1
|
Yetti H, Naito H, Yuan Y, Jia X, Hayashi Y, Tamada H, Kitamori K, Ikeda K, Yamori Y, Nakajima T. Bile acid detoxifying enzymes limit susceptibility to liver fibrosis in female SHRSP5/Dmcr rats fed with a high-fat-cholesterol diet. PLoS One 2018; 13:e0192863. [PMID: 29438418 PMCID: PMC5811017 DOI: 10.1371/journal.pone.0192863] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/31/2018] [Indexed: 02/07/2023] Open
Abstract
During middle age, women are less susceptible to nonalcoholic steatohepatitis (NASH) than men. Thus, we investigated the underlying molecular mechanisms behind these sexual differences using an established rat model of NASH. Mature female and male stroke-prone spontaneously hypertensive 5/Dmcr rats were fed control or high-fat-cholesterol (HFC) diets for 2, 8, and 14 weeks. Although HFC-induced hepatic fibrosis was markedly less severe in females than in males, only minor gender differences were observed in expression levels of cytochrome P450 enzymes (CYP)7A1, CYP8B1 CYP27A1, and CYP7B1, and multidrug resistance-associated protein 3, and bile salt export pump, which are involved in fibrosis-related bile acid (BA) kinetics. However, the BA detoxification-related enzymes UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) 2A1, and the nuclear receptors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), were strongly suppressed in HFC-fed males, and were only slightly changed in HFC-diet fed females. Expression levels of the farnesoid X receptor and its small heterodimer partner were similarly regulated in a gender-dependent fashion following HFC feeding. Hence, the pronounced female resistance to HFC-induced liver damage likely reflects sustained expression of the nuclear receptors CAR and PXR and the BA detoxification enzymes UGT and SULT.
Collapse
MESH Headings
- Animals
- Bile Acids and Salts/metabolism
- Cholesterol, Dietary/administration & dosage
- Cholesterol, Dietary/adverse effects
- Constitutive Androstane Receptor
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Disease Susceptibility
- Female
- Gene Expression
- Glucuronosyltransferase/metabolism
- Liver Cirrhosis/etiology
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Male
- Non-alcoholic Fatty Liver Disease/etiology
- Non-alcoholic Fatty Liver Disease/metabolism
- Non-alcoholic Fatty Liver Disease/pathology
- Pregnane X Receptor
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred SHR
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Sex Characteristics
- Sulfotransferases/metabolism
Collapse
Affiliation(s)
- Husna Yetti
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisao Naito
- Department of Public Health, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yuan Yuan
- College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Xiaofang Jia
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yumi Hayashi
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hazuki Tamada
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuya Kitamori
- College of Human Life and Environment, Kinjo Gakuin University, Nagoya, Japan
| | - Katsumi Ikeda
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya, Japan
| | - Yukio Yamori
- Institute for World Health Development, Mukogawa Women’s University, Nishinomiya, Japan
| | - Tamie Nakajima
- College of Life and Health Sciences, Chubu University, Kasugai, Japan
- * E-mail:
| |
Collapse
|
2
|
Lyu C, Zhang Y, Zhou W, Zhang S, Kou F, Wei H, Zhang N, Zuo Z. Gender-Dependent Pharmacokinetics of Veratramine in Rats: In Vivo and In Vitro Evidence. AAPS JOURNAL 2016; 18:432-44. [PMID: 26791530 DOI: 10.1208/s12248-016-9870-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 01/07/2016] [Indexed: 12/25/2022]
Abstract
Veratramine, a major alkaloid from Veratrum nigrum L., has distinct anti-tumor and anti-hypertension effects. Our previous study indicated that veratramine had severe toxicity toward male rats. In order to elucidate the underling mechanism, in vivo pharmacokinetic experiments and in vitro mechanistic studies have been conducted. Veratramine was administrated to male and female rats intravenously via the jugular vein at a dose of 50 μg/kg or orally via gavage at 20 mg/kg. As a result, significant pharmacokinetic differences were observed between male and female rats after oral administration with much lower concentrations of veratramine and 7-hydroxyl-veratramine and higher concentrations of veratramine-3-O-sulfate found in the plasma and urine of female rats. The absolute bioavailability of veratramine was 0.9% in female rats and 22.5% in male rats. Further experiments of veratramine on Caco-2 cell monolayer model and in vitro incubation with GI content or rat intestinal subcellular fractions demonstrated that its efficient passive diffusion mediated absorption with minimal intestinal metabolism, suggesting no gender-related difference during its absorption process. When veratramine was incubated with male or female rat liver microsomes/cytosols, significant male-predominant formation of 7-hydroxyl-veratramine and female-predominant formation of veratramine-3-O-sulfate were observed. In conclusion, the significant gender-dependent hepatic metabolism of veratramine could be the major contributor to its gender-dependent pharmacokinetics.
Collapse
Affiliation(s)
- Chunming Lyu
- Technology Laboratory Center, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.,School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, People's Republic of China
| | - Yufeng Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, People's Republic of China
| | - Wenbin Zhou
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shen Zhang
- Department of Rehabilitation, Changzheng Hospital Affiliated to Second Military Medicine University, Shanghai, 200003, People's Republic of China
| | - Fang Kou
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Hai Wei
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Ning Zhang
- Technology Laboratory Center, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
3
|
Wang Y, Ye F, Ke Q, Wu Q, Yang R, Bu H. Gender-dependent Histone Deacetylases Injury May Contribute to Differences in Liver Recovery Rates of Male and Female Mice. Transplant Proc 2013; 45:463-73. [DOI: 10.1016/j.transproceed.2012.06.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 06/08/2012] [Accepted: 06/26/2012] [Indexed: 02/07/2023]
|
4
|
Alnouti Y, Klaassen CD. Mechanisms of gender-specific regulation of mouse sulfotransferases (Sults). Xenobiotica 2010; 41:187-97. [PMID: 21091322 DOI: 10.3109/00498254.2010.535923] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. Marked gender differences in the expression of sulfotransferases (Sults) are known to exist in several species including rats, mice and hamsters. However, the mechanism for this gender difference is not known. Therefore, in the present study, it was determined whether sex and/or growth hormone (GH) are responsible for the gender difference in the expression of Sults using gonadectomized (GNX), hypophysectomized (HX) and GH-releasing hormone receptor-deficient little (lit/lit) mouse models. 2. Sult1a1 and Papss2 in liver and kidney, and Sult1d1 in liver are female-predominant in mice because of suppressive effects of both androgens and male-pattern GH secretion. Sult2a1/a2 is the most markedly female-predominant Sult in mouse liver due to suppressive effects of androgens and male-pattern GH secretion, as well as stimulatory effects by estrogens and female-pattern GH secretion. Sult3a1 is female-predominant in mouse liver due to suppressive effects of androgens as well as stimulatory effects of estrogens and female-pattern GH secretion. Sult1c1 expression is male-predominant in mouse liver and kidney because of stimulatory effects of androgens in males. Sult4a1 expression is female-predominant in mouse brain due to stimulatory effects of estrogens. 3. In conclusion, gender-divergent Sults are mostly female-predominant and Sult1c1 is the only male-dominant Sult. The gender differences in expression of various mouse Sults are influenced by various mechanisms involving sex and/or GHs.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
5
|
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
|
6
|
Panella-Riera N, Moe M, Grindflek E, Oliver M, Wood J, Doran O. Effect of sex steroids on expression of sulfotransferase 2B1 immunoreactive protein in primary cultured porcine hepatocytes. Livest Sci 2008. [DOI: 10.1016/j.livsci.2008.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
7
|
Alnouti Y, Klaassen CD. Regulation of sulfotransferase enzymes by prototypical microsomal enzyme inducers in mice. J Pharmacol Exp Ther 2008; 324:612-21. [PMID: 17993606 DOI: 10.1124/jpet.107.129650] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
In the present study, the regulation of the mRNA of 11 sulfotransferases (Sults) and two 3'-phosphoadenosine 5'-phosphosulfate synthase (PAPSs) isozymes by 15 microsomal enzyme inducers (MEI) in livers of male mice and five MEIs in livers of female mice was examined. These MEIs represent the transcriptionally mediated pathways: aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR), constitutive androstane receptor (CAR), peroxisomal proliferator-activated receptor alpha (PPARalpha), and NF-E2-related factor 2 (Nrf2). AhR ligands suppress the expression of Sults, especially the Sult1 isoenzymes in female mice. CAR activators up-regulate several Sults and PAPSs2 in female but not in male mice. PXR ligands cause marked induction of Sult1e1 in male, Sult2a1/2a2 in female, and PAPSs2 in both male and female mice. PPARalpha ligands do not have a marked effect on Sult expression in males, but they tend to suppress the expression of several Sult isoforms in female mice. Nrf2 activators appear to induce the mRNA expression of Sults in male and have mixed effects in female mice. In silico analysis indicated the presence of putative binding sites for all five transcription factors in the promoter region of many Sult and PAPSs isoforms. In conclusion, induction of Sults by typical MEIs is not as marked as the induction of P450 enzymes in mice. In addition to gender differences in basal expression of Sults, there is also a marked gender difference in the inducibility of various Sult isoenzymes in mice by MEIs.
Collapse
Affiliation(s)
- Yazen Alnouti
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160-7417, USA
| | | |
Collapse
|
8
|
Hodson MP, Dear GJ, Roberts AD, Haylock CL, Ball RJ, Plumb RS, Stumpf CL, Griffin JL, Haselden JN. A gender-specific discriminator in Sprague–Dawley rat urine: The deployment of a metabolic profiling strategy for biomarker discovery and identification. Anal Biochem 2007; 362:182-92. [PMID: 17266915 DOI: 10.1016/j.ab.2006.12.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 12/17/2006] [Accepted: 12/20/2006] [Indexed: 11/22/2022]
Abstract
The use of nuclear magnetic resonance (NMR) spectroscopy and liquid chromatography-mass spectrometry (LC-MS) as complementary analytical techniques for open metabolic profiling is illustrated in the context of defining urinary biochemical discriminators between male and female Sprague-Dawley rats. Subsequent to the discovery of a female-specific urinary discriminator by LC-MS, further LC, MS, and NMR methods have been applied in a coordinated effort to identify this urinary component. Thereafter, the biological relevance and context of the identified component, in this case a steroid metabolite, has been achieved. This approach will be deployed in future studies of disease, drug efficacy, and toxicity to discover and identify biologically relevant markers.
Collapse
Affiliation(s)
- Mark P Hodson
- Safety Assessment Division, GlaxoSmithKline, Ware, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
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
|
10
|
Duanmu Z, Weckle A, Koukouritaki SB, Hines RN, Falany JL, Falany CN, Kocarek TA, Runge-Morris M. Developmental expression of aryl, estrogen, and hydroxysteroid sulfotransferases in pre- and postnatal human liver. J Pharmacol Exp Ther 2006; 316:1310-7. [PMID: 16339912 DOI: 10.1124/jpet.105.093633] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aryl- (SULT1A1), estrogen- (SULT1E1), and hydroxysteroid- (SULT2A1) sulfotransferases (SULTs) are active determinants of xenobiotic detoxication and hormone metabolism in the adult human liver. To investigate the role of these conjugating enzymes in the developing human liver, the ontogeny of immunoreactive SULT1A1, SULT1E1, and SULT2A1 expression was characterized in a series of 235 pre- and postnatal human liver cytosols ranging in age from early gestation to a postnatal age of 18 years. Interindividual variability in expression levels was apparent for all three SULTs in pre- and postnatal liver samples. Expression of the three SULTs displayed distinctly different developmental profiles. Semiquantitative Western blot analyses indicated that SULT1A1 and SULT2A1 immunoreactive protein levels were readily detectable in the majority of developmental human liver cytosols throughout the prenatal period. Whereas SULT1A1 expression did not differ significantly among the various developmental stages, SULT2A1 expression increased during the third trimester of gestation and continued to increase during postnatal life. By contrast, SULT1E1, a cardinal estrogen-inactivating enzyme, achieved the highest levels of expression during the earliest periods of gestation in prenatal male livers, indicating a requisite role for estrogen inactivation in the developing male. The present analysis suggests that divergent regulatory mechanisms are responsible for the differential patterns of hepatic SULT1A1, SULT1E1, and SULT2A1 immunoreactive protein levels that occur during pre- and postnatal human development, and implicates a major role for sulfotransferase expression in the developing fetus.
Collapse
Affiliation(s)
- Zhengbo Duanmu
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Avenue, Room 4000, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Mattioli F, Garbero C, Gosmar M, Manfredi V, Carrozzino R, Martelli A, Brambilla G. DNA fragmentation, DNA repair and apoptosis induced in primary rat hepatocytes by dienogest, dydrogesterone and 1,4,6-androstatriene-17β-ol-3-one acetate. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2004; 564:21-9. [PMID: 15474407 DOI: 10.1016/j.mrgentox.2004.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2004] [Revised: 06/17/2004] [Accepted: 07/16/2004] [Indexed: 10/26/2022]
Abstract
Four steroids that share the 17-hydroxy-3-oxopregna-4,6-diene structure - cyproterone acetate, chlormadinone acetate, megestrol acetate, and potassium canrenoate - have been shown previously to behave with different potency as liver-specific genotoxic agents, the response being markedly higher in female than in male rats, but similar in humans of both genders. In this study, performed to better define the relationship between chemical structure and genotoxicity, dydrogesterone (DGT) with double bonds C4=C5 and C6=C7, dienogest (DNG) with double bonds C4=C5 and C9=C10, and 1,4,6-androstatriene-17beta-ol-3-one acetate (ADT) with double bonds C1=C2, C4=C5 and C6=C7, were compared with cyproterone acetate (CPA) for their ability to induce DNA fragmentation and DNA repair synthesis in primary cultures of hepatocytes from three rats of each sex. At subtoxic concentrations, ranging from 10 to 90 microM, all four steroids consistently induced a dose-dependent increase of DNA fragmentation, which in all cases was higher in females than in males; their DNA damaging potency decreased in the order CPA > DNG > ADT > DGT. Under the same experimental conditions, the responses provided by the DNA repair-synthesis assay were positive or inconclusive in hepatocytes from female rats and consistently negative in hepatocytes from male rats. In the induction of apoptotic cells, examined in primary hepatocytes from female rats, CPA was more active than ADT and DGT, and DNG was inactive. Considered as a whole these findings suggest that a liver-specific genotoxic effect more marked in female than in male rats might be a common property of steroids with two or three double bonds.
Collapse
Affiliation(s)
- Francesca Mattioli
- Department of Internal Medicine, Division of Clinical Pharmacology and Toxicology, University of Genoa, Viale Benedetto XV 2, I-16132 Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
12
|
You L. Steroid hormone biotransformation and xenobiotic induction of hepatic steroid metabolizing enzymes. Chem Biol Interact 2004; 147:233-46. [PMID: 15135080 DOI: 10.1016/j.cbi.2004.01.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2004] [Indexed: 11/30/2022]
Abstract
Normal reproductive development depends on the interplay of steroid hormones with their receptors at specific tissue sites. The concentrations of hormone ligands in the circulation and at target sites are maintained through coordinated regulation on steroid biosynthesis and degradation. Changed bioavailability of steroids, through alteration of steroidogenesis or biotransformation rates, leads to changes in endocrine function. Steroid hormones lose their receptor reactivity in most cases when they are bound to binding proteins, while metabolic conversion can result in either active or inactive metabolites. Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. The expression of these biotransformation enzymes can be induced by many xenobiotics. The barbiturate phenobarbital and the environmental toxicant 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) are among the well characterized inducers for the CYP 2B and 3A enzymes and selected conjugation enzymes. The induction of the steroid biotransformation enzymes is partly mediated through the activation of a group of nuclear receptors including the glucocorticoid receptor, the constitutive androstane receptor (CAR), the pregnane X receptor (PXR), and the peroxisome proliferator activated receptors (PPAR). Drug or chemical-induced increases in hepatic enzyme activities are often a basis for drug-drug interactions that lead to enhanced elimination and reduced therapeutic efficacy of steroidal drugs. The effects of enzyme induction on endogenous steroid clearance, along with its possible consequence, are less well understood. While enzyme induction by xenobiotics may increase clearance of the endogenous steroid, regulatory mechanisms for steroid homeostasis may adapt and compensate for altered clearance.
Collapse
Affiliation(s)
- Li You
- CIIT Centers for Health Research, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709-2137, USA.
| |
Collapse
|
13
|
Sugahara T, Yang YS, Liu CC, Pai TG, Liu MC. Sulphonation of dehydroepiandrosterone and neurosteroids: molecular cloning, expression, and functional characterization of a novel zebrafish SULT2 cytosolic sulphotransferase. Biochem J 2003; 375:785-91. [PMID: 12885295 PMCID: PMC1223711 DOI: 10.1042/bj20031050] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2003] [Accepted: 07/29/2003] [Indexed: 11/17/2022]
Abstract
By searching the zebrafish EST (expressed-sequence tag) database, we have identified two partial cDNA clones encoding the 5' and 3' regions of a putative zebrafish sulphotransferase (ST). Using the reverse transcription-PCR technique, a full-length cDNA encoding this zebrafish ST was successfully cloned. Sequence analysis revealed that this novel zebrafish ST displays 44%, 43% and 40% amino acid identity with mouse SULT2B1, human SULT2B1b and human SULT2A1 ST respectively. This zebrafish ST therefore appears to belong to the SULT2 cytosolic ST gene family. Recombinant zebrafish ST, expressed using the pGEX-2TK prokaryotic expression system and purified from transformed Escherichia coli cells, migrated as a 34 kDa protein upon SDS/PAGE. Purified zebrafish ST displayed a strong sulphonating activity toward DHEA (dehydroepiandrosterone), with a optimum pH of 9.5. The enzyme also exhibited activities toward several neurosteroids with differential K(m) and V(max) values. A thermostability experiment revealed the enzyme to be relatively stable over a temperature range between 20 degrees C and 43 degrees C. Among ten different divalent metal cations tested, Fe2+ and Cd(2+ exhibited small, but significant, stimulatory effects, whereas Hg2+ and Cu2+ displayed considerably stronger inhibitory effects on the DHEA-sulphonating activity of the enzyme. These results constitute the first study on the molecular cloning, expression, and characterization of a zebrafish cytosolic SULT2 ST.
Collapse
Affiliation(s)
- Takuya Sugahara
- Biomedical Research Center, The University of Texas Health Center, Tyler, TX 75708, USA
| | | | | | | | | |
Collapse
|
14
|
Gardmo C, Swerdlow H, Mode A. Growth hormone regulation of rat liver gene expression assessed by SSH and microarray. Mol Cell Endocrinol 2002; 190:125-33. [PMID: 11997186 DOI: 10.1016/s0303-7207(02)00004-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The sexually dimorphic secretion of growth hormone (GH) that prevails in the rat leads to a sex-differentiated expression of GH target genes, particularly in the liver. We have used subtractive suppressive hybridization (SSH) to search for new target genes induced by the female-characteristic, near continuous, pattern of GH secretion. Microarrays and dot-blot hybridizations were used in an attempt to confirm differential ratios of expression of obtained SSH clones. Out of 173 unique SSH clones, 41 could be verified as differentially expressed. Among these, we identified 17 known genes not previously recognized as differentially regulated by the sex-specific GH pattern. Additional SSH clones may also represent genes subjected to sex-specific GH regulation since only transcripts abundantly expressed could be verified. Optimized analyses, specific for each gene, are required to fully characterize the degree of differential expression.
Collapse
Affiliation(s)
- Cissi Gardmo
- Department of Medical Nutrition, Karolinska Institutet, Novum, S-141 86 Huddinge, Sweden.
| | | | | |
Collapse
|
15
|
Takehara K, Kubushiro K, Iwamori Y, Tsukazaki K, Nozawa S, Iwamori M. Expression of an isoform of the testis-specific estrogen sulfotransferase in the murine placenta during the late gestational period. Arch Biochem Biophys 2001; 394:201-8. [PMID: 11594734 DOI: 10.1006/abbi.2001.2545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytosolic sulfotransferases play essential roles in regulating the activities and transfer of steroids. To evaluate their biological significance in the murine uterus and placenta during the course of gestation, we determined their activities with several steroids as substrates. Activated estrogen sulfotransferase (EST) was found in the placenta and uterus during the late gestational period. Reverse-transcribed cDNA of murine placental EST (mpEST) was isolated from mouse placenta at 18 days of gestation and its expression in the tissue coincided with a change in its enzyme activity. The open-reading frame of mpEST encodes a protein composed of 296 amino acids with a predicted molecular mass of 35.5 kDa and was revealed to be an isoform of the murine testis-specific EST gene (99.7%). Also, the amino acid sequence of mpEST showed 49.6 and 77.9% homology with human placental and endometrial EST, respectively, showing that it corresponds to human endometrial EST. COS-7 cells transfected with mpEST exhibited sulfotransferase activity with the phenolic hydroxy groups of steroids and artificial substrates. The best acceptor substrate was estrogen.
Collapse
Affiliation(s)
- K Takehara
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Wu W, Kocarek TA, Runge-Morris M. Sex-dependent regulation by dexamethasone of murine hydroxysteroid sulfotransferase gene expression. Toxicol Lett 2001; 119:235-46. [PMID: 11246177 DOI: 10.1016/s0378-4274(01)00263-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To determine whether glucocorticoid-inducible expression of hepatic hydroxysteroid sulfotransferase is conserved in mouse, the effects of dexamethasone (DEX) on hydroxysteroid sulfotransferase (mSULT2A) gene expression were investigated in primary cultured hepatocytes prepared from C57BL/6J mice. In female mouse hepatocytes, DEX (10(-7) and 10(-5) M, respectively) produced 8.2- and 17.8-fold increases, respectively, in the amounts of mSULT2A mRNA relative to control. By contrast, mSULT2A mRNA levels were undetectable in male mouse hepatocytes. Female-predominant mSULT2A mRNA expression was also observed in liver samples from C57BL/6J and three other mouse strains. Treatment of primary cultured female mouse hepatocytes with dihydrotestosterone in the presence of DEX suppressed mSULT2 expression. Transfection of primary cultured male or female mouse hepatocytes with a rat SULT2-40/41 reporter construct revealed that hepatocytes of both sexes have sufficient machinery to achieve DEX-inducible SULT2 transcription. However, treatment with the potent histone deacetylase inhibitor trichostatin A failed to elicit mSULT2A expression in male mouse hepatocytes.
Collapse
Affiliation(s)
- W Wu
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Avenue, Detroit, MI 48201, USA
| | | | | |
Collapse
|
17
|
Abstract
Sex-dependent differences in xenobiotic metabolism have been most extensively studied in the rat. Because sex-dependent differences are most pronounced in rats, this species quickly became the most popular animal model to study sexual dimorphisms in xenobiotic metabolism. Exaggerated sex-dependent variations in metabolism by rats may be the result of extensive inbreeding and/or differential evolution of isoforms of cytochromes P450 in mammals. For example, species-specific gene duplications and gene conversion events in the CYP2 and CYP3 families have produced different isoforms in rats and humans since the species division over 80 million years ago. This observation can help to explain the fact that CYP2C is not found in humans but is a major subfamily in rats (Table 11). Animal studies are used to help determine the metabolism and toxicity of many chemical agents in an attempt to extrapolate the risk of human exposure to these agents. One of the most important concepts in attempting to use rodent studies to identify sensitive individuals in the human population is that human cytochromes P450 differ from rodent cytochromes P450 in both isoform composition and catalytic activities. Xenobiotic metabolism by male rats can reflect human metabolism when the compound of interest is metabolized by CYP1A or CYP2E because there is strong regulatory conservation of these isoforms between rodents and humans. However, problems can arise when rats are used as animal models to predict the potential for sex-dependent differences in xenobiotic handling in humans. Information from countless studies has shown that the identification of sex-dependent differences in metabolism by rats does not translate across other animal species or humans. The major factor contributing to this observation is that CYP2C, a major subfamily in rats, which is expressed in a sex-specific manner, is not found in humans. To date, sex-specific isoforms of cytochromes P450 have not been identified in humans. The lack of expression of sex-dependent isoforms in humans indicates that the male rat is not an accurate model for the prediction of sex-dependent differences in humans. Differences in xenobiotic metabolism among humans are more likely the consequence of intraindividual variations as a result of genetics or environmental exposures rather than from sex-dependent differences in enzyme composition. A major component of the drug discovery and development process is to identify, at as early a stage as possible, the potential for toxicity in humans. Earlier identification of individual differences in xenobiotic metabolism and the potential for toxicity will be facilitated by improving techniques to make better use of human tissue to prepare accurate in vitro systems such as isolated hepatocytes and liver slices to study xenobiotic metabolism and drug-induced toxicities. Accurate systems should possess an array of bioactivation enzymes similar to the in vivo expression of human liver. In addition, the compound concentrations and exposure times used in these in vitro test systems should mimic those achieved in the target tissues of humans. Consideration of such factors will allow the development of compounds with improved efficacy and low toxicity at a more efficient rate. The development of accurate in vitro systems utilizing human tissue will also aid in the investigation of the molecular mechanisms by which the CYP genes are regulated in humans. Such studies will facilitate the study of the basis for differences in expression of isoforms of CYP450 in humans.
Collapse
Affiliation(s)
- C A Mugford
- Chemical Industry Institute of Toxicology, Research Triangle Park, North Carolina 27709-2137, USA
| | | |
Collapse
|
18
|
Runge-Morris M. Regulation of sulfotransferase gene expression by glucocorticoid hormones and xenobiotics in primary rat hepatocyte culture. Chem Biol Interact 1998; 109:315-27. [PMID: 9566755 DOI: 10.1016/s0009-2797(97)00142-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the rat liver, hydroxysteroid sulfotransferase-a (HST-a) and aryl sulfotransferase IV (ASTIV) represent two major rat hepatic sulfotransferases that are important to xenobiotic metabolism. Prototypic CYP1A1 and CYP2B/3A inducers regulate rat hepatic sulfotransferase gene expression although not necessarily in a coordinate direction. It has been previously reported that in vivo treatment with CYP1A1 inducer 3-methylcholanthrene (3-MC) suppresses rat hepatic HST-a mRNA expression in a dose-dependent manner. Similarly, HST-a and ASTIV mRNA levels become suppressed or induced, respectively, following in vivo treatment with phenobarbital (PB)-like CYP2B/3A inducers or prototypic CYP3A inducers such as glucocorticoid hormones. In the whole animal, sulfotransferase gene expression is modulated by members of the hypothalamic/pituitary-adrenal gonadal hormone axis. However, studies in primary rat hepatocyte culture suggest that prototypic P450 inducers regulate HST-a and ASTIV gene expression directly at the level of the hepatocyte. Glucocorticoid-mediated sulfotransferase expression was compared with the regulation of tyrosine amino transferase (TAT), a gene that is transcriptionally regulated by ligand bound glucocorticoid receptor. It was found that lower doses of dexamethasone (DEX, 10(-7) M) produced concomitant increases in ASTIV and TAT mRNA expression, whereas HST-a mRNA expression continued to rise as the DEX dose was increased through 10(-5) M. When hepatocytes were co-incubated with DEX and antiglucocorticoid/antiprogestin RU-486, DEX-stimulated HST-a mRNA expression was not significantly inhibited by RU-486, but ASTIV and TAT mRNA expression were inhibited to a similar extent. The results suggested that ASTIV, like TAT, is likely regulated by a classical glucocorticoid receptor mediated mechanism, whereas HST-a is probably regulated by glucocorticoids via an alternative mechanism. In contrast to the positive effects of glucocorticoid hormones, HST-a and ASTIV mRNA expression was negatively regulated by xenobiotics such as PB-like CYP2B/3A inducers or aryl hydrocarbon receptor (AhR) agonist CYP1A1 inducers. Incubation of primary cultured rat hepatocytes with PB or structurally dissimilar PB-like inducers clotrimazole, diphenylhydantoin, heptachlor, gamma-hexachlorocyclohexane or 2,2',4,4',5,5'-hexachlorobiphenyl suppressed HST-a and ASTIV mRNA levels. Also, incubation of primary cultured rat hepatocytes with CYP1A1 inducer beta-naphthoflavone or with archetypic AhR agonist, 2,3,7,8-tetrachloro-p-dioxin (TCDD) markedly suppressed HST-a and ASTIV mRNA expression. These data suggest that the rat HST-a and ASTIV genes are positively regulated by glucocorticoid hormones and negatively regulated by xenobiotics as a result of molecular and cellular mechanisms that act directly on the hepatocyte.
Collapse
Affiliation(s)
- M Runge-Morris
- Institute of Chemical Toxicology, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
19
|
Lin B, Kubushiro K, Akiba Y, Cui Y, Tsukazaki K, Nozawa S, Iwamori M. Alteration of acidic lipids in human sera during the course of pregnancy: characteristic increase in the concentration of cholesterol sulfate. JOURNAL OF CHROMATOGRAPHY. B, BIOMEDICAL SCIENCES AND APPLICATIONS 1997; 704:99-104. [PMID: 9518183 DOI: 10.1016/s0378-4347(97)00450-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this study, we determined the concentrations of acidic lipids, including cholesterol sulfate (CS), sulfatide and GM3 ganglioside, in human sera of non-pregnant state and during the course of pregnancy. In human sera of non-pregnant women, GM3 was present at a concentration of 8 nmol/ml and the concentrations of CS and sulfatides were less than 20% of that of GM3. The concentration of sulfatides in sera at the second trimester of gestation was decreased, but CS gradually increased from the first to the third trimester of gestation with a correlation coefficient of 0.66, and a correlation between the concentration of CS and weeks of gestation (p<0.01). CS was also contained in the placental villi, and its concentration increased from the first to the third trimester of gestation, suggesting that placental CS is one of the source of CS in the blood by shedding.
Collapse
Affiliation(s)
- B Lin
- Department of Biochemistry, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Falany CN, Comer KA, Dooley TP, Glatt H. Human dehydroepiandrosterone sulfotransferase. Purification, molecular cloning, and characterization. Ann N Y Acad Sci 1995; 774:59-72. [PMID: 8597484 DOI: 10.1111/j.1749-6632.1995.tb17372.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human tissues possess at least four distinct forms of cytosolic ST, three of which are involved in the sulfation of steroids. DHEA-ST is responsible for the majority of hydroxysteroid and bile acid sulfation in human tissues and abundant levels of the enzyme are present in human liver and adrenal tissues. In the adult human adrenal, DHEA-ST has been localized immunologically to the zona reticularis of the adrenal cortex. No age- or gender-related differences in the expression of DHEA-ST activity in adult human liver cytosols have been reported. The cDNA encoding DHEA-ST has been isolated from a human liver cDNA library and expressed in both mammalian COS cells and E. coli. Purification and molecular characterization studies suggest a single form of DHEA-ST in human tissues. The properties of DHEA-ST expressed in either mammalian or bacterial cells are very similar to those of the native enzyme. DHEA-ST can also bioactivate a number of procarcinogens to reactive electrophilic forms. Hydroxymethyl PAHs are sulfated and bioactivated at a relatively rapid rate by DHEA-ST, whereas 1'-hydroxysafrole and N-hydroxy-2-acetylaminofluorene are bioactivated to a lesser extent.
Collapse
Affiliation(s)
- C N Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham 35294, USA
| | | | | | | |
Collapse
|
21
|
Schauss SJ, Henry T, Palmatier R, Halvorson L, Dannenbring R, Beckmann JD. Characterization of bovine tracheobronchial phenol sulphotransferase cDNA and detection of mRNA regulation by cortisol. Biochem J 1995; 311 ( Pt 1):209-17. [PMID: 7575456 PMCID: PMC1136140 DOI: 10.1042/bj3110209] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Phenol sulphotransferases esterify both endogenous and foreign hydroxylated aromatic compounds with sulphate. Since these enzymes participate in both hormone and drug metabolism, elucidating their regulation at both the enzymic and molecular levels may provide new understanding in several metabolic pathways. The primary structure of a bovine phenol sulphotransferase has been determined by isolation of the corresponding cDNA. Two partial bovine cDNAs were first isolated by probing a tracheal epithelial cell lambda gt11 cDNA library with a rat phenol sulphotransferase cDNA. These clones provided the sequences of the 5' and 3' ends of the predicted coding region. A contiguous cDNA was subsequently isolated by PCR using 5' and 3' oligonucleotide primers and the cDNA library as the template. The sequence of the resulting approx. 1 kbp cDNA predicted an amino acid sequence that included sequences determined for several tryptic peptides of the purified protein. Antiserum directed to a synthetic N-terminal peptide predicted by the cDNA sequence showed reactivity with the purified enzyme. High-level Trc-promoter-driven expression of the recombinant bovine enzyme was achieved in Escherichia coli. The bovine cDNA was used to determine relative steady-state levels of phenol sulphotransferase transcripts in bovine lung tissues; distal lung parenchymal RNA levels were 6-10-fold greater than those in tracheobronchial epithelium. Using a bronchial epithelial cell culture model, however, cortisol was observed to increase mRNA levels by 5-fold in both a dose- and time-dependent manner; this corresponds to previously reported glucocorticoid stimulation of phenol sulphotransferase activity in this system [Beckmann, Illig and Bartzatt (1994) J. Cell Physiol. 160, 603-610].
Collapse
Affiliation(s)
- S J Schauss
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha 68193-5300, USA
| | | | | | | | | | | |
Collapse
|
22
|
Matsui M, Homma H. Biochemistry and molecular biology of drug-metabolizing sulfotransferase. THE INTERNATIONAL JOURNAL OF BIOCHEMISTRY 1994; 26:1237-47. [PMID: 7851628 DOI: 10.1016/0020-711x(94)90093-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Sulfation is an important conjugation reaction in the metabolism of various xenobiotics and endogenous compounds and is catalyzed by sulfotransferase (ST) present in cytosols. The cloning studies on STs have provided the basis for the understanding of the ST multigene family. STs are classified into hydroxysteroid (or alcohol), aryl (or phenol), estrogen, flavonol and polysaccharide STs and recent developments in the molecular characterization of these isoforms are reviewed. Regulation and localization of ST isoforms in various tissues are characterized at the molecular level by virtue of the specific antibodies and the corresponding cDNA probes. The recent developments are summarized. ST inhibitors are potent tools for the study on ST multiplicity and for the characterization of the enzyme structure. It also appears to be important to understand exogenous and endogenous ST inhibitors in clinical environment. The recent developments are reviewed.
Collapse
Affiliation(s)
- M Matsui
- Kyoritsu College of Pharmacy, Tokyo, Japan
| | | |
Collapse
|
23
|
Abstract
Enzymatic sulfation has been implicated to play a key role in a number of essential biological pathways including xenobiotic detoxication, carcinogen activation, and the regulation of intra-tissue hormone activity. In order to increase our understanding of the critical determinants governing the regulation of sulfotransferase gene expression, we investigated age-, gender-, and xenobiotic-related alterations in hydroxysteroid sulfotransferase-a or aryl sulfotransferase-IV gene expression. Northern blot and slot blot analyses showed that rat hepatic hydroxysteroid sulfotransferase-a mRNA expression was responsive to age- and gender-related signals. The results also suggested that the rat hepatic aryl sulfotransferase-IV and hydroxysteroid sulfotransferase-a genes are differentially regulated. Northern blot and reverse transcriptase polymerase chain reaction analyses demonstrated that hydroxysteroid sulfotransferase-a mRNA was expressed to a greater extent in female rat liver than in lung or kidney tissue. In addition, rat hepatic hydroxysteroid sulfotransferase-a gene expression in mature female rats, although not substantially altered in response to short-term fasting or high-dose dexamethasone treatment, was suppressed after treatment with the polycyclic aromatic hydrocarbon, 3-methylcholanthrene.
Collapse
Affiliation(s)
- M A Runge-Morris
- Institute of Chemical Toxicology, Wayne State University, Detroit, MI 48201
| |
Collapse
|
24
|
Surh YJ, Miller JA. Roles of electrophilic sulfuric acid ester metabolites in mutagenesis and carcinogenesis by some polynuclear aromatic hydrocarbons. Chem Biol Interact 1994; 92:351-62. [PMID: 8033269 DOI: 10.1016/0009-2797(94)90076-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hydroxylation of meso-methyl groups with subsequent formation of reactive benzylic esters bearing a good leaving group (e.g. sulfate) was proposed as a possible biochemical mechanism of activation and tumorigenicity of methyl-substituted polycyclic aromatic hydrocarbons (PAHs). In support of this postulation, recent studies have demonstrated the formation by rodent hepatic sulfotransferase activity of electrophilic, mutagenic, and carcinogenic sulfuric acid esters of several hydroxymethyl aromatic hydrocarbons including hydroxymethyl derivatives of benz[a]anthracene, 6-hydroxymethylbenzo[a]pyrene, 5-hydroxymethylchrysene, 9-hydroxymethyl-10-methylanthracene, and 1-hydroxymethylpyrene. Besides these hydroxymethyl PAHs containing a primary benzylic alcoholic group, some aromatic hydrocarbons with secondary benzylic hydroxyl functional group(s) are also metabolically activated through sulfuric acid esterification.
Collapse
Affiliation(s)
- Y J Surh
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06510
| | | |
Collapse
|
25
|
Otterness DM, Weinshilboum R. Human dehydroepiandrosterone sulfotransferase: molecular cloning of cDNA and genomic DNA. Chem Biol Interact 1994; 92:145-59. [PMID: 8033249 DOI: 10.1016/0009-2797(94)90060-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human tissues contain at least three well-characterized cytoplasmic sulfotransferase (ST) enzymes, dehydroepiandrosterone (DHEA) ST and two of phenol ST (PST). DHEA ST catalyzes the sulfation of DHEA and other steroids. We cloned and expressed two cDNAs for human liver DHEA ST. The cloning strategy involved the design of PCR primers directed against two conserved domains in ST proteins. These primers were used to generate a specific PCR product that was then used successfully to clone cDNAs for DHEA ST from a human liver cDNA library. Two cDNAs were isolated that were approximately 1.1 and 1.8 kb in length. These two clones had identical open reading frames. Both cDNAs produced enzymatically active DHEA ST protein in a mammalian expression system. Northern blot analysis confirmed the presence of 1.1 and 1.8 kb transcripts in human liver. cDNAs for a number of eukaryotic enzymes have now been cloned, and they share significant sequence homology. These ST cDNAs appear to fall into distinct groups on the basis of amino acid sequences of the proteins that they encode, thus demonstrating that the enzymes comprise a gene superfamily. We have also isolated, a genomic clone for human DHEA ST that contains approximately 3 kb of 5'-flanking sequence, exon 1 and 1.7 kb of intron 1. Characterization of the structure and regulatory elements of this gene should help to elucidate mechanisms involved in the regulation of DHEA ST in humans.
Collapse
Affiliation(s)
- D M Otterness
- Department of Pharmacology, Mayo Medical School, Rochester, MN 55905
| | | |
Collapse
|
26
|
Homma H, Nakagome I, Kamakura M, Hirota M, Takahashi M, Matsui M. Studies on rat hepatic hydroxysteroid sulfotransferase--immunochemistry, development and pI variants. Chem Biol Interact 1994; 92:15-24. [PMID: 8033250 DOI: 10.1016/0009-2797(94)90049-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Rat hepatic hydroxysteroid sulfotransferase with sulfoconjugates androsterone (androsterone-sulfating sulfotransferase) is an oligomer consisting of several subunits with distinct pI values but with the same molecular mass (pI variants). N-terminal amino acid sequences of the pI variants are all identical. The enzyme is exclusively present in the liver, in which its lobular localization is sex-dependent. The localization of the enzyme is markedly different from that of an isoenzyme of phenol sulfotransferase. In weanling and adult female rats, the relative abundance of the pI variants is different. During development from weanling stage to adulthood, the amounts of acidic variants increase, whereas the relative levels of alkaline variants remain constant.
Collapse
Affiliation(s)
- H Homma
- Kyoritsu College of Pharmacy, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Bernier F, Lopez Solache I, Labrie F, Luu-The V. Cloning and expression of cDNA encoding human placental estrogen sulfotransferase. Mol Cell Endocrinol 1994; 99:R11-5. [PMID: 8187949 DOI: 10.1016/0303-7207(94)90159-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Using two oligoprimers derived from the bovine placental estrogen sulfotransferase sequence, we amplified a probe for human placental estrogen sulfotransferase. Using this probe to screen a human placental cDNA library constructed in lambda gt11, we isolated a cDNA clone of 1.3 kb encoding human estrogen sulfotransferase. DNA analysis predicts a protein of 295 amino acids with a calculated molecular weight of 34,199. Alignment of the amino acid sequence with other sulfotransferases indicates that human placental estrogen sulfotransferase shares 68.6, 68.2 and 65.9% similarity with bovine placental, guinea pig adrenocortical, and rat liver estrogen sulfotransferase, respectively. It shows also 95.6, 57.6, 85.3, and 54.2% similarity to human phenol, human DHEA, rat phenol, and rat hydroxysteroid sulfotransferase, respectively. Transfection of expression vectors encoding human estrogen sulfotransferase and dehydroepiandrosterone (DHEA) sulfotransferase in human adrenal adenocarcinoma SW-13 cells indicates that estrogen sulfotransferase transforms estrone more specifically, whereas DHEA sulfotransferase is more specific for DHEA and pregnenolone.
Collapse
Affiliation(s)
- F Bernier
- Medical Research Council Group in Molecular Endocrinology, CHUL Research Center, Quebec, Canada
| | | | | | | |
Collapse
|
28
|
Borthwick EB, Burchell A, Coughtrie MW. Purification and immunochemical characterization of a male-specific rat liver oestrogen sulphotransferase. Biochem J 1993; 289 ( Pt 3):719-25. [PMID: 8435070 PMCID: PMC1132234 DOI: 10.1042/bj2890719] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Sulphation of oestrogens represents an important regulatory mechanism for these biologically active compounds. We have characterized and purified a form of rat liver sulphotransferase (ST), existing as a 32,500 Da monomer, which sulphates oestrogens, and have used this preparation to produce antibodies against oestrogen ST. The enzyme was active against oestrone, oestriol and beta-oestradiol, but not towards androgens. Using the antibody as a probe for immunoblotting, it was determined that the enzyme is expressed solely in male rats, and predominantly in the liver. Of the tissues examined, the only major extrahepatic tissue found to have any oestrogen ST was the brain (although the levels were very low), indicating that there might be a role for the sulphation of oestrogens in the brain. Examination of human liver and platelet cytosols by immunoblotting showed that the antibody recognized two major proteins of 32 and 34 kDa, which were presumed to correspond to the two principal phenol ST isoenzymes present in man.
Collapse
Affiliation(s)
- E B Borthwick
- Department of Biochemical Medicine, University of Dundee, Ninewells Hospital and Medical School, Scotland, U.K
| | | | | |
Collapse
|
29
|
Homma H, Nakagome I, Kamakura M, Matsui M. Immunochemical characterization of developmental changes in rat hepatic hydroxysteroid sulfotransferase. BIOCHIMICA ET BIOPHYSICA ACTA 1992; 1121:69-74. [PMID: 1599953 DOI: 10.1016/0167-4838(92)90338-e] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A major isoenzyme of hepatic androsterone-sulfating sulfotransferase (AD-ST) was purified from adult female rats. The activity was purified 122-fold over that found in the cytosol and showed a single protein band with a subunit molecular mass of 30 kDa after sodium dodecyl sulfate polyacrylamide gel electrophoresis. The purified enzyme exhibited four isoelectric variants of subunits on denaturing isoelectrofocusing gels (pI = 5.8, 6.1, 6.7 and 7.2). Rabbit antiserum raised against the enzyme specifically detected AD-ST polypeptide in rat liver cytosol. Immunoblot analysis of liver cytosol from female and male rats at various ages showed good correlation between the levels of AD-ST activity and AD-ST polypeptide. Significant levels of AD-ST activity and polypeptide were detected in senescent male rats, though normal adult male rats have very low levels of AD-ST activity and protein. The relative content of the isoelectric variants of AD-ST were different in liver cytosol of weanling and adult females, indicating that age- and gender-related alterations of hepatic AD-ST activity are primarily determined by the levels of AD-ST polypeptide and the relative amounts of the four isoelectric variants of the enzyme.
Collapse
Affiliation(s)
- H Homma
- Kyoritsu College of Pharmacy, Tokyo, Japan
| | | | | | | |
Collapse
|