation(s)
- M A T Hildebrandt
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Mayo Foundation, Rochester, MN 55985, USA
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Nagar S, Walther S, Blanchard RL. Sulfotransferase (SULT) 1A1 polymorphic variants *1, *2, and *3 are associated with altered enzymatic activity, cellular phenotype, and protein degradation. Mol Pharmacol 2006; 69:2084-92. [PMID: 16517757 DOI: 10.1124/mol.105.019240] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The superfamily of sulfotransferase (SULT) enzymes catalyzes the sulfate conjugation of several pharmacologically important endo- and xenobiotics. SULT1A1 catalyzes the sulfation of small planar phenols such as neurotransmitters, steroid hormones, acetaminophen, and p-nitrophenol (PNP). Genetic polymorphisms in the human SULT1A1 gene define three alleles, SULT1A1*1, *2, and *3. The enzyme activities of the SULT1A1 allozymes were studied with a variety of substrates, including PNP, 17beta-estradiol, 2-methoxyestradiol, catecholestrogens, the antiestrogen 4-hydroxytamoxifen (OHT), and dietary flavonoids. Using purified recombinant SULT1A1 protein, marked differences in *1, *2, and *3 activity toward every substrate studied were noted. Substrate inhibition was observed for most substrates. In general, the trend in V(max) estimates was *1 > *3 > *2; however, V(max)/K(m) estimate trends varied with substrate. In MCF-7 cells stably expressing either SULT1A1*1 or *2, the antiestrogenic response to OHT was found to be allele-specific: the cells expressing *2 exhibited a better antiproliferative response. The intracellular stability of the *1 and *2 allozymes was examined in insect as well as mammalian cells. The SULT1A1*2 protein had a shorter half-life than the *1 protein. In addition, the *2 protein was ubiquitinated to a greater extent than *1, suggesting increased degradation via a proteasome pathway. The results of this study suggest marked differences in activity of polymorphic SULT1A1 variants, including SULT1A1*3, toward a variety of substrates. These differences are potentially critical for understanding interindividual variability in drug response and toxicity, as well as cancer risk and incidence.
Collapse
Affiliation(s)
- Swati Nagar
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | |
Collapse
|
124
|
Aitken AE, Richardson TA, Morgan ET. Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev Pharmacol Toxicol 2006; 46:123-49. [PMID: 16402901 DOI: 10.1146/annurev.pharmtox.46.120604.141059] [Citation(s) in RCA: 350] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inflammation and infection have long been known to downregulate the activity and expression of cytochrome P450 (CYP) enzymes involved in hepatic drug clearance. This can result in elevated plasma drug levels and increased adverse effects. Recent information on regulation of human CYP enzymes is presented, as are new developments in our understanding of the mechanisms of regulation. Experiments to study the effects of modulating CYP activities on the inflammatory response have yielded possible insights into the physiological consequences, if not the purpose, of the downregulation. Regulation of hepatic flavin monooxygenases, UDP-glucuronosyltransferases, sulfotransferases, glutathione S-transferases, as well as of hepatic transporters during the inflammatory response, exhibits similarities and differences with regulation of CYPs.
Collapse
Affiliation(s)
- Alison E Aitken
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | |
Collapse
|
125
|
Chen X, Maiti S, Zhang J, Chen G. Nuclear receptor interactions in methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1). J Biochem Mol Toxicol 2006; 20:309-17. [PMID: 17163485 DOI: 10.1002/jbt.20149] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cytosolic sulfotransferases (SULTs) are a major family of phase II drug-metabolizing enzymes. SULT-catalyzed sulfonation regulates hormone activities metabolizes drugs detoxifies xenobiotic toxicants bioactivates carcinogens. Human dehydroepiandrosterone sulfotransferase (hSULT2A1 DHEA-ST) plays a very important role in sulfating endogenous hydroxysteroids and exogenousxenobiotics. Our recent studies have shown that methotrexate can induce hSULT2A1 expression. To investigate the molecular mechanism involved in hSULT2A1 induction we generated the promoter sequence of hSULT2A1 by PCR and constructed a reporter gene vector. Both reporter gene assay and endogenous induction results suggested that human constitutive active receptor (hCAR) mediates the methotrexate induction of hSULT2A1 in both Caco-2 and Hep G2 cells. Human vitamin D receptor (hVDR) also upregulated hSULT2A1 gene expression while human pregnane X receptor (hPXR) downregulated it. Human pregnane X receptor suppressed hCAR-mediated methotrexate induction of hSULT2A1 in both Caco-2 and Hep G2 cells. hVDR competed with hCAR for the hSULT2A1 promoter in Caco-2 cells. hCAR inhibited hVDR-mediated vitamin D3 induction of hSULT2A1 but not methotrexate induction of hSULT2A1. These results strongly support the hypothesis that cross-talk occurs among nuclear receptors in the signal transduction pathway of hSULT2A1 and that interactions among nuclear receptors also depend on ligands (inducers) in the system.
Collapse
Affiliation(s)
- Xinrong Chen
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | |
Collapse
|
126
|
Yang G, Gao YT, Cai QY, Shu XO, Cheng JR, Zheng W. Modifying Effects of Sulfotransferase 1A1 Gene Polymorphism on the Association of Breast Cancer Risk with Body Mass Index or Endogenous Steroid Hormones. Breast Cancer Res Treat 2005; 94:63-70. [PMID: 16175316 DOI: 10.1007/s10549-005-7280-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Sulfotransferase (SULT) 1A1 is involved in the inactivation and elimination of estrogens and catechol estrogens. A common functional polymorphism (Arg213His) has been linked in our previous study of postmenopausal Caucasian women to an elevated risk of breast cancer and the association appeared to be modified by factors related to high endogenous estrogen exposures. We further evaluated this polymorphism and levels of BMI and steroid hormones in association with breast cancer risk in a population-based case-control study of Chinese women, involving 1102 incident cases aged 25-64 years and 1147 age-matched population controls. The SULT1A1 genotype was not associated with overall breast cancer risk in this population. A possible association was suggested for postmenopausal breast cancer (adjusted odds ratio [OR] = 1.4, 95% CI = 0.9-2.1 for subject carrying the variant His allele). The SULT1A1 genotype was found to significantly modify postmenopausal breast cancer risk associated with a high BMI (>or=25 kg/m2) (p for interaction = 0.02), with an adjusted OR of 3.6 (95% CI = 1.5-8.7) for women with the Arg/His genotype compared with 1.1 (0.8-1.5) for women with the Arg/Arg genotype (no His/His genotype was identified in this study population). Similarly, the risk associated with a long duration (>or=30 years) of menstruation also substantially differed by the SULT1A1 genotype (p for interaction = 0.05), with an OR of 4.0 (95% CI = 1.3-12.8) for women with the Arg/His genotype and 1.4 (0.8-2.5) for women with the Arg/Arg genotype. Positive associations with blood levels of steroid hormones were also found generally to be more pronounced among women carrying the His allele. No similar effect modification was found for premenopausal breast cancer, however. These data suggest that the SULT1A1 Arg213His polymorphism may modify the effect of endogenous sex hormone exposures on postmenopausal breast cancer risk.
Collapse
Affiliation(s)
- Gong Yang
- Department of Medicine, Center for Health Services Research and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232-8300, USA.
| | | | | | | | | | | |
Collapse
|
127
|
Aust S, Jaeger W, Klimpfinger M, Mayer K, Baravalle G, Ekmekcioglu C, Thalhammer T. Biotransformation of melatonin in human breast cancer cell lines: role of sulfotransferase 1A1. J Pineal Res 2005; 39:276-82. [PMID: 16150108 DOI: 10.1111/j.1600-079x.2005.00246.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The biologically active melatonin metabolite, 6-hydroxymelatonin (6-OHMel), is conjugated to form 6-hydroxymelatonin sulfate (6-OHMelS). To elucidate the role of the sulfotransferase (SULT) enzyme 1A1, considerably expressed in normal and malignant human breast cells, we measured the formation of 6-OHMelS by ELISA in hormone-dependent MCF-7 and hormone-independent MDA-MB231 (MDA) breast cancer cell lines after stable transfection with SULT1A1. In parent MDA cells, low SULT1A1 mRNA expression was associated with moderate 6-OHMelS formation as determined after application (24 hr) of 0.1 microM 6-OHMel. As expected, overexpression of SULT1A1 in MDA cells resulted in a 2.9- and 110-fold increase in 6-OHMelS in the cytosol and cellular supernatant respectively. Furthermore, 6.3- and 115-fold increases were observed after 0.5 microM, and 12.6- and 101-fold increases after 1 microM 6-OHMel respectively. In MCF-7 cells, because of high basal SULT1A1 expression, only two- to threefold increases in 6-OHMelS were observed after transfection with the enzyme. In total, 866 and 539 pmol/mg protein 6-OHMelS were formed from 1 microM 6-OHMel in SULT1A1 overexpressing MDA and MCF-7 cells, respectively, whereas application of 1 microM melatonin produced only <1% of 6-OHMelS. Possible interactions with the SULT1A1 substrate tamoxifen (tam), an anti-estrogen applied in the therapy of breast cancer, were also studied. A concentration of 1 microM tam increased 6-OHMelS formation by approximately threefold in the presence of 1 microM melatonin or 1 microM 6-OHMel respectively. However, no alterations were detected after application of 1 microM 4-hydroxy-tamoxifen. In summary, we demonstrate the importance of SULT1A1 for the biotransformation of 6-OHMel in human breast cancer cells. Our data further suggest that tam can modulate melatonin biotransformation.
Collapse
Affiliation(s)
- Sylvia Aust
- Center for Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
128
|
Stanley EL, Hume R, Coughtrie MWH. Expression profiling of human fetal cytosolic sulfotransferases involved in steroid and thyroid hormone metabolism and in detoxification. Mol Cell Endocrinol 2005; 240:32-42. [PMID: 16024168 DOI: 10.1016/j.mce.2005.06.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2005] [Revised: 06/01/2005] [Accepted: 06/09/2005] [Indexed: 10/25/2022]
Abstract
Protection against chemical insult is essential for normal development of the fetus, however many detoxification enzymes are poorly expressed during fetal development. A major exception is the sulfotransferase (SULT) family, which appears to be widely expressed in the developing human. These enzymes also play a key role in biosynthesis and homeostasis of a number of hormones, including estrogens and iodothyronines. We therefore examined the enzyme activity, protein and mRNA expression of SULT 1A, 1B, 1C, 1E and 2A families in a variety of human fetal and adult tissues. Our results show that these SULTs are expressed in the human fetus, with most present at levels equivalent to or higher than the adult. As there are no isoform-selective substrates for SULTs 1B1 and 1C2 we used immunoblot analysis to show for the first time expression of SULT1B1 at high levels in fetal small intestine, and expression of SULT1C2 in fetal liver, kidney and small intestine. SULT1C2 was not expressed in adult liver or colon. Sulfotransferase expression in the developing fetus is therefore more widespread than in the adult, and this has significant implication for our understanding of human developmental physiology.
Collapse
Affiliation(s)
- Emma L Stanley
- Division of Pathology & Neuroscience, University of Dundee, Ninewells Hospital & Medical School, Scotland, UK
| | | | | |
Collapse
|
129
|
Maiti S, Dutta SM, Baker SM, Zhang J, Narasaraju T, Liu L, Chen G. In vivo and in vitro oxidative regulation of rat aryl sulfotransferase IV (AST IV). J Biochem Mol Toxicol 2005; 19:109-18. [PMID: 15849721 DOI: 10.1002/jbt.20064] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sulfotransferase catalyzed sulfation is important in the regulation of different hormones and the metabolism of hydroxyl containing xenobiotics. In the present investigation, we examined the effects of hyperoxia on aryl sulfotransferase IV in rat lungs in vivo. The enzyme activity of aryl sulfotransferase IV increased 3- to 8-fold in >95% O2 treated rat lungs. However, hyperoxic exposure did not change the mRNA and protein levels of aryl sulfotransferase IV in lungs as revealed by Western blot and RT-PCR. This suggests that oxidative regulation occurs at the level of protein modification. The increase of nonprotein soluble thiol and reduced glutathione (GSH)/oxidized glutathione (GSSG) ratios in treated lung cytosols correlated well with the aryl sulfotransferase IV activity increase. In vitro, rat liver cytosol 2-naphthol sulfation activity was activated by GSH and inactivated by GSSG. Our results suggest that Cys residue chemical modification is responsible for the in vivo and in vitro oxidative regulation. The molecular modeling structure of aryl sulfotransferase IV supports this conclusion. Our gel filtration chromatography results demonstrated that neither GSH nor GSSG treatment changed the existing aryl sulfotransferase IV dimer status in cytosol, suggesting that oxidative regulation of aryl sulfotransferase IV is not caused by dimer-monomer status change.
Collapse
Affiliation(s)
- Smarajit Maiti
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | | | | | | | |
Collapse
|
130
|
Schneider H, Glatt H. Sulpho-conjugation of ethanol in humans in vivo and by individual sulphotransferase forms in vitro. Biochem J 2005; 383:543-9. [PMID: 15281911 PMCID: PMC1133748 DOI: 10.1042/bj20040925] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We studied whether ethanol is sulphonated in humans with the perspective of using the urinary excretion of ethyl sulphate after ethanol consumption as a biomarker for SULT (sulphotransferase) activity. We developed a sensitive and selective HPLC-MS/MS method for determining ethyl sulphate in urine. Ten volunteers received a low dose of ethanol (0.1 g/kg of body mass). In general, excretion of ethyl sulphate was maximal in the first or second hour after dosage. Within 8 h, 2.5-6.8 micromol of ethyl sulphate was excreted. A 5-fold increase in the dose of ethanol led to an increase in the amount of ethyl sulphate excreted within 8 h (28-95 micromol) and the presence of this metabolite in urine for at least 24 h. Since ethyl sulphate was still being excreted for a substantial period after the elimination of ethanol, it might be used as a medium-time biomarker for preceding ethanol consumption. We have expressed previously all human SULT forms identified in Salmonella typhimurium. Ethanol sulphonation was studied in cytosolic preparations of these strains. The highest activities were observed with SULT1A2, 1B1 and 1C2, followed by 1A3. Activities were markedly lower with SULT1E1, 1A1 and 2A1, and were negligible with SULT1C1, 2B1a, 2B1b and 4A1. If the expression levels in tissues are additionally taken into account, SULT1A3 might be the predominant form for the sulphonation of ethanol in vivo, although a robust estimate requires further studies. With this limitation, urinary ethyl sulphate excretion appears very promising as a biomarker for SULT activity in vivo.
Collapse
Affiliation(s)
- Heiko Schneider
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Department of Toxicology, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Hansruedi Glatt
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Department of Toxicology, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
131
|
He D, Frost AR, Falany CN. Identification and immunohistochemical localization of Sulfotransferase 2B1b (SULT2B1b) in human lung. Biochim Biophys Acta Gen Subj 2005; 1724:119-26. [PMID: 15878639 DOI: 10.1016/j.bbagen.2005.03.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 03/11/2005] [Accepted: 03/21/2005] [Indexed: 11/29/2022]
Abstract
Sulfotransferase 2B1b (SULT2B1b) is a member of SULT 2 gene family. SULT2B1a and SULT2B1b are transcribed from the same gene using different transcriptional start sites and contain different first exons as the result of alternative splicing. SULT2B1a and SULT2B1b are 350 and 365 AA in length, respectively. Northern blot analysis and SULT2B1 isoform specific RT-PCR detected only SULT2B1b message in human lung tissue. Immunoblot analysis of human lung tissue with a specific rabbit anti-SULT2B1 polyclonal antibody detected only SULT2B1b immunoreactive protein in human lung cytosol. Immunoprecipitation and MALDI mass spectroscopic analysis verified that the immunoreactive protein was SULT2B1b. Immunohistochemical localization of SULT2B1b in human tissues showed expression in the cytoplasm of ciliated columnar or cuboidal epithelial cells in terminal bronchia. No staining was observed in alveolar cells. SULT2B1b is selective for the sulfation of 3beta-hydroxysteroids such as dehydroepiandrosterone and pregnenolone as well as cholesterol. The presence of SULT2B1b in lung tissues suggests a role in the regulation of local steroid hormone synthesis and metabolism.
Collapse
Affiliation(s)
- Dongning He
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall, Room G133M, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
132
|
Schwonbeck S, Krause-Griep A, Gajovic-Eichelmann N, Ehrentreich-Förster E, Meinl W, Glatt H, Bier FF. Cohort analysis of a single nucleotide polymorphism on DNA chips. Biosens Bioelectron 2005; 20:956-66. [PMID: 15530792 DOI: 10.1016/j.bios.2004.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A method has been developed to determine SNPs on DNA chips by applying a flow-through bioscanner. As a practical application we demonstrated the fast and simple SNP analysis of 24 genotypes in an array of 96 spots with a single hybridisation and dissociation experiment. The main advantage of this methodical concept is the parallel and fast analysis without any need of enzymatic digestion. Additionally, the DNA chip format used is appropriate for parallel analysis up to 400 spots. The polymorphism in the gene of the human phenol sulfotransferase SULT1A1 was studied as a model SNP. Biotinylated PCR products containing the SNP (The SNP summary web site: ) (mutant) and those containing no mutation (wild-type) were brought onto the chips coated with NeutrAvidin using non-contact spotting. This was followed by an analysis which was carried out in a flow-through biochip scanner while constantly rinsing with buffer. After removing the non-biotinylated strand a fluorescent probe was hybridised, which is complementary to the wild-type sequence. If this probe binds to a mutant sequence, then one single base is not fully matching. Thereby, the mismatched hybrid (mutant) is less stable than the full-matched hybrid (wild-type). The final step after hybridisation on the chip involves rinsing with a buffer to start dissociation of the fluorescent probe from the immobilised DNA strand. The online measurement of the fluorescence intensity by the biochip scanner provides the possibility to follow the kinetics of the hybridisation and dissociation processes. According to the different stability of the full-match and the mismatch, either visual discrimination or kinetic analysis is possible to distinguish SNP-containing sequence from the wild-type sequence.
Collapse
Affiliation(s)
- Susanne Schwonbeck
- Department of Molecular Bioanalytics and Bioelectronics, Fraunhofer Institute for Biomedical Engineering, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | | | | | | | | | | | | |
Collapse
|
133
|
Abstract
Sulfonation has a major function in modulating the biological activities of a wide number of endogenous and foreign chemicals, including: drugs, toxic chemicals, hormones, and neurotransmitters. The activation as well as inactivation of many xenobiotics and endogenous compounds occurs via sulfonation. The process is catalyzed by members of the cytosolic sulfotransferase (SULT) superfamily consisting of at least ten functional genes in humans. The reaction in intact cells may be reversed by arylsulafatase present in the endoplasmic reticulum. Under physiological conditions, sulfonation is regulated, in part, by the supply of the co-substrate/donor molecule 3'-phosphadensoine-5-phosphosulfate (PAPS), and transport mechanisms by which sulfonated conjugates enter and leave cells. Variation in the response of individuals to certain drugs and toxic chemicals may be related to genetic polymorphisms documented to occur in each of the above pathways. Sulfonation has a major function in regulating the endocrine status of an individual by modulating the receptor activity of estrogens and androgens, steroid biosynthesis, and the metabolism of catecholamines and iodothyronines Sulfonation is a key reaction in the body's defense against injurious chemicals and may have a major function during early development since SULTs are highly expressed in the human fetus. As with many Phase I and Phase II reactions, sulfonation may also serve as the terminal step in activating certain dietary and environmental agents to very reactive toxic intermediates implicated in carcinogenesis.
Collapse
Affiliation(s)
- Frederick C Kauffman
- Laboratory for Cellular and Biochemical Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA.
| |
Collapse
|
134
|
Phylogenomic approaches to common problems encountered in the analysis of low copy repeats: the sulfotransferase 1A gene family example. BMC Evol Biol 2005; 5:22. [PMID: 15752422 PMCID: PMC555591 DOI: 10.1186/1471-2148-5-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Accepted: 03/07/2005] [Indexed: 11/30/2022] Open
Abstract
Background Blocks of duplicated genomic DNA sequence longer than 1000 base pairs are known as low copy repeats (LCRs). Identified by their sequence similarity, LCRs are abundant in the human genome, and are interesting because they may represent recent adaptive events, or potential future adaptive opportunities within the human lineage. Sequence analysis tools are needed, however, to decide whether these interpretations are likely, whether a particular set of LCRs represents nearly neutral drift creating junk DNA, or whether the appearance of LCRs reflects assembly error. Here we investigate an LCR family containing the sulfotransferase (SULT) 1A genes involved in drug metabolism, cancer, hormone regulation, and neurotransmitter biology as a first step for defining the problems that those tools must manage. Results Sequence analysis here identified a fourth sulfotransferase gene, which may be transcriptionally active, located on human chromosome 16. Four regions of genomic sequence containing the four human SULT1A paralogs defined a new LCR family. The stem hominoid SULT1A progenitor locus was identified by comparative genomics involving complete human and rodent genomes, and a draft chimpanzee genome. SULT1A expansion in hominoid genomes was followed by positive selection acting on specific protein sites. This episode of adaptive evolution appears to be responsible for the dopamine sulfonation function of some SULT enzymes. Each of the conclusions that this bioinformatic analysis generated using data that has uncertain reliability (such as that from the chimpanzee genome sequencing project) has been confirmed experimentally or by a "finished" chromosome 16 assembly, both of which were published after the submission of this manuscript. Conclusion SULT1A genes expanded from one to four copies in hominoids during intra-chromosomal LCR duplications, including (apparently) one after the divergence of chimpanzees and humans. Thus, LCRs may provide a means for amplifying genes (and other genetic elements) that are adaptively useful. Being located on and among LCRs, however, could make the human SULT1A genes susceptible to further duplications or deletions resulting in 'genomic diseases' for some individuals. Pharmacogenomic studies of SULT1Asingle nucleotide polymorphisms, therefore, should also consider examining SULT1A copy number variability when searching for genotype-phenotype associations. The latest duplication is, however, only a substantiated hypothesis; an alternative explanation, disfavored by the majority of evidence, is that the duplication is an artifact of incorrect genome assembly.
Collapse
|
135
|
Wang L, Yee VC, Weinshilboum RM. Aggresome formation and pharmacogenetics: sulfotransferase 1A3 as a model system. Biochem Biophys Res Commun 2004; 325:426-33. [PMID: 15530410 DOI: 10.1016/j.bbrc.2004.10.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2004] [Indexed: 01/16/2023]
Abstract
A common cause for pharmacogenetic alteration in drug response is genetic variation in encoded amino acid sequence. We have used the catecholamine and drug-metabolizing enzyme sulfotransferase (SULT)1A3 to create an artificial model system to study mechanisms-especially possible aggresome formation-by which genetic alteration in amino acid sequence might influence function. Specifically, we created a double variant SULT1A3 allozyme that included the naturally occurring Asn234 polymorphism plus an additional Trp172Arg mutation. Analysis of the SULT1A3 X-ray crystal structure had indicated that the Trp172Arg mutation might destabilize the protein's structure. Expression of SULT1A3 Arg172,Asn234 in COS-1 cells resulted in undetectable enzyme activity and a virtual lack of enzyme protein. Rabbit reticulocyte lysate degradation studies showed that the double variant allozyme was degraded much more rapidly than was wild type SULT1A3 by a ubiquitin-proteasome-dependent process. In addition, after expression in COS-1 cells, the double variant allozyme localized to aggresomes, a process not previously described or studied in pharmacogenetics. Therefore, the alteration of only one or two amino acids can lead to decreased levels of protein as a result of both aggresome formation and accelerated degradation. The possible role of aggresome formation in pharmacogenetics should be evaluated in naturally occurring systems with inherited alteration in encoded amino acid sequence.
Collapse
Affiliation(s)
- Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Medical School-Mayo Clinic-Mayo Foundation, Rochester, MN 55905, USA
| | | | | |
Collapse
|
136
|
Abstract
Posttranslational modifications of proteins such as phosphorylation have been recognized as pivotal modulators of biological activity in healthy and diseased tissues. Sulfation is a key posttranslational modification the role of which in physiology and pathology is only now becoming appreciated. Whereas phosphorylation is central to intracellular signal transduction, sulfation modulates cell-cell and cell-matrix communication. Sulfation involves a class of enzymes known as sulfotransferases, which transfer sulfate from the ATP-like sulfate donor 3'phosphoadenosine-5'phosphosulate to glycoproteins, glycolipids or metabolites. This review focuses on Golgi-localized sulfotransferases, their molecular biology and biochemistry, and strategies towards discovery of sulfotransferase inhibitors that could have potential as therapeutics in inflammation, cancer and infectious diseases.
Collapse
Affiliation(s)
- Stefan Hemmerich
- Thios Pharmaceuticals, 5980 Horton Street #400, Emeryville, CA 94608, USA.
| | | | | |
Collapse
|
137
|
Hildebrandt MAT, Salavaggione OE, Martin YN, Flynn HC, Jalal S, Wieben ED, Weinshilboum RM. Human SULT1A3 pharmacogenetics: gene duplication and functional genomic studies. Biochem Biophys Res Commun 2004; 321:870-8. [PMID: 15358107 DOI: 10.1016/j.bbrc.2004.07.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Indexed: 10/26/2022]
Abstract
Sulfotransferase (SULT) 1A3 catalyzes the sulfate conjugation of catecholamines. Inheritance is an important factor responsible for individual variation in SULT1A3 activity, and gene resequencing studies have shown the presence of one functionally significant SULT1A3 nonsynonymous cSNP. However, following completion of the Human Genome Project, it appeared that SULT1A3 might be duplicated. We used specific PCR-based assays and fluorescence in situ hybridization to verify that 2 SULT1A3 genes-SULT1A3 and SULT1A4-were present on chromosome 16 in all human DNA samples studied. Furthermore, reanalysis of previous gene resequencing data confirmed the presence of the SULT1A3 SNPs identified previously, but also revealed 11 novel polymorphisms, including 3 nonsynonymous cSNPs. Functional genomic studies showed that two of those cSNPs, C302T, and C302A, resulted in decreased enzyme activity without striking changes in substrate kinetics but with parallel changes in levels of immunoreactive protein. In addition, RT-PCR revealed that both SULT1A3 and SULT1A4 can be transcriptionally active. The duplication of SULT1A3 will have to be taken into account in future efforts to understand individual variation in SULT1A3 activity or properties.
Collapse
Affiliation(s)
- Michelle A T Hildebrandt
- Department of Molecular Pharmacology, Mayo Clinic College of Medicine, Mayo Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
138
|
Wilson LA, Reyns GE, Darras VM, Coughtrie MWH. cDNA cloning, functional expression, and characterization of chicken sulfotransferases belonging to the SULT1B and SULT1C families. Arch Biochem Biophys 2004; 428:64-72. [PMID: 15234270 DOI: 10.1016/j.abb.2004.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Revised: 05/10/2004] [Indexed: 10/26/2022]
Abstract
A search of the chicken expressed sequence tag (EST) database identified 2 cDNA clones that appeared to represent members of the SULT1B and SULT1C enzyme families. These cDNAs were fully sequenced and found to contain full-length inserts. Phylogenetic analysis of the derived amino acid sequences clearly placed them as the first members of the chicken SULT1B and SULT1C families, respectively, to be identified, and we propose they be named SULT1B1 and SULT1C1. (CHICK)SULT1B1 shares approximately 60% amino acid sequence identity with mammalian SULT1B enzymes, whereas the closest neighbor to (CHICK)SULT1C1 was the ortholog (RAT)SULT1C1, with 68% identity. We cloned these cDNAs into the bacterial expression vectors from the pET series. Transformed Escherichia coli cells strongly expressed the recombinant proteins. Purification of the recombinant enzymes from E. coli was accomplished by a three-step procedure involving ammonium sulfate precipitation, anion exchange chromatography, and affinity chromatography. The purified enzymes displayed subunit molecular weights of approximately 35,000Da on SDS-PAGE, as predicted, and were both able to sulfate a wide range of compounds, including xenobiotics and endogenous substrates such as iodothyronines. Detailed kinetic analysis showed SULT1C1 was more prolific in that it was able to sulfate dopamine, tyramine, and apomorphine, which SULT1B1 was not. 2-Bromophenol was the best substrate for both enzymes. We also raised antibodies against these proteins, which were able to detect the SULTs by ELISA, and which were able to strongly inhibit the recombinant enzymes. This is the first detailed characterization of sulfotransferases from the chicken, and it demonstrates that the avian and mammalian SULT1 enzymes are closely related in both structure and function.
Collapse
Affiliation(s)
- Lesley A Wilson
- Division of Pathology and Neuroscience, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, Scotland, UK
| | | | | | | |
Collapse
|
139
|
Thomas NL, Coughtrie MWH. Sulfation of apomorphine by human sulfotransferases: evidence of a major role for the polymorphic phenol sulfotransferase, SULT1A1. Xenobiotica 2004; 33:1139-48. [PMID: 14660177 DOI: 10.1080/00498250310001609192] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
1. The relative roles of various members of the human sulfotransferase (SULT) enzyme family in the metabolism of apomorphine, a dopamine receptor antagonist used in the treatment of Parkinson's disease and, more recently, erectile dysfunction, were examined. In humans, sulfation is the major route of metabolism of this drug. 2. Using recombinant SULTs expressed in Escherichia coli, R(--)-apomorphine sulfation was studied using the universal barium precipitation assay in the presence of [35S] 3'-phosphoadenosine 5'-phosphosulfate and SULTs 1A1, 1A2, 1A3, 1B1, 1C2, 1E1 and 2A1. It was shown that SULTs 1A1, 1A2, 1A3 and 1E1 all sulfated apomorphine to varying extents. Low activity with SULT1B1 was only seen at the highest concentration (100 microM) and no activity with SULT1C2 or SULT2A1 was observed. 3. Kinetic analysis using purified recombinant SULTs showed that 1A1, 1A3 and 1E1 all had similar Vmax/Km values, although SULT1E1 had a slightly lower Km at around 1 microM compared with approximately 4 microM for the other SULTs. 4. By correlating apomorphine sulfation (at 10 microM) in a bank of 28 liver cytosols with SULT activity towards 10 microM 4-nitrophenol (SULT1A1) and 0.2 microM 17beta-oestradiol (SULT1E1), a strong correlation with SULT1A1 activity was clearly demonstrated, suggesting this enzyme was primarily responsible for hepatic apomorphine sulfation. 5. These findings were confirmed using immuno-inhibition experiments with antibodies against SULT1A and SULT1E1, which showed preferential inhibition of apomorphine sulfation in human liver cytosol by anti-SULT1A. 6. The results strongly implicate SULT1A1 as the major enzyme responsible for hepatic apomorphine metabolism. As SULT1A1 is subject to a common functional polymorphism, sulfation phenotype may be an important determinant of susceptibility to side-effects of apomorphine and/or efficacy of treatment.
Collapse
Affiliation(s)
- N L Thomas
- Department of Molecular and Cellular Pathology, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | | |
Collapse
|
140
|
Mougous JD, Petzold CJ, Senaratne RH, Lee DH, Akey DL, Lin FL, Munchel SE, Pratt MR, Riley LW, Leary JA, Berger JM, Bertozzi CR. Identification, function and structure of the mycobacterial sulfotransferase that initiates sulfolipid-1 biosynthesis. Nat Struct Mol Biol 2004; 11:721-9. [PMID: 15258569 DOI: 10.1038/nsmb802] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 05/19/2004] [Indexed: 11/08/2022]
Abstract
Sulfolipid-1 (SL-1) is an abundant sulfated glycolipid and potential virulence factor found in Mycobacterium tuberculosis. SL-1 consists of a trehalose-2-sulfate (T2S) disaccharide elaborated with four lipids. We identified and characterized a conserved mycobacterial sulfotransferase, Stf0, which generates the T2S moiety of SL-1. Biochemical studies demonstrated that the enzyme requires unmodified trehalose as substrate and is sensitive to small structural perturbations of the disaccharide. Disruption of stf0 in Mycobacterium smegmatis and M. tuberculosis resulted in the loss of T2S and SL-1 formation, respectively. The structure of Stf0 at a resolution of 2.6 A reveals the molecular basis of trehalose recognition and a unique dimer configuration that encloses the substrate into a bipartite active site. These data provide strong evidence that Stf0 carries out the first committed step in the biosynthesis of SL-1 and establish a system for probing the role of SL-1 in M. tuberculosis infection.
Collapse
Affiliation(s)
- Joseph D Mougous
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Sparks R, Ulrich CM, Bigler J, Tworoger SS, Yasui Y, Rajan KB, Porter P, Stanczyk FZ, Ballard-Barbash R, Yuan X, Lin MG, McVarish L, Aiello EJ, McTiernan A. UDP-glucuronosyltransferase and sulfotransferase polymorphisms, sex hormone concentrations, and tumor receptor status in breast cancer patients. Breast Cancer Res 2004; 6:R488-98. [PMID: 15318931 PMCID: PMC549165 DOI: 10.1186/bcr818] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Revised: 05/11/2004] [Accepted: 05/20/2004] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) enzymes are involved in removing sex hormones from circulation. Polymorphic variation in five UGT and SULT genes - UGT1A1 ((TA)6/(TA)7), UGT2B4 (Asp458Glu), UGT2B7 (His268Tyr), UGT2B15 (Asp85Tyr), and SULT1A1 (Arg213His)--may be associated with circulating sex hormone concentrations, or the risk of an estrogen receptor-negative (ER-) or progesterone receptor-negative (PR-) tumor. METHODS Logistic regression analysis was used to estimate the odds ratios of an ER- or PR- tumor associated with polymorphisms in the genes listed above for 163 breast cancer patients from a population-based cohort study of women in western Washington. Adjusted geometric mean estradiol, estrone, and testosterone concentrations were calculated within each UGT and SULT genotype for a subpopulation of postmenopausal breast cancer patients not on hormone therapy 2-3 years after diagnosis (n = 89). RESULTS The variant allele of UGT1A1 was associated with reduced risk of an ER- tumor (P for trend = 0.03), and variants of UGT2B15 and SULT1A1 were associated with non-statistically significant risk reductions. There was some indication that plasma estradiol and testosterone concentrations varied by UGT2B15 and SULT1A1 genotypes; women with the UGT2B15 Asp/Tyr and Tyr/Tyr genotypes had higher concentrations of estradiol than women with the Asp/Asp genotype (P = 0.004). Compared with women with the SULT1A1 Arg/Arg and Arg/His genotypes, women with the His/His genotype had elevated concentrations of testosterone (P = 0.003). CONCLUSIONS The risk of ER- breast cancer tumors may vary by UGT or SULT genotype. Further, plasma estradiol and testosterone concentrations in breast cancer patients may differ depending on some UGT and SULT genotypes.
Collapse
Affiliation(s)
- Rachel Sparks
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Cornelia M Ulrich
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jeannette Bigler
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Shelley S Tworoger
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Yutaka Yasui
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kumar B Rajan
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peggy Porter
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Rachel Ballard-Barbash
- Applied Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, USA
| | - Xiaopu Yuan
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ming Gang Lin
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lynda McVarish
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Erin J Aiello
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Group Health Cooperative, Center for Health Studies, Seattle, Washington, USA
| | - Anne McTiernan
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
142
|
Helander A, Beck O. Mass spectrometric identification of ethyl sulfate as an ethanol metabolite in humans. Clin Chem 2004; 50:936-7. [PMID: 15105353 DOI: 10.1373/clinchem.2004.031252] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Anders Helander
- Karolinska Institutet and University Hospital, Department of Clinical Neuroscience, Stockholm, Sweden.
| | | |
Collapse
|
143
|
Tabrett CA, Coughtrie MWH. Phenol sulfotransferase 1A1 activity in human liver: kinetic properties, interindividual variation and re-evaluation of the suitability of 4-nitrophenol as a probe substrate. Biochem Pharmacol 2004; 66:2089-97. [PMID: 14609733 DOI: 10.1016/s0006-2952(03)00582-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sulfation is an important metabolic pathway in humans for xenobiotics, hormones and neurotransmitters, and is catalysed by the cytosolic sulfotransferase (SULT) enzymes. Phenol SULTs, especially SULT1A1, are particularly important in xenobiotic and drug metabolism because of their broad substrate specificity and extensive tissue distribution. A common variant SULT1A1 allozyme (SULT1A1*2) exists in the population, and is less stable than the wild-type SULT1A1*1. 4-Nitrophenol is widely used as a substrate for quantifying SULT1A1 activity. However, our kinetic experiments suggest that 4-nitrophenol is not an ideal substrate when determining SULT1A1 activity in human liver. Assays with a bank of 68 human liver cytosols revealed three distinct kinetic profiles for 4-nitrophenol sulfation in the population: linear, biphasic and inhibition. Sulfation of 4-nitrophenol by purified, recombinant SULT1A1*1 and SULT1A1*2 shows marked substrate inhibition, with inhibition at 4-nitrophenol concentrations greater than 4 and 10 microM, respectively. Furthermore, sulfation of 4-nitrophenol by purified recombinant SULT1B1 was significant at concentrations of 4-nitrophenol less than 10 microM. Western blots showed that the SULT1A1 levels in liver are highly variable between liver samples and that no correlation was observed between SULT1A1 activity and protein level in liver cytosols. However, a correlation between SULT1A1 activity and protein level was observed in human placental cytosols, where SULT1B1 is not expressed. We believe that in human liver other SULT isoforms (particularly SULT1B1) contribute to the sulfation of 4-nitrophenol. Therefore, 4-nitrophenol is not an ideal substrate with which to quantitate SULT1A1 activity in human liver tissue.
Collapse
Affiliation(s)
- Catherine A Tabrett
- Department of Molecular & Cellular Pathology, University of Dundee, Ninewells Hospital & Medical School, Dundee DD1 9SY, Scotland, UK
| | | |
Collapse
|
144
|
Ansell PJ, Espinosa-Nicholas C, Curran EM, Judy BM, Philips BJ, Hannink M, Lubahn DB. In vitro and in vivo regulation of antioxidant response element-dependent gene expression by estrogens. Endocrinology 2004; 145:311-7. [PMID: 14551226 DOI: 10.1210/en.2003-0817] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Understanding estrogen's regulation of phase II detoxification enzymes is important in explaining how estrogen exposure increases the risk of developing certain cancers. Phase II enzymes such as glutathione-S-transferases (GST) and quinone reductase protect against developing chemically induced cancers by metabolizing reactive oxygen species. Phase II enzyme expression is regulated by a cis-acting DNA sequence, the antioxidant response element (ARE). It has previously been reported that several antiestrogens, but not 17beta-estradiol, could regulate ARE-mediated gene transcription. Our goal was to determine whether additional estrogenic compounds could regulate ARE-mediated gene expression both in vitro and in vivo. We discovered that physiological concentrations (10 nm) of 17beta-estradiol repressed GST Ya ARE-dependent gene expression in vitro. Treatment with other endogenous and anti-, xeno-, and phytoestrogens showed that estrogen receptor/ARE signaling is ligand, receptor subtype, and cell type specific. Additionally, GST and quinone reductase activities were significantly lowered in a dose-dependent manner after 17beta-estradiol exposure in the uteri of mice. In conclusion, we have shown that 17beta-estradiol, and other estrogens, down-regulate phase II enzyme activities. We propose estrogen-mediated repression of phase II enzyme activities may increase cellular oxidative DNA damage that ultimately can result in the formation of cancer in some estrogen-responsive tissues.
Collapse
Affiliation(s)
- P J Ansell
- Department of Biochemistry, MU Center for Phytonutrient and Phytochemical Studies, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | | | | | | | |
Collapse
|
145
|
Kester MHA, Kaptein E, Roest TJ, van Dijk CH, Tibboel D, Meinl W, Glatt H, Coughtrie MWH, Visser TJ. Characterization of rat iodothyronine sulfotransferases. Am J Physiol Endocrinol Metab 2003; 285:E592-8. [PMID: 12773305 DOI: 10.1152/ajpendo.00046.2003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sulfation appears to be an important pathway for the reversible inactivation of thyroid hormone during fetal development. The rat is an often used animal model to study the regulation of fetal thyroid hormone status. The present study was done to determine which sulfotransferases (SULTs) are important for iodothyronine sulfation in the rat, using radioactive T4, T3, rT3, and 3,3'-T2 as substrates, 3'-phosphoadenosine-5'-phosphosulfate (PAPS) as cofactor, and rat liver, kidney and brain cytosol, and recombinant rat SULT1A1, -1B1, -1C1, -1E1, -2A1, -2A2, and -2A3 as enzymes. Recombinant rat SULT1A1, -1E1, -2A1, -2A2, and -2A3 failed to catalyze iodothyronine sulfation. For all tissue SULTs and for rSULT1B1 and rSULT1C1, 3,3'-T2 was by far the preferred substrate. Apparent Km values for 3,3'-T2 amounted to 1.9 microM in male liver, 4.4 microM in female liver, 0.76 microM in male kidney, 0.23 microM in male brain, 7.7 microM for SULT1B1, and 0.62 microM for SULT1C1, whereas apparent Km values for PAPS showed less variation (2.0-6.9 microM). Sulfation of 3,3'-T2 was inhibited dose dependently by other iodothyronines, with similar structure-activity relationships for most enzymes except for the SULT activity in rat brain. The apparent Km values of 3,3'-T2 in liver cytosol were between those determined for SULT1B1 and -1C1, supporting the importance of these enzymes for the sulfation of iodothyronines in rat liver, with a greater contribution of SULT1C1 in male than in female rat liver. The results further suggest that rSULT1C1 also contributes to iodothyronine sulfation in rat kidney, whereas other, yet-unidentified forms appear more important for the sulfation of thyroid hormone in rat brain.
Collapse
Affiliation(s)
- Monique H A Kester
- Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|