Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Steroid Biochem Mol Biol ; 102(1-5): 214-21, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17055258

ABSTRACT

Sulfation is an important Phase II conjugation reaction involved in the synthesis and metabolism of steroids in humans. Two different isoforms (2B1a and 2B1b) are encoded by the sulfotransferase (SULT) 2B1 gene utilizing different start sites of transcription resulting in the incorporation of different first exons. SULT2B1a and SULT2B1b are 350 and 365 amino acids in length, respectively, and the last 342 aa are identical. Message for both SULT2B1 isoforms is present in human tissues although SULT2B1b message is generally more abundant. However, to date only SULT2B1b protein has been detected in human tissues or cell lines. SULT2B1b is localized in the cytosol and/or nuclei of human cells. A unique 3'-extension of SULT2B1b is required for nuclear localization in human BeWo placental choriocarcinoma cells. Nuclear localization is stimulated by forskolin treatment in BeWo cells and serine phosphorylation has been identified in the 3'-extension. SULT2B1b is selective for the sulfation of 3beta-hydroxysteroids such as dehydroepiandrosterone and pregnenolone, and may also have a role in cholesterol sulfation in human skin. The substrate specificity, nuclear localization, and tissue localization of SULT2B1b suggest a role in regulating the responsiveness of cells to adrenal androgens via their direct inactivation or by preventing their conversion to more potent androgens and estrogens.


Subject(s)
Cytosol/enzymology , Sulfotransferases/metabolism , Amino Acid Sequence , Animals , Breast/enzymology , Breast/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Nucleus , Cholesterol/chemistry , Cholesterol/metabolism , Choriocarcinoma/metabolism , Cloning, Molecular , Dehydroepiandrosterone/chemistry , Dehydroepiandrosterone/metabolism , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Placenta/metabolism , Pregnenolone/chemistry , Pregnenolone/metabolism , Protein Isoforms , Rats , Sequence Homology, Amino Acid , Skin/chemistry , Skin/metabolism , Subcellular Fractions , Substrate Specificity , Sulfates/metabolism , Sulfotransferases/genetics , Transcription, Genetic
2.
Biochem J ; 346 Pt 3: 857-64, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10698717

ABSTRACT

The sulphotransferase (SULT) gene family is involved with the conjugation of many small drugs, xenobiotics and endogenous compounds. In this report, we describe the cloning and expression of novel cDNAs from human and rat brain, which are structurally related to the SULTs. These cDNAs have been termed 'brain sulphotransferase-like' (BR-STL), because of their similarity to the SULTs and their selective expression in brain tissue. The proteins encoded by the human and rat BR-STL cDNAs (hBR-STL-1 and rBR-STL cDNA respectively), denoted as hBR-STL and rBR-STL, are 98% identical and 99% similar in sequence. The hBR-STL-1 cDNA contains an 852-nt open reading frame encoding a 284-amino-acid protein with a calculated molecular mass of 33083 Da. Northern-blot analyses of RNA isolated from eight human tissues indicate that the hBR-STL message is selectively expressed in brain. Characterization of different brain regions showed that message levels were highest in cortical brain regions. rBR-STL message levels were relatively low in brains of 1-day-old male and female rats, but increased to adult levels in RNA from 7-day-old rats, and remained at that level in adult animals. The hBR-STL and rBR-STL cDNAs were expressed in both Escherichia coli and Sf9 insect cells in the presence or absence of an N-terminal histidine-affinity tag (His-tag). Polyclonal antibodies were raised in chickens against purified His-tagged hBR-STL, and were used to detect the presence of rBR-STL in adult male and female rat brain cytosol. The high degree of sequence conservation, and the selective localization of the BR-STL message in brain, suggest an important function in the central nervous system.


Subject(s)
Brain/enzymology , Sulfotransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cloning, Molecular , DNA, Complementary , Female , Humans , Male , Molecular Sequence Data , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
3.
Chem Biol Interact ; 124(2): 133-47, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10670824

ABSTRACT

Low molecular weight dimethylcyclosiloxanes (DMCS) are important precursors in the synthesis of polydimethysiloxane polymers widely used in industry, and in medical and personal care products. The objective of this study was to characterize the ability of two DMCS, octamethylcyclosiloxane (D4) and decamethylcyclopentasiloxane (D5) to induce drug metabolizing enzymes in rats. Male and female Sprague-Dawley rats were administered 1, 5, 20, or 100 mg/kg D4 or D5 in corn oil daily by gavage for 4 days. Changes in the levels of activity and/or immunoreactivity of CYP1A1/2, CYP2B1/2, CYP3A1/2 and NADPH cytochrome P450 reductase in liver microsomes were examined. Significant increases were observed in the liver to body weight ratio in female rats administered either D4 or D5 at doses > or = 20 mg/kg. Increases in the liver to body weight ratio were observed in male rats treated with > or = 100 mg/kg D5 but not with D4. Relatively large increases in CYP2B1/2 enzymatic activity and immunoreactive protein were observed with increasing concentrations of both D4 and D5. Significant increases in 7-pentoxyresorufin O-depentylase (PROD) activity were also detected in male and female rats given D4 at doses > or = 5 mg/kg. D5 increased PROD activity in male rats at doses > or = 20 mg/kg and in female rats at doses > or = 5 mg/kg. 7-Ethoxyresorufin O-deethylase (EROD) activity was increased in both male and female rats receiving > or = 20 mg/kg D4 or > or = 5 mg/kg D5; however, no changes were detected in CYP1A1/2 immunoreactive protein in rats of either sex. D4 and D5 caused significant increases in CYP3A1/2 immunoreactive protein in only male rats treated with 100 mg/kg of either compound. However, significant increases were detected in CYP3A1/2 immunoreactive protein in female rats at D4 doses > or = 20 mg/kg and D5 doses > or = 5 mg/kg. Induction of NADPH cytochrome P-450 reductase immunoreactive protein was observed with D4 in female rats and in both male and female rats with D5. Induction of CYP2B/1/2, CYP3A1/2 and NADPH cytochrome P450 reductase was observed in rats treated with 50 mg/kg phenobarbital by intraperitoneal injection. Maximal CYP2B induction detected with D4 was approximately 50% of the increase observed with phenobarbital. In summary, D4 and D5 induced CYP2B1/2 in adult rat liver in a manner similar to that observed with phenobarbital; however, differences were observed between D4 and D5 in their ability to induce CYP3A1/2 and NADPH cytochrome P450 reductase. Female rats were more sensitive to the inductive properties of low doses of both DMCS than male rats whereas male rats were more responsive to phenobarbital induction.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Siloxanes/toxicity , Animals , Biocompatible Materials/toxicity , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP2B1/biosynthesis , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Female , Male , Mixed Function Oxygenases/biosynthesis , NADPH-Ferrihemoprotein Reductase/biosynthesis , Rats , Rats, Sprague-Dawley , Sex Characteristics , Steroid Hydroxylases/biosynthesis
4.
Drug Metab Dispos ; 27(9): 1057-63, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460806

ABSTRACT

The metabolism of the local anesthetics lidocaine and ropivacaine (ropi) involves several steps in humans. Lidocaine is mainly hydrolyzed and hydroxylated to 4-OH-2,6-xylidine (4-OH-xyl). The metabolism of ropi, involving dealkylation and hydroxylation, gives rise to 3-OH-ropi, 4-OH-ropi, 3-OH-2'6'-pipecoloxylidide (3-OH-PPX), and 2-OH-methyl-ropi. Because the metabolites are hydroxylated, they are particularly prone to subsequent Phase II conjugation reactions such as sulfation and glucuronidation. This study focused on the in vitro sulfation of these metabolites as well as another suspected metabolite of ropi, 2-carboxyl-ropi. All the metabolites were synthesized for the subsequent enzymatic studies. Five cloned human sulfotransferases (STs) were used in this study, namely, the phenol-sulfating form of ST (P-PST-1), the monoamine-sulfating form of ST (M-PST), estrogen-ST (EST), ST1B2, and dehydroepiandrosterone-ST (DHEA-ST), all of which are expressed in human liver. The results demonstrate that all of the metabolites except 2-OH-methyl-ropi and 2-carboxyl-ropi can be sulfated. It was also found that all of the STs can conjugate the remaining hydroxylated metabolites except DHEA-ST. However, there are large differences in the capacity of the individual human ST isoforms to conjugate the different metabolites. P-PST-1 sulfates 3-OH-PPX, 3-OH-ropi, and 4-OH-xyl; M-PST and EST conjugate 3-OH-PPX, 3-OH-ropi, and 4-OH-ropi whereas ST1B2 sulfates only 4-OH-xyl. The most extensively sulfated ropi metabolite is 3-OH-PPX. In conclusion, all of the hydroxylated metabolites of lidocaine and ropi can be sulfated if the hydroxyl group is attached to the aromatic ring in the metabolites. The human ST enzymes that are considered to be responsible for the sulfation of these metabolites in vivo are P-PST-1, M-PST, EST, and ST1B2. These enzymes are also found in the liver; this is the most important tissue for the metabolism of ropi in humans, demonstrated by.


Subject(s)
Amides/chemistry , Amides/metabolism , Anesthetics, Local/chemistry , Anesthetics, Local/metabolism , Lidocaine/chemistry , Lidocaine/metabolism , Sulfotransferases/metabolism , Amides/chemical synthesis , Anesthetics, Local/chemical synthesis , Biotransformation , Cytosol/enzymology , Humans , Hydroxylation , Isoenzymes/metabolism , Kinetics , Lidocaine/chemical synthesis , Mass Spectrometry , Recombinant Proteins/metabolism , Ropivacaine , Sulfates/chemical synthesis , Sulfates/metabolism
5.
J Steroid Biochem Mol Biol ; 68(3-4): 137-44, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10369411

ABSTRACT

Sulfation is an important conjugation reaction in the metabolism of steroids. Steroids sulfates do not interact with the appropriate hormone receptors; additionally, the presence of the charged sulfate moiety increases the aqueous solubility and excretion of most steroids. Estrogen sulfotransferase (EST) is the major form of human cytosolic ST involved in the conjugation of estrogens. EST is important in the inactivation of beta-estradiol (E2) during the luteal phase of the menstrual cycle. EST has a significantly higher affinity for the sulfation of E2 and 17alpha-ethinylestradiol (EE2) than for other potent estrogens such as diethylstilbestrol (DES) and equine estrogens. The ability of EST to sulfate these estrogenic compounds at physiologic concentrations is important in regulating their activation of the ER in estrogen responsive cells. Human Ishikawa endometrial adenocarcinoma (ISH) cells possess an estrogen receptor (ER)-regulated alkaline phosphatase (AlkPhos) which is used to assay ER activation. To study the effects of EST activity on the ER activation of different estrogenic compounds, ISH cells were stably transformed with an EST expression vector. Dose-response curves for the induction of AlkPhos activity by the different estrogenic compounds were generated with EST/ISH and control pcDNA/ISH cells. EST/ISH cells were 200-fold less sensitive to E2 and EE2 than were control cells. No differences were observed in the dose response curves for DES between EST/ISH and pcDNA/ISH cells. EST/ISH cells were approximately 3-10-fold less sensitive to the equine estrogens equilin and 17-equilin as compared to control cells. The ability of EST to decrease the ER activation of an estrogen correlates with the sulfation of these compounds at nanomolar concentrations by EST/ISH and pcDNA/ISH ISH cells. These results indicate that EST is capable of efficiently inactivating E2 and EE2 but is significantly less effective in inhibiting the ER binding of other potent estrogenic compounds.


Subject(s)
Adenocarcinoma/metabolism , Endometrial Neoplasms/metabolism , Estrogens/metabolism , Receptors, Estrogen/metabolism , Sulfotransferases/metabolism , Alkaline Phosphatase/metabolism , Cytosol/enzymology , Diethylstilbestrol/metabolism , Ethinyl Estradiol/metabolism , Female , Humans , Kinetics , Substrate Specificity , Tumor Cells, Cultured
6.
Chem Biol Interact ; 109(1-3): 329-39, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9566756

ABSTRACT

Understanding the factors which alter estrogen metabolism and activity in endometrial tissue is important because unopposed estrogen stimulation is an important risk factor in the development of endometrial carcinoma. The cyclic progression of the endometrium through proliferative and secretory phases is normally under the control of the ovarian hormones beta-estradiol (E2) and progesterone. One mechanism by which progesterone inhibits the activity of E2 in secretory endometrium is by elevating the degree of E2 sulfation, thereby reducing its ability to bind to the estrogen receptor and elicit a cellular response. Our laboratories have investigated the cytosolic sulfotransferases (STs) found in biopsies of both proliferative and secretory endometrium obtained from five normal pre-menopausal women who were not taking any drugs or steroids. Two of the human cytosolic STs were detected in human endometrial tissues. The phenol-sulfating form of phenol ST (P-PST) was found at varying levels in cytosol from both proliferative and secretory endometrium in all of the women studied but with no consistent correlation to the phase of the menstrual cycle. In contrast, estrogen ST (EST) was not detected in the proliferative endometrial cytosol of any of the women studied but was consistently found in all of the secretory endometrial cytosols. The presence and levels of these STs was confirmed by ST activity studies, immunoblot analysis and Northern blot analysis. These results indicate that the expression of EST in human endometrial tissues varies with the phase of the menstrual cycle and is most likely regulated by progesterone secreted from the ovaries.


Subject(s)
Endometrium/enzymology , Menstrual Cycle/metabolism , Sulfotransferases/isolation & purification , Sulfotransferases/metabolism , Adult , Blotting, Northern , Cytosol/enzymology , Female , Humans , Immunoblotting
7.
Mol Pharmacol ; 53(2): 274-82, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9463486

ABSTRACT

Sulfation is an important conjugation reaction for a wide range of endogenous and exogenous compounds in humans, including steroids, bile acids, catecholamine neurotransmitters and thyroid hormones. The cDNA for a distinct human cytosolic sulfotransferase (ST), hST1B2, has been isolated from a human liver lambdaZap cDNA library. The hST1B2 cDNA consists of 1144 bp and contains the coding region for a novel human cytosolic ST that has been termed hST1B2 on the basis of its sequence similarity to a rat sulfotransferase, ST1B1. The hST1B2 cDNA contains an 888-bp open reading frame that encodes a 296-amino acid protein with a calculated molecular mass of 34,897 Da. The hST1B2 cDNA also has a 127-bp 5' untranslated region (UTR) and a 129-bp 3'-UTR, including a 22-bp poly(A)+ tract. The amino acid sequence of hST1B2 is 74%, 53%, 53%, 52%, 56%, and 34% identical to the amino acid sequences of rat ST1B1 and human P-PST-1, P-PST-2, M-PST, EST, and DHEA-ST, respectively. Enzymatically active hST1B2 was expressed in the bacterial expression vector pKK233-2 for kinetic characterization and in the bacterial expression vector pQE-31, which generates a histidine-tagged fusion protein for the generation of antibodies. Expressed hST1B2 sulfates small phenols such as 1-naphthol and p-nitrophenol and thyroid hormones, including 3,3'-diiodothyronine, triiodothyronine, reverse triiodothyronine, and thyroxine. No activity was detected when several steroids or dopamine were tested as substrates. High levels of hST1B2 message were detected by Northern blot analysis in RNA isolated from human liver, colon, small intestine, and blood leukocytes. Immunoblot analysis detected a protein with the same mass as expressed hST1B2 in several human tissues that also possessed hST1B2 message. These results indicate that a novel cytosolic ST is present in human tissues, which may have an important role in thyroid hormone and xenobiotic metabolism.


Subject(s)
Liver/enzymology , Sulfotransferases/metabolism , Thyroid Hormones/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Cytosol/enzymology , DNA, Complementary/genetics , Gene Expression , Humans , Immunologic Techniques , Molecular Sequence Data , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
8.
Oncol Res ; 9(11-12): 589-96, 1997.
Article in English | MEDLINE | ID: mdl-9563006

ABSTRACT

Estrogen metabolism is closely associated with the growth, progression, and treatment of breast cancer because many breast cancers are dependent upon estrogens for both growth and progression. Factors that affect the intracellular metabolism of estrogens may be critical in altering the effects of estrogens on breast cancer cells. MCF-7 cells have been used as a model system to study the effects of estrogens on breast cancer cellular growth. Because normal human mammary epithelial (HME) cells contain estrogen sulfotransferase (EST), which is involved in the inactivation of estrogens via sulfation, and MCF-7 cells do not possess this enzyme, the absence of EST may be critical to the growth of MCF-7 cells in the presence of estrogens. To study the effects of EST on cellular growth, MCF-7 cells stably transformed with an EST expression vector were compared to control cells transformed with vector only. Sulfation of 20 nM E2 occurs more rapidly with MCF-7 cells transformed with EST than with the control cells, thereby rendering E2 physiologically inactive. Additionally, these EST/MCF-7 cells sulfate 20 nM 17 alpha-ethinylestradiol (EE2) at a rate similar to that for E2 but sulfate 20 nM diethylstibestrol (DES) much more slowly; these results correlate with the kinetic characteristics of EST for these steroids. EST/MCF-7 cells require higher concentrations of E2 to stimulate growth than do control MCF-7 cells, hypothetically because EST is inactivating E2 via sulfation, rendering it incapable of binding to the estrogen receptor (ER). The effects of EE2 are similar to those of E2 whereas DES is effective at lower concentrations because it is not inactivated by EST. Neither control nor EST/MCF-7 cells grow well in the complete absence of estrogens, as would be expected because MCF-7 cells are estrogen dependent. However, in medium that has not been treated to remove endogenous estrogens, EST/MCF-7 cells grow more slowly than control cells, most likely because EST is inactivating the estrogens in the medium, making them ineffective in stimulating growth. EST/MCF-7 cells possess EST at levels similar to HME cells and are less responsive to estrogens than are MCF-7 cells lacking EST. The loss of EST may be a factor in oncogenesis, which leads to altered estrogen metabolism in breast carcinoma cells.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Sulfates/metabolism , Breast Neoplasms/enzymology , Cell Transformation, Neoplastic/metabolism , Cytosol/enzymology , Cytosol/metabolism , Estrogens, Conjugated (USP)/metabolism , Female , Humans , Sulfotransferases/metabolism , Tumor Cells, Cultured
9.
Endocrinology ; 137(4): 1395-401, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8625916

ABSTRACT

During the secretory phase of the human menstrual cycle, the endometrium is minimally responsive to the estrogens secreted from the ovaries. Conjugation of beta-estradiol (E2) with sulfate is thought to be an important mechanism in the regulation of the levels of active E2 in endometrial tissue. Estrogen sulfation is reportedly increased during the secretory phase in response to the high levels of progesterone secreted by the ovaries. Estrogen sulfotransferase (hEST), a distinct form of human cytosolic sulfotransferase (ST) with an affinity for E2 and estrone at low nanomolar concentrations, has recently been cloned and expressed in mammalian cells and in bacteria (J Steroid Biochem Mol Biol 52:529, 1995). At least two other forms of human cytosolic ST, dehydroepiandrosterone ST (hDHEA-ST) and the phenol-sulfating form of phenol-ST (hP-PST), also conjugate estrogens but at micromolar concentrations. This report describes the specific induction of hEST in human Ishikawa endometrial adenocarcinoma cells by progesterone as a model for the increases in estrogen sulfation observed in women during the secretory phase of the menstrual cycle. Treatment of Ishikawa cells with 10 microns progesterone for 48 h resulted in a 7-fold increase in the sulfation of 20 nM E2. The sulfation of selective substrates for human dehydroepiandrosterone sulfotransferase (hDHEA-ST) and the two forms of phenol sulfotransferase (hP-PST, hM-PST) were not affected by treatment with progesterone. The levels of immunoreactive hEST and hEST mRNA in the Ishikawa cells were both increased by progesterone, whereas the levels of immunoreactive hDHEA-ST, hP-PST, and hM-PST were not altered. hEST activity was not induced by treatment of Ishikawa cells with varying concentrations of E2, testosterone, or cortisol. The induction of hEST by progesterone was inhibited by RU-486, indicating that progesterone is acting via the progesterone receptor. These results indicate that progesterone is capable of specifically inducing hEST and estrogen sulfation in human Ishikawa adenocarcinoma cells and suggest a mechanism for increasing estrogen sulfation in the endometrium during the secretory phase of the menstrual cycle.


Subject(s)
Adenocarcinoma/enzymology , Endometrial Neoplasms/enzymology , Progesterone/pharmacology , Sulfotransferases/metabolism , Adenocarcinoma/pathology , Endometrial Neoplasms/pathology , Estrone/metabolism , Female , Humans , Mifepristone/pharmacology , RNA, Messenger/metabolism , Sulfates/metabolism , Tumor Cells, Cultured
10.
Cancer Res ; 56(7): 1551-5, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8603401

ABSTRACT

Breast cancers require the presence of estrogens for the maintenance of growth at some time in the course of their development, as does normal breast tissue. Sulfation is an important process in the metabolism and inactivation of steroids, including estrogens, because the addition of the charged sulfonate group prevents the binding of the steroid to its receptor. Also, many of the therapeutic and chemopreventive agents used in the treatment of breast cancer are substrates for the sulfotransferases (STs). The activity and expression of four cytosolic STs, which are the human phenol-sulfating and monoamine-sulfating forms of phenol ST (PST), dehydroepiandrosterone ST, and estrogen ST (hEST), were assayed in normal breast cells and in breast cancer cell lines. ST activities and immunoreactivities were assayed in the estrogen receptor-positive human breast cancer cell lines ZR-75-1, T-47D and MCF-7; in the estrogen receptor-negative breast cancer cell lines BT-20, MDA-MB-468, and MDA-MB-231; and in normal human mammary epithelial cells. The PSTs were the most highly expressed ST activities in the breast cancer cell lines, although the levels of activity varied significantly. ZR-75-1 and BT-20 cells possessed the highest levels of activity of the human phenol-sulfating form of PST. The breast cancer cell lines showed only trace levels of dehydroepiandrosterone ST and hEST activities. In contrast, hEST was the only ST detectable in human mammary epithelial cells. Understanding the regulation of ST activity in these breast cancer and normal breast cell lines will improve our knowledge of the role of sulfation in breast cancer and provide a model with which to study the mechanism of action of estrogens in mammary cells.


Subject(s)
Breast Neoplasms/enzymology , Breast/enzymology , Sulfotransferases/metabolism , Blotting, Northern , Cytosol/enzymology , Epithelium/enzymology , Estradiol/metabolism , Female , Humans , Immunoblotting , Tumor Cells, Cultured
11.
Eur J Pharmacol ; 293(2): 173-81, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7589232

ABSTRACT

Human hydroxysteroid sulfotransferase, human phenol-sulfating form of phenol sulfotransferase, rat hydroxysteroid sulfotransferase a and rat phenol sulfotransferase IV were expressed in Escherichia coli. Cytosol preparations of transformed bacteria were used as activating systems in mutagenicity tests with Salmonella typhimurium TA98. All test compounds, 1-hydroxymethylpyrene, 2-hydroxymethylpyrene, 1-(1-pyrenyl)ethanol, 9-hydroxymethylanthracene, 7-hydroxymethyl-12-methylbenz[a]anthracene and 4H-cyclopenta[def]chrysen-4-ol, were activated by both hydroxysteroid sulfotransferases investigated. However, 1-(1-pyrenyl)ethanol was 67-fold more efficiently activated by the human enzyme, whereas 7-hydroxymethyl-12-methylbenz[a]anthracene was 27-fold more efficiently activated by the rat enzyme. The phenol sulfotransferases showed relatively low activities with the benzylic alcohols investigated. The only exception was 4H-cyclopenta[def]chrysen-4-ol, which was activated efficiently by rat phenol sulfotransferase IV. We had previously tested the ability of rat and human hepatic cytosol preparations to activate the same compounds. The results of a statistical analysis suggest that the activities of human hydroxysteroid sulfotransferase, rat hydroxysteroid sulfotransferase a and phenol sulfotransferase IV can account for a substantial portion of the activation of benzylic alcohols in human, female rat and male rat liver, respectively.


Subject(s)
Benzyl Alcohols/metabolism , Benzyl Alcohols/toxicity , Escherichia coli/enzymology , Escherichia coli/genetics , Mutagens/metabolism , Mutagens/toxicity , Sulfotransferases/metabolism , Animals , Base Sequence , Biotransformation , Cloning, Molecular , Cytosol/enzymology , DNA, Complementary/biosynthesis , Female , Humans , In Vitro Techniques , Liver/drug effects , Liver/enzymology , Male , Molecular Sequence Data , Mutagenicity Tests , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
12.
J Steroid Biochem Mol Biol ; 52(6): 529-39, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7779757

ABSTRACT

A distinct human estrogen sulfotransferase (hEST-1) cDNA has been isolated from a human liver lambda Zap cDNA library using a PCR procedure. The enzymatically active protein has been expressed in two bacterial expression systems and the kinetic and immunologic properties of the enzyme have been characterized. The full-length cDNA for hEST-1 is 994 base pairs in length and encodes a 294 amino acid protein with a calculated molecular mass of 35,123 Da. Purified hEST-1 migrated with an apparent molecular mass of 35,000 Da during SDS-polyacrylamide gel electrophoresis. Immunoblot analysis of hEST-1 expressed in E. coli with a rabbit anti-hEST-1 antibody yields a band of approximately 35,000 Da. The anti-hEST-1 antibody also detects a single band in human liver and jejunum cytosol which migrates with the same molecular mass as expressed hEST-1. There was also no cross-reactivity of hEST-1 with rabbit anti-hP-PST or rabbit anti-hDHEA-ST antibodies upon immunoblot analysis. hEST-1 was expressed in bacteria and purified to homogeneity. Expressed hEST-1 activity has a significantly greater affinity for estrogen sulfation than that found for the other human STs which conjugate estrogens. hEST-1 maximally sulfates beta-estradiol and estrone at concentrations of 20 nM. hEST-1 also sulfates dehydroepiandrosterone, pregnenolone, ethinylestradiol, and 1-naphthol, at significantly higher concentrations; however, cortisol, testosterone and dopamine are not sulfated. The results presented in this paper describe the expression and characterization of a human EST distinct from other human STs which sulfate estrogens. The high affinity of hEST-1 for estrogens indicates that this ST may be important in both the metabolism of estrogens and in the regulation of their activities.


Subject(s)
DNA, Complementary/genetics , Liver/enzymology , Sulfotransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , Escherichia coli/genetics , Estrogens/metabolism , Gene Expression , Humans , Kinetics , Molecular Sequence Data , Molecular Weight , Polymerase Chain Reaction , Rabbits , Sequence Homology, Amino Acid , Species Specificity , Substrate Specificity , Sulfotransferases/immunology , Sulfotransferases/metabolism
13.
J Steroid Biochem Mol Biol ; 52(1): 35-44, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7857871

ABSTRACT

A new isoform of rat liver estrogen sulfotransferase (EST), rEST-6, which is distinct from the previously reported rat EST [Demyan et al., Molec. Endocrinol. 6 (1992) 589], has been cloned, expressed, purified and characterized. A PCR procedure using oligonucleotide primers synthesized to the 5'-nontranslated and 3'-nontranslated regions of the published rEST sequence was used to isolate rEST-6 cDNA. The cloned DNA is 1000 bp in length and encodes a protein of 295 amino acids with a calculated molecular mass of 35,300 Da. rEST-6 is selectively expressed in male rats, as confirmed by Northern blot and immunoblot analyses. Northern blot analysis of male and female rat liver RNA with the rEST-6 cDNA as a probe shows a band with male RNA but not with female RNA. Similarly, immunoblot analysis of male and female rat liver cytosols with an antibody to rat EST yields a strong immunoreactive band in rat liver cytosol from male rats but not from females. Subsequent to bacterial expression and purification of rEST-6, the enzyme was analyzed kinetically and shown to sulfate estrogens but not dehydroepiandrosterone, pregnenolone, cortisol or testosterone. Maximal sulfation activity towards both beta-estradiol and estrone occurred at a concentration of 1 microM with substrate inhibition at higher concentrations. These results indicate that multiple, closely related forms of EST are present in rat liver. Analysis of the activity and regulation of these different EST enzymes is important in understanding estrogen metabolism in rats.


Subject(s)
Estradiol/metabolism , Isoenzymes/genetics , Sex Characteristics , Sulfotransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli/genetics , Female , Gene Expression , Liver/enzymology , Male , Molecular Sequence Data , Rats , Recombinant Proteins/biosynthesis , Sequence Homology, Amino Acid , Sulfotransferases/biosynthesis
14.
Blood ; 84(2): 650-6, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-7517724

ABSTRACT

In our initial immunochemical study of the red blood cell (RBC) membrane proteins targeted in 20 cases of warm-antibody autoimmune hemolytic anemia (AHA), RBC eluates of 6 patients mediated immunoprecipitation (IP) of both band 3 and glycophorin A (GPA). This dual IP pattern had previously been observed with murine monoclonal antibodies (MoAbs) against the high frequency blood group antigen, Wrb (Wright), suggesting that the Wrb epitope may depend on a band 3-GPA interaction. Earlier, anti-Wrb had been identified serologically as a prominent non-Rh specificity of AHA autoantibodies. In the present study, 6 autoantibody eluates immunoprecipitating band 3 and GPA from common Wr(b+) RBCs were retested, in parallel with murine anti-Wrb MoAbs, against very rare Wr(a+b-)En(a+)RBCs. One patient's autoantibodies were unreactive with the Wr(b-) RBCs by either IP or indirect antiglobulin test (IAT) and were judged to have "pure" anti-Wrb specificity. Two other patients' autoantibodies displayed both IP and serologic evidence for anti-Wrb as a major component in combination with one or more additional specificities. However, among 3 other patients whose autoantibodies coprecipitated band 3 and GPA, there was no reduction in IP or IAT reactivity with Wr(b-) RBCs in 2 and only slight reduction in the third. We conclude (1) that human anti-Wrb autoantibodies, like their murine monoclonal counterparts, coprecipitate band 3 and GPA from human RBCs; but (2) that not all antibodies with this IP behavior have anti-Wrb serologic specificity, as defined by this donor's Wr(b-) RBCs. The possibility of an additional (non-Wrb) RBC epitope dependent on a band 3-GPA interaction is raised.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/immunology , Autoantibodies/immunology , Blood Group Antigens/immunology , Erythrocytes/immunology , Glycophorins/immunology , Immunoglobulin G/immunology , Animals , Antibodies, Monoclonal/immunology , Epitopes , Humans , Mice , Precipitin Tests
15.
Chem Biol Interact ; 92(1-3): 57-66, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8033270

ABSTRACT

The cDNA for human liver phenol-sulfating phenol sulfotransferase (P-PST) has been cloned and the active enzyme expressed in Cos cells and bacteria. Analysis of the sequence identified two cysteine residues, one of which is highly conserved in the phenol sulfotransferase gene family. Previous studies with the pure human liver enzyme suggested that the conserved cysteine may be involved in binding substrates. Bacterial expression of P-PST with the cysteine converted to a serine indicates that the cysteine is not essential for activity or substrate binding, however, the mutant enzyme is significantly more sensitive to thermal inactivation.


Subject(s)
Arylsulfotransferase/metabolism , Amino Acid Sequence , Animals , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Base Sequence , Binding Sites , Cloning, Molecular , Cysteine/chemistry , Humans , Liver/enzymology , Molecular Sequence Data , Mutagenesis , Sequence Homology, Amino Acid , Serine/chemistry , Substrate Specificity , Temperature
16.
J Steroid Biochem Mol Biol ; 48(4): 369-75, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8142314

ABSTRACT

The human cytosolic sulfotransferases (STs), dehydroepiandrosterone sulfotransferase (DHEA-ST) and the phenol-sulfating form of phenol sulfotransferase, (P-PST), have been expressed in bacteria and used to investigate the ability of the cloned enzymes to conjugate steroids and related compounds. DHEA-ST was capable of sulfating all of the 3-hydroxysteroids, testosterone and estrogens tested as substrates. The 3-hydroxysteroids, androsterone, epiandrosterone and androstenediol, were conjugated at 50-60% of the rate of DHEA. Of the steroids tested, P-PST was capable of conjugating only the estrogens. The catechol estrogens, 2-hydroxyestradiol, 4-hydroxyestradiol and 4-hydroxyestrone, and compounds with estrogenic activity such as 17 alpha-ethynyl-estradiol and trans-4-hydroxytamoxifen, were also tested as substrates. DHEA-ST showed little or no sulfation activity with these compounds; however, all of these compounds were sulfated by P-PST. These results indicate that the expressed human STs are valuable in analyzing the overlapping substrate specificities of these enzymes and that P-PST may have an important role in the metabolism of estrogens and estrogenic compounds in human tissues.


Subject(s)
Arylsulfotransferase/metabolism , Steroids/metabolism , Sulfates/metabolism , Sulfotransferases/metabolism , Amino Acid Sequence , Androstenediol/metabolism , Androsterone/metabolism , Arylsulfotransferase/genetics , Base Sequence , Cytosol/enzymology , Escherichia coli/genetics , Estrogens/metabolism , Gene Expression , Humans , Molecular Sequence Data , Recombinant Proteins/metabolism , Substrate Specificity , Sulfotransferases/genetics , Testosterone/metabolism
17.
J Steroid Biochem Mol Biol ; 46(4): 481-7, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8217878

ABSTRACT

MCF-7 human mammary carcinoma cells have been reported to possess beta-estradiol and dehydroepiandrosterone sulfotransferase activities. These steroid sulfotransferase activities may be important in the metabolism and activity of different steroids in these cells. This report describes and characterizes both the enzymatic activity of three cytosolic sulfotransferases found in MCF-7 cells and the corresponding immunoblot analysis of these enzymes with specific anti-sulfotransferase antibodies. Two cytosolic sulfotransferases have been purified and characterized from human tissues which are capable of sulfating estrogens. These are the phenol-sulfating form of phenol sulfotransferase (P-PST) and the hydroxysteroid sulfotransferase, dehydroepiandrosterone sulfotransferase (DHEA-ST). The results of this study show that P-PST is the major cytosolic sulfotransferase found in MCF-7 cytosol and is responsible for most of the beta-estradiol sulfation in these cells. Although DHEA-ST activity was found in MCF-7 cytosol, this activity was only about 3% of the P-PST activity. Immunoblot analysis of MCF-7 cytosol detected both P-PST and lower levels of the monoamine-sulfating form of PST; however DHEA-ST could not be detected apparently because of low levels of expression. Human liver P-PST was expressed in Cos-7 Green monkey kidney fibroblasts and the ability of the cloned enzyme to sulfate beta-estradiol was investigated. This study indicates that P-PST is the prevalent cytosolic sulfotransferase in MCF-7 cytosol and is responsible for the majority of beta-estradiol sulfation in these cells.


Subject(s)
Arylsulfotransferase/metabolism , Breast Neoplasms/enzymology , Sulfotransferases/metabolism , Blotting, Western , Cytosol/enzymology , Estradiol/metabolism , Humans , In Vitro Techniques , Kinetics , Liver/enzymology , Substrate Specificity , Tumor Cells, Cultured
18.
J Clin Invest ; 91(4): 1672-80, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8473510

ABSTRACT

Immunoglobulin G (IgG) autoantibodies of 20 patients with autoimmune hemolytic anemia (AHA) were used in immunoaffinity assays with surface-radioiodinated human red blood cells (RBCs), and detergent-solubilized products were analyzed by SDS-PAGE/autoradiography. Four membrane proteins were identified as candidate autoantigens: a nonglycosylated polypeptide with an apparent molecular mass of 34 kD (p34) that was expressed in all available RBC phenotypes except Rhnull but differed consistently in apparent molecular mass from the 32-kD Rh(D) polypeptide co-isolated by IgG allo-anti-D; a heterogenous 37-55-kD glycoprotein, also deficient in Rhnull RBCs, which disappeared after deglycosylation by N-glycanase, with the appearance of a sharp, new approximately 31-kD band distinct from p34 and from Rh(D) polypeptide; a approximately 100-kD major membrane glycoprotein identified by immunoblotting as the band 3 anion transporter; and glycophorin A (GPA), also confirmed by immunoblotting. GP37-55 was not seen in the absence of p34, and both proteins are likely to be members of the Rh family. Indeed, a 34-kD polypeptide band and 37-55-kD poly-disperse "smear," isolated concurrently from the same labeled RBCs by IgG allo-anti-e, were indistinguishable from their autoantibody-isolated counterparts and may well be the same protein identified at different epitopes by the auto- and allo-antibodies. Individual AHA patients' autoantibodies isolated p34 and gp37-55, alone or in combination with band 3 (nine cases); strong band 3 alone (five cases); and combinations of band 3 with GPA (six cases). The autoantibodies of three additional patients whose AHA had been induced by alpha-methyldopa also isolated p34 and gp37-55.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Autoantibodies/immunology , Blood Proteins/immunology , Erythrocyte Membrane/immunology , Isoantibodies/immunology , Membrane Proteins/immunology , Rh-Hr Blood-Group System , Anion Exchange Protein 1, Erythrocyte/analysis , Blood Proteins/analysis , Erythrocyte Membrane/chemistry , Glycophorins/analysis , Humans , Membrane Proteins/analysis , Methyldopa/immunology , Rh-Hr Blood-Group System/chemistry
19.
Biochem J ; 289 ( Pt 1): 233-40, 1993 Jan 01.
Article in English | MEDLINE | ID: mdl-7678732

ABSTRACT

Dehydroepiandrosterone sulphotransferase (DHEA-ST) catalyses the 3'-phosphoadenosine 5'-phosphosulphate-dependent sulphation of a wide variety of steroids in human liver and adrenal tissue and is responsible for most, if not all, of the sulphation of bile acids in human liver. This report describes the isolation, characterization and expression of a cDNA which encodes human liver DHEA-ST. The DHEA-ST cDNA, designated DHEA-ST8, was isolated from a Uni-Zap XR human liver cDNA library and is composed of 1060 bp and contains an open reading frame encoding a 285-amino-acid protein with a molecular mass of approx. 33765 Da. Translation of DHEA-ST8 in vitro generated a protein identical in molecular size with that of DHEA-ST. Expression of DHEA-ST8 in COS-7 cells produces an active DHEA-ST protein which is capable of sulphating DHEA, has the same molecular mass as human liver DHEA-ST and is recognized by rabbit anti-(human liver DHEA-ST) antibodies. Northern-blot analysis of human liver RNA detects the presence of three different size transcripts; however, Southern-blot analysis of human DNA suggests that only one gene may be present in the genome. These results describe the cloning of a human ST which has an important role in the sulphation of steroids and bile acids in human liver and adrenals.


Subject(s)
Liver/enzymology , Sulfotransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Cell Line, Transformed , Chlorocebus aethiops , Cloning, Molecular , DNA , Humans , Molecular Sequence Data , Protein Biosynthesis , RNA/genetics , Rats , Sequence Homology, Amino Acid , Transcription, Genetic
20.
J Biochem Toxicol ; 7(4): 241-8, 1992.
Article in English | MEDLINE | ID: mdl-1293312

ABSTRACT

The bioactivation of 7-hydroxymethyl-12-methylbenz[a]anthracene (HMBA) to an electrophilic sulfuric acid ester metabolite has been shown to be catalyzed by rat liver bile acid sulfotransferase I (BAST I). The sulfation and activation of HMBA by BAST I was determined by the ability of sulfated HMBA to form DNA adducts. The BAST I was also shown to react with rabbit anti-human dehydroepiandrosterone sulfotransferase antisera and to represent a major form of hydroxysteroid/bile acid sulfotransferase in female rat liver cytosol. Higher levels of BAST I activity and immunoreactivity as well as HMBA-DNA adduct formation were detected in female rat liver cytosol than in male rat liver cytosol. The bioactivation of HMBA by pure BAST I was dependent on the presence of 3'-phosphoadenosine 5'-phosphosulfate (PAPS) in the reaction and was inhibited by dehydroepiandrosterone, a physiological substrate for BAST I. Glutathione, a cellular nucleophile with important protective properties, decreased DNA adduct formation in the HMBA sulfation reaction in the absence of glutathione S-transferase activity. These results indicate the usefulness of BAST I to investigate the sulfation and activation of HMBA and probably other hydroxymethylated polyaromatic hydrocarbons to electrophilic and mutagenic metabolites under defined reaction conditions.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/analogs & derivatives , Liver/enzymology , Sulfotransferases/metabolism , 9,10-Dimethyl-1,2-benzanthracene/metabolism , Animals , Biotransformation , Cytosol/enzymology , DNA/metabolism , Female , Male , Rats , Rats, Sprague-Dawley , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...