SULT1A1 catalyzes 2-methoxyestradiol sulfonation in MCF-7 breast cancer cells. (9/185)

In a previous study of nine human breast-derived cell lines, rates of metabolism of 17beta-estradiol (E(2)) were greatly enhanced when cultures were exposed to the aromatic hydrocarbon receptor agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin. Elevated rates of E(2) hydroxylation at the C-2, -4, -6alpha and -15alpha positions were observed concomitant with the induction of cytochromes P450 1A1 and 1B1. In each cell line, 2- and 4-hydroxyestradiol (2- and 4-OHE(2)) were converted to 2- and 4-methoxyestradiol (2- and 4-MeOE(2)) by the action of catechol O:-methyltransferase. In this study, conjugation of these estrogen metabolites was investigated. A comparison of the levels of metabolites determined with and without prior treatment of the media with a crude beta-glucuronidase/sulfatase preparation showed that most of the 2-MeOE(2) present was in conjugated form, whereas 4-MeOE(2), 6alpha-OHE(2) and 15alpha-OHE(2) were minimally conjugated. Inhibitor studies suggested that it was the sulfatase activity of the preparation that hydrolyzed the 2-MeOE(2) conjugates in MCF-7 cell media; the presence of 2-MeOE(2)-3-sulfate in MCF-7 culture media was confirmed by electrospray ion-trap mass spectrometry. To identify the enzyme catalyzing this conjugation, the expression of mRNAs encoding five sulfotransferases (SULT1A1, SULT1A2, SULT1A3, SULT1E1 and SULT2A1) was evaluated in the nine cell lines by use of the reverse transcription-polymerase chain reaction. Only expression of SULT1A1 mRNA correlated with the observed conjugation of nanomolar levels of 2-MeOE(2) in these cell lines. Cloning and sequencing of SULT1A1 cDNA from MCF-7 cells revealed that mRNAs encoding two previously identified allelic variants, SULT1A1*1 ((213)Arg) and SULT1A1*2 ((213)His), were expressed in these cells. Heterologous cDNA-directed expression of either variant in MDA-MB-231 cells, which do not normally express SULT1A1, conferred 2-MeOE(2) sulfonation activity. The SULT1A1 allelic variants were also expressed in SF:9 insect cells, from which post-microsomal supernatants were used to determine K:(m) values of 0.90 +/- 0.12 and 0.81 +/- 0.06 microM for SULT1A1*1 and SULT1A1*2, respectively, with 2-MeOE(2) as substrate. These results show that SULT1A1 is an efficient and selective catalyst of 2-MeOE(2) sulfonation and, as such, may be important in modulating the anticarcinogenic effects of 2-MeOE(2) that have been described recently.  (+info)

Phenol sulfotransferases: hormonal regulation, polymorphism, and age of onset of breast cancer. (10/185)

In recent years, significant effort has been made to identify genes that influence breast cancer risk. Because the high-penetrance breast cancer susceptibility genes BRCA1 and 2 play a role only in a small fraction of breast cancer cases, understanding the genetic risk of the majority of breast cancers will require the identification and analysis of several lower penetrance genes. The estrogen-signaling pathway plays a crucial role in the pathophysiology of breast cancer; therefore, polymorphism in genes involved in this pathway is likely to influence breast cancer risk. Our detailed analysis of gene expression profiles of estrogen- and 4-OH-tamoxifen-treated ZR75-1 breast cancer cells identified members of the sulfotransferase 1A (SULT1A) phenol sulfotransferase family as downstream targets of tamoxifen. On the basis of the induction of SULT1A by 4-OH-tamoxifen and the known inherited variability in SULT1A enzymatic activity, we hypothesized that polymorphism in sulfotransferase genes might influence the risk of breast cancer. Using an RFLP that distinguishes an arginine to histidine change in exon 7 of the SULT1A1 gene, we characterized SULT1A1 genotypes in relation to breast cancer risk. An analysis of 444 breast cancer patients and 227 controls revealed no effect of SULT1A1 genotype on the risk of breast cancer (P = 0.69); however, it did appear to influence the age of onset among early-onset affected patients (P = 0.04). Moreover, individuals with the higher activity SULT1A1*1 allele were more likely to have other tumors in addition to breast cancer (P = 0.004; odds ratio, 3.02; 95% confidence interval, 1.32, 8.09). The large number of environmental mutagens and carcinogens activated by sulfotransferases and the high frequency of the SULT1A1*1 allele in human populations warrants additional studies to address the role of SULT genes in human cancer.  (+info)

Effects of organic solvents on the activities of cytochrome P450 isoforms, UDP-dependent glucuronyl transferase, and phenol sulfotransferase in human hepatocytes. (11/185)

We studied the effects of acetonitrile, dimethyl sulfoxide (DMSO), and methanol (MeOH) in human hepatocytes on cytochrome P450 (CYP) and phase II conjugation activities: phenacetin O-deethylation (CYP1A2), coumarin 7-hydroxylation (CYP2A6), tolbutamide 4-hydroxylation (CYP2C9), S-mephenytoin 4'-hydroxylation (CYP2C19), dextromethorphan O-demethylation (CYP2D6), chlorzoxazone 6-hydroxylation (CYP2E1), testosterone 6beta-hydroxylation (CYP3A4), and umbelliferone glucuronidation and sulfation. The solvents were evaluated at concentrations (v/v) of 0.1, 1, and 2%. Previously cryopreserved human hepatocytes pooled from multiple donors were used as suspension cultures in this study. DMSO was found to inhibit CYP2C9 and CYP2C19, CYP2E1, and CYP3A4 in a concentration-dependent manner. At 2% DMSO, the activities for the four isoforms were approximately 40% (CYP2C9), 23% (CYP2C19), and 11% (CYP2E1) of that observed for 0.1% acetonitrile and 45% (CYP3A4) of that observed for 1% acetonitrile. No apparent inhibitory effects were observed for the other activities evaluated. Methanol was found to inhibit CYP2C9 and CYP2E1 activities, but to a lesser extent than DMSO. Acetonitrile had no apparent effects on any of the on any of the activities evaluated. These findings should be considered when choosing an organic solvent for metabolism studies with human hepatocytes.  (+info)

Enzymatic characterization and interspecies difference of phenol sulfotransferases, ST1A forms. (12/185)

Cytosolic sulfotransferases, which mediate activation and detoxification of both endogenous and exogenous compounds, consist of at least five different gene families (ST1 to ST5) in mammals. Several cDNAs corresponding to ST1A forms have been reported, but their functional properties are not well characterized. In addition, only a single form of ST1A sulfotransferase has been reported in each experimental animal species despite the expressions of plural forms in humans. Therefore, enzymatic properties of human ST1A3, ST1A5, rat ST1A1, mouse St1a4, and newly isolated rabbit ST1A8 have been characterized and compared by use of their recombinant proteins to clarify the functional difference between human and experimental animal ST1A forms. From the results using more than 25 phenolic chemicals, all the experimental animal ST1A forms showed substrate specificities similar to human ST1A3 rather than ST1A5. They showed high affinities toward p-nitrophenol and 6-hydroxymelatonin as found in human ST1A3. These forms also showed high activities toward umbelliferone and naringenin, but very low activities toward catecholamines, representative substrates of human ST1A5. Hepatic contents of experimental animal ST1A forms varied (66-250 pmol/mg of cytosolic protein) but showed the same order as observed with human ST1A3 (120 pmol/mg). Hepatic content of human ST1A5 was about 19-fold less than that of ST1A3. Therefore, ST1A forms identified in experimental animal species correspond to human ST1A3 functionally. For chemicals such as troglitazone and 2-amino-4'-hydroxy-1-methyl-6-phenylimidazo[4,5-b]pyridine, clear species differences were detected among the ST1A forms examined.  (+info)

Sulfation is rate limiting in the futile cycling between estrone and estrone sulfate in enriched periportal and perivenous rat hepatocytes. (13/185)

The metabolic activities and tissue binding of estrone (E1) and estrone sulfate (E1S) on futile cycling were examined. Desulfation of E1S in the 9000g supernatant fraction (S9) of periportal (PP) and perivenous (PV) rat hepatocytes were of similar V (2.9 +/- 1.0 and 2.4 +/- 0.9 nmol/min/mg of S9 protein), K (30.4 +/- 8.3 and 34.8 +/- 6.6 microM), and desulfation intrinsic clearances (V/K of 77 and 55 microl/min/10(6) cells). The intrinsic clearance towards E1 sulfation (1 microM) in cytosolic preparations of PV hepatocytes was 4 times that of PP hepatocytes (V/K of 26.4 +/- 9.5 versus 6.1 +/- 2.2 microl/min/mg of cytosolic protein or 13 +/- 5 versus 3.1 +/- 1.1 microl/min/10(6) cells). The observation was consistent with the immunolocalization of estrogen sulfotransferase (PV/PP ratio of 3.4 +/- 1.1) but not hydroxysteroid sulfotransferase (PV/PP ratio of 0.29 +/- 0.21) nor phenol sulfotransferase (PV/PP ratio of 1.13 +/- 0.23). Upon incubation of E1S (1-125 microM) with hepatocytes (30 min), higher concentrations of E1S and E1 were observed within PP than in PV cells, and saturation was evident at the higher concentrations. Based on the in vitro metabolic and tissue binding parameters for E1S and E1 and the published zonal uptake clearances of E1S (116 microl/min/10(6) cells), fitting revealed that uptake of E1 (1484 and 1463 microl/min/10(6) cells) by PP and PV cells was rapid and similar, and E1 sulfation was the slowest step in futile cycling. The greater metabolism of E1 in PV region led to higher levels of E1 and E1S in PP hepatocytes, and the nonlinear uptake, binding, and vesicular accumulation of E1S resulted in different t(1/2) values for E1S and E1.  (+info)

Sulfotransferase 1A1 polymorphism, endogenous estrogen exposure, well-done meat intake, and breast cancer risk. (14/185)

Sulfotransferase (SULT) 1A1 is involved in the inactivation of estrogens and bioactivation of heterocyclic amines and polycyclic aromatic hydrocarbons. A G--->A transition at codon 213 (CGC/Arg to CAC/His) of the SULT1A1 gene was reported recently, and individuals homozygous for the His allele have a substantially lower activity of this enzyme than those with other genotypes. We hypothesized that the His allele may be a risk factor for breast cancer, particularly among women who had risk factors related to higher endogenous estrogen exposure. This hypothesis was investigated in a case-control study conducted in a cohort of postmenopausal Iowa women who completed a mailed questionnaire in 1986 on lifestyle factors including information on major breast cancer risk factors. DNA samples and information related to well-done meat intake were obtained from breast cancer cases diagnosed between 1992 and 1994 and a random sample of cancer-free cohort members. Multivariate analysis was performed on data from 156 cases and 332 controls who donated a blood sample. The frequency of the His allele was 41.6% in cases and 34.1% in controls (P = 0.03), and the risk of breast cancer was increased with the number of His alleles (P for trend = 0.02). Compared with women with the Arg/Arg genotype, an 80% elevated risk was observed among women homozygous for the His allele (95% confidence interval, 1.0-3.2; P = 0.04). This positive association was more pronounced among women who drank alcohol and had a high body mass index, early age at menarche, and late age at menopause, factors related to high endogenous estrogen exposure, than among those who did not have these risk factors. The risk of breast cancer was elevated with increasing doneness level of red meat intake among women with the Arg/Arg genotype (P for trend, 0.01) or the Arg/His genotype (P for trend, 0.10), whereas this association was not evident for women with the His/His genotype. The results from this study suggest that homozygosity for the SULT1A1 His213 allele may be a risk factor for breast cancer, and its effect may be modified by the exposure level of endogenous estrogens and heterocyclic amines.  (+info)

Cloning and sequencing of the astA gene encoding arylsulfate sulfotransferase from Salmonella typhimurium. (15/185)

Arylsulfate sulfotransferase (ASST) transfers a sulfate group from a phenolic sulfate ester to a phenolic acceptor substrate. In the present study, the gene encoding ASST was cloned from a genomic library of Salmonella typhimurium. The gene was subcloned into the vector pKF3 and was sequenced. A recombinant clone harboring the gene was directly identified using a fluorescent assay. Sequencing revealed two contiguous open reading frames (ORFs) on the same strand. Based on amino acid sequence homology, ORF1 and ORF2 are designated as astA and dsbA, respectively. The deduced amino acid sequence of astA from S. typhimurium was highly similar to those of the Enterobacter amnigenus, Klebsiella, and Campylobacter jejuni ASSTs, encoded by the astA genes. However, an ASST activity assay revealed a different acceptor specificity. Using p-nitrophenyl sulfate (PNS) as a donor substrate, phenol is the best acceptor substrate, followed by alpha-naphthol, resorcinol, tyramine, acetaminophen, and tyrosine.  (+info)

Transcriptional regulation of rat hepatic aryl sulfotransferase (SULT1A1) gene expression by glucocorticoids. (16/185)

The 5'-flanking region [1892 base pairs (bp)] of the rat aryl sulfotransferase (SULT1A1) gene was cloned and the cis-acting sequences involved in glucocorticoid-inducible SULT1A1 gene transcription were characterized. SULT1A1 promoter and 5'-flanking sequences lacked a TATA box and a consensus glucocorticoid response element. Using a 5'-rapid amplification of cDNA ends approach, four SULT1A1 transcription start sites were identified. Transient transfection studies with SULT1A1-5':luciferase reporter constructs in primary cultured rat hepatocytes revealed that treatment with the potent glucocorticoid dexamethasone (10(-9)-10(-5) M) produced concentration-dependent increases in luciferase activity in constructs containing from 1892 to 119 bp of the SULT1A1 5'-flanking region. Relative to the most upstream SULT1A1 transcription start site, the minimal cis-acting sequences that were required for dexamethasone-inducible SULT1A1 expression were located between -84 and -69 bp. Treatment of transfectants with a panel of steroids, including dexamethasone, triamcinolone acetonide, hydrocortisone, dihydrotestosterone, 17beta-estradiol, and pregnenolone-16alpha-carbonitrile, revealed that steroid-inducible SULT1A1 gene expression was specific for glucocorticoid-class steroids. Concentration-response studies, coupled with a robust inhibition of glucocorticoid-inducible SULT1A1-5':luciferase reporter activity by antiglucocorticoid/antiprogestin RU-486, recapitulated earlier findings on endogenous SULT1A1 gene expression and implicated a major role for the glucocorticoid receptor transcription factor in the regulation of glucocorticoid-inducible SULT1A1 gene expression.  (+info)