Structural formula of estrone
Find information on thousands of medical conditions and prescription drugs.

Estrone

Estrone is an estrogenic hormone secreted by the ovary. Its molecular formula is C18H22O2. Estrone has a melting point of 254.5 degrees Celsius.

Synthesis

Estrone is synthesized from androstenedione, a derivative of progesterone. The conversion consists of the de-methylation of C-19 and the aromaticity of the 'A' ring. This reaction is similar to the conversion of testosterone to estradiol.

Home
Diseases
Medicines
A
B
C
D
E
E-Base
Ecstasy (drug)
Edecrin
Edrophonium
Edrophonium chloride
Efavirenz
Effexor
Eflornithine
Elavil
Eldepryl
Elidel
Eligard
Elitek
Elixomin
Elixophyllin
Ellagic acid
Elmiron
Eloxatin
Elspar
Emtriva
Emylcamate
Enalapril
Enalaprilat
Enalaprilat
Endep
Enflurane
Enoxaparin sodium
Entacapone
Enulose
Epi-pen
Epinephrine
Epirubicin
Epitol
Epivir
Epogen
Eprosartan
Ergocalciferol
Ergoloid Mesylates
Ergotamine
Eryc
Eryped
Erythromycin
Esgic
Eskalith
Esmolol
Estazolam
Estazolam
Estrace
Estraderm
Estradiol
Estradiol
Estradiol valerate
Estring
Estrogel
Estrone
Estrostep
Ethacridine
Ethambutol
Ethchlorvynol
Ethosuximide
Ethotoin
Etiracetam
Etodolac
Etopophos
Etoposide
Etorphine
Evista
Exelon
Exemestane
Hexal Australia
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z

Read more at Wikipedia.org


[List your site here Free!]


Organotins disrupt the 11[beta]-hydroxysteroid dehydrogenase type 2-dependent local inactivation of glucocorticoids
From Environmental Health Perspectives, 11/1/05 by Atanas G. Atanasov

Organotins, important environmental pollutants widely used in agricultural and industrial applications, accumulate in the food chain and induce imposex in several marine species as well as neurotoxic and immunotoxic effects in higher animals. Reduced birth weight and thymus involution, observed upon exposure to organotins, can also be caused by excessive glucocorticoid levels. We now demonstrate that organotins efficiently inhibit 11[beta]-hydroxysteroid dehydrogenase type 2 (11[beta]-HSD2), converting active 11[beta]-hydroxyglucocorticoids into inactive 11-ketoglucocorticoids, but not 11[beta]-HSD1, which catalyzes the reverse reaction. Di- and tributyltin as well as di--and triphenyltin inhibited recombinant and endogenous 11[beta]-HSD2 in lysates and intact cells with [IC.sub.50] values between 500 nM and 3 [micro]M. Dithiothreitol protected 11[beta]-HSD2 from organotin-dependent inhibition, indicating that organotins act by binding to one or more cysteines. Mutational analysis and 3-D structural modeling revealed several important interactions of cysteines in 11[beta]-HSD2. [Cys.sup.90], [Cys.sub.228], and [Cys.sup.264] were essential for enzymatic stability and catalytic activity, suggesting that disruption of such interactions by organotins leads to inhibition of 11[beta]-HSD2. Enhanced glucocorticoid concentrations due to disruption of 11[beta]-HSD2 function may contribute to the observed organotin-dependent toxicity in some glucocorticoid-sensitive tissues such as thymus and placenta. Key words: cortisol, dibutyltin, 11[beta]-hydroxysteroid dehydrogenase, glucocorticoid, inhibition, organotin, toxicity, tributyltin, triphenyltin.

**********

Organotins belong to the most widely used organometallic compounds, with an estimated annual production of approximately 50,000 tons. Derivatives of dialkyltin compounds such as dibutyltin (DBT), diphenyltin (DPT), and dioctyltin (DOT) are used in industry as stabilizers in polyvinyl chloride (PVC) and as catalysts in various products, whereas trialkyltins, including tributyltin (TBT) and triphenyhin (TPT) are used in agriculture as fungicides and pesticides and as antifouling agents for large ships (Fent 1996). Organotins are ubiquitous environmental pollutants especially relevant for water ecosystems. Accumulation of these lipophilic compounds has been observed in various species of snails, mussels, and fish (Bhosle et al. 2004; Coelho et al. 2002), causing an increased incidence of sterility or imposex (imposition of male sex characters onto the female) (Evans and Nicholson 2000; Fent 2003).

The main sources of organotin intake for humans are seafood contaminated because of the exposure to antifouling agents (Takahashi et al. 1999), and drinking water contaminated because of the leaching from PVC water pipes (Sadiki and Williams 1999). Additional sources are indoor dust, and liquids stored in plastic containers, including various alcoholic beverages (Liu and Jiang 2002). In higher species, including mammals, organotins tend to accumulate in certain organs, namely liver, kidney, and brain (Fait et al. 1994). Organotins efficiently penetrate through the skin and easily cross the placenta and blood-brain barrier (Adeeko et al. 2003; Cooke et al. 2004; Hasan et al. 1984).

Comparison of the effects of various tri-alkyltins indicated that the compounds with short alkyl groups such as trimethyltin (TMT) and triethyhin were mainly neurotoxic, whereas organotins with alkyl chains of intermediate length (tripropyltin and TBT) were primarily immunotoxic (Snoeij et al. 1985). The higher trialkyhin homologs trihexyhin and trioctyhin were found to be only slightly toxic; however, further metabolism in vivo converted them to their dialkyltin forms, which are also highly immunotoxic (Penninks et al. 1985; Seinen and Willems 1976; Snoeij et al. 1988). A single oral dose of DOT, DBT, or TBT induces a dose-related reduction of the relative thymus weight in rats, and impaired cell-mediated immunity was observed after dietary exposure to TPT for several weeks (Krajnc et al. 1984; Seinen et al. 1977a, 1977b; Snoeij et al. 1988; Vos et al. 1984a, 1984b, 1990). Furthermore, exposure of pregnant rats to organotins causes reduced birth weight (Adeeko et al. 2003; Cooke et al. 2004; Crofton et al. 1989).

Reduced birth weight has also been observed with prolonged intrauterine glucocorticoid exposure (Benediktsson et al. 1993; Lindsay et al. 1996a, 1996b; Stewart et al. 1995). After such an insult, circulating cortisol levels remained elevated throughout adult life, indicating a permanently disturbed regulation of the hypothalamic-pituitary-adrenal axis, which leads to a higher susceptibility for cardiovascular and metabolic disorders including obesity, insulin resistance, and type II diabetes (Drake et al. 2005; Seckl et al. 2000). In the placenta the fetus is protected from the high maternal glucocorticoid concentration through the activity of 11[beta]-hydroxysteroid dehydrogenase type 2 (11[beta]-HSD2), which converts active 11[beta]-hydroxyglucocorticoids (cortisol in human, corticosterone in rodents) into inactive 11-ketoglucocorticoids (cortisone in human, 11-dehydrocorticosterone in rodents) (reviewed in Stewart and Krozowski 1999). Impaired 11[beta]-HSD2 activity, due to mutations or the presence of inhibitors such as glycyrrhetinic acid (GA), strongly correlates with reduced birth weight and metabolic complications in later life of the offspring (Drake et al. 2005; Lindsay et al. 1996b; Odermatt 2004; Seckl et al. 2000).

Moreover, exposure of rats to excessive levels of glucocorticoids causes thymus involution (Schuurman et al. 1992), a phenomenon also evident after exposure to organotins. Treatment of rats with high doses of the 11[beta]-HSD inhibitor GA led to a significant elevation of systemic glucocorticoid levels accompanied by thymocyte apoptosis (Horigome et al. 1999).

Despite the fact that both exposure to excessive levels of organotins and glucocorticolds cause low birth weight and thymus involution in animal models, the impact of organotins on the control of the intracellular availability of glucocorticoids has not been studied. Therefore, we investigated the effect of various organotins on the activities of 11[beta]-HSD1, converting inactive 11-ketoglucocorticoids to active 11 [beta]-hydroxyglucocorticoids, and of 11[beta]-HSD2, catalyzing the opposite reaction. We also studied the mechanism of organotin-dependent inhibition of 11[beta]-HSD2.

Materials and Methods

Chemicals and reagents. We purchased [1,2,6,7-[sup.3]H]-cortisol, [2,4,6,7-[sup.3]H]-estrone, and [2,4,6,7-[sup.3]H]-estradiol from Amersham Pharmacia (Piscataway, NJ, USA); [1,2,6,7[sup.3]H]-cortisone from American Radiolabeled Chemicals (St. Louis, MO, USA); cell culture media and supplements from Invitrogen (Carlsbad, CA, USA); and steroid hormones from Steraloids (Wilton, NH, USA). All other chemicals were obtained from Fluka AG (Buchs, Switzerland) and were of the highest grade available. Organotins were dissolved in dimethyl sulfoxide (DMSO) and stored as 20-mM stock solutions at -70[degrees]C. Human 11[beta]-HSD1 and 11[beta]-HSD2 expression constructs in pcDNA3 vector (Invitrogen) were described previously (Odermatt et al. 1999). Plasmids containing cDNA from human 17[beta]-HSD1 or 17[beta]-HSD2, kindly provided by Stefan Andersson, were recloned into pcDNA3 vector by PCR with primers at the 5' end containing a HindIII restriction site (17[beta]-HSD1) or a BamHI restriction site (17[beta]-HSD2), a Kozak consensus sequence (Kozak 1989) and the initiation codon, and primers at the 3' end containing a stop codon followed by an XbaI restriction site. All constructs were verified by sequencing.

Cell culture. HEK-293 (human embryonic kidney) cells stably transfected with FLAG (Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys)-tagged human 11 [beta]-HSD2 (Schweizer et al. 2003) were grown at 37[degrees]C under 5% carbon dioxide to 60-70% confluence in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal calf serum, 4.5 g/L glucose, 50 U/mL penicillin/streptomycin, and 2 mM glutamine. SW-620 (human colorectal adenocarcinoma) cells and JEG-3 (human choriocarcinoma) cells were cultured according to the recommendations of the supplier (American Type Culture Collection, Manassas, VA, USA). The recently described RCCD-2 aldosterone-sensitive rat cortical collecting duct cells (Djelidi et al. 2001) were cultured in DMEM/Ham's F-12 (1:1), 14 mM NaHC[O.sub.3], 2 mM glutamine, 10 U/mL penicillin/streptomycin, and 20 mM HEPES, pH 7.4.

Activity assays in cell lysates. To measure 11[beta]-HSD2 activity, stably transfected HEK-293 cells (Schweizer et al. 2003) were grown in 10-cm culture dishes to 90% confluence. Cells were rinsed 3 times with phosphate-buffered saline (PBS) and resuspended in 2 mL ice-cold buffer TS2 (100 mM NaCl, 1 mM EGTA, 1 mM EDTA, 1 mM Mg[Cl.sub.2], 250 mM sucrose, 20 mM Tris-HCl, pH 7.4). Aliquots of the cell suspension were frozen at -20[degrees]C, retaining full enzymatic activity for at least 3 months. For determination of 11[beta]-HSD2 activity, aliquots were thawed, sonicated, and diluted 1:12 in buffer TS2 (4[degrees]C). We carried out reactions in a final volume of 20 [micro]L containing 10 nCi [1,2,6,7-[sup.3]H]-cortisol, 400-[micro]M [NAD.sup.+], and different concentrations of unlabeled cortisol. Final cortisol concentrations were 40 nM for measurements of inhibitors and ranged between 10 nM and 200 nM for determination of apparent [K.sub.m] values. Incubations were for 10 min at 37[degrees]C.

For determination of 11[beta]-HSD1, 17[beta]-HSD1, and 17[beta]-HSD2 activity, HEK-293 cells transfected by the calcium-phosphate precipitation method were harvested 48 hr later, washed with PBS, and centrifuged for 3 min at 150 x g. Supernatants were removed, cell pellets quick-frozen in a dry ice ethanol bath, and stored at -70[degrees]C. For assaying 11[beta]-HSD1, we dissolved pellets in TS2 buffer; for 17[beta]-HSD1 or 17[beta]-HSD2, we used a buffer containing 50 mM potassium phosphate, 20% glycerol, and 1 mM EDTA. We measured 11[beta]-HSD 1 oxoreductase activity as described recently (Atanasov et al. 2004), using radiolabeled cortisone as substrate. 17[beta]-HSD1 and 17[beta]-HSD2 activities were measured in the presence of radiolabeled estrone or estradiol and unlabeled steroid at final concentrations of 200 nM and 500 [micro]M NADPH or [NAD.sup.+], respectively.

Determination of 11[beta]-HSD2 activity in intact cells and MTT cytotoxicity assay. HEK-293 cells stably transfected with 11[beta]-HSD2 (25,000 cells per well) were seeded 24 hr prior to the assay in poly-D-lysine coated 96-well Biocoat plates (Becton Dickinson, Basel, Switzerland). The medium was carefully removed, followed by the addition of 30 [micro]L fresh medium, 10 [micro]L medium containing various concentrations of organotins, and 10 [micro]L radiolabeled cortisol. The reaction volume was 50 [micro]L, with a final cortisol concentration of 40 nM. The cells were incubated for 2 hr at 37[degrees] C under 5% C[O.sub.2]. We stopped reactions by adding an excess of unlabeled cortisone and cortisol in methanol, and separated steroids using thin-layer chromatography, followed by scintillation counting. 11[beta]-HSD2 activity in JEG-3, SW-620, and RCCD-2 cells was measured similarly by adjusting the cell density and reaction time to obtain a maximal conversion of cortisol between 15 and 25%. To ensure that the observed inhibition of 11[beta]-HSD2 activity was not due to cell death, we assessed cytotoxicity of organotin compounds parallel to the activity assay under identical conditions. The corresponding organotin compound was added to the cells, followed by incubation for 2 hr at 37[degrees] C under 5% C[O.sub.2]. Cells were washed with PBS and incubated in fresh medium containing 0.5 mg/mL 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide (MTT). After conversion of MTT, we removed the medium and added 200 [micro]L DMSO to the insoluble fraction. Conversion of MTT was kept below OD 0.9 (A570 A690)

Site-directed mutagenesis and analysis of mutant 11[beta]-HSD2 enzymes. Mutations were introduced into the C-terminally FLAG-tagged 11[beta]-HSD2 cDNA in Bluescript vector by site-directed mutagenesis according to the Quick Change mutagenesis kit (Stratagene, Amsterdam, the Netherlands) (Odermatt et al. 1999). All constructs were verified by sequencing and recloned into pcDNA3 expression vector. Wild-type and mutant enzymes were expressed in HEK-293 cells, lysates were prepared, and proteins were separated by sodium dodecyl sulfate (SDS) gel electrophoresis. Proteins were transferred to nitrocellulose, and expression levels of 11[beta]-HSD2 constructs were detected using mouse monoclonal antibody M2 raised against the FLAG epitope and visualized with a horseradish peroxidase conjugated anti-mouse antibody and enhanced chemiluminescence Western kit (Pierce, Rockford, IL, USA). After detection of 11[beta]-HSD2 constructs, nitrocellulose membranes were stripped and incubated with rabbit polyclonal anti-actin IgG (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) and horseradish peroxidase-conjugated goat anti-rabbit IgG to adjust for the amount of proteins loaded on the gel. The expression of mutant relative to wild-type enzyme was adjusted for calculation of kinetic parameters.

Three-dimensional modeling. The 3-D model of human 11[beta]-HSD2 from Arnold et al. (2003) was used with [NAD.sup.+] extracted from protein data bank file 1AHI (Tanaka et al. 1996). Human 11[beta]-HSD2 was then minimized for 100,000 iterations with Discover 3 (Accelrys Inc., San Diego, CA, USA) using the extensible and systematic force field (ESFF), with a distant dependent dielectric constant of 2, to model water in the protein.

Statistical analysis. Enzyme kinetics were analyzed by nonlinear regression using Data Analysis Toolbox (MDL Information Systems Inc., Nashville, TN, USA) assuming first-order rate kinetics. Data represent mean [+ or -] SD of at least four independent experiments.

Results

Inhibition of 11[beta]-HSD2 but not 11[beta]-HSD1 by organotins. To investigate whether organotins disrupt the control of the ratio of active to inactive glucocorticoids by inhibition of 11[beta]-HSD enzymes, we incubated lysates of HEK-293 cells expressing recombinant 11[beta]-HSD constructs with various concentrations of TBT and TPT and determined enzymatic activities. TBT and TPT did not inhibit the 11[beta]-HSD1-dependent conversion of cortisone to cortisol at concentrations up to 200 [micro]M (Table 1). In contrast the 11[beta]-HSD2-dependent conversion of cortisol to cortisone was efficiently inhibited by both compounds with [IC.sub.50] (median inhibitory concentration) values in the low micromolar range, indicating that organotins selectively abolish the 11[beta]-HSD2-dependent inactivation of glucocorticoids.

Despite that 11[beta]-HSD1 and 11[beta]-HSD2 interconvert the same substrate, they are phylogenetically relatively distant enzymes, sharing only 18% identical amino acid sequence (Baker 2004). 11[beta]-HSD2 is more closely related to 17[beta]-HSD2, with an amino acid sequence about 45% identical. Thus, we also determined the conversion of estrone to the more potent estrogen estradiol by 17[beta]-HSD1 and the reverse reaction by 17[beta]-HSD2 in the presence of various concentrations of organotins. As shown in Table 1, neither TBT nor TPT inhibited 17[beta]-HSD1. 17[beta]-HSD2 activity was inhibited by TPT at a 6-fold higher [IC.sub.50] value compared with that of 11[beta]-HSD2, indicating that organotins preferentially inhibit 11[beta]-HSD2.

Because trialkyltins are progressively dealkylated by microorganisms in the environment and in mammalian organs including brain, liver, and kidneys (Gadd 2000), and because the dialkyltins DBT and DPT are major metabolites with significant toxicity (Penninks et al. 1985; Seinen and Willems 1976; Snoeij et al. 1988), we included these two compounds in the study. Analysis of all four organotin compounds revealed comparable inhibitory properties both in lysates and intact cells expressing 11[beta]-HSD2 with [IC.sub.50] values in the low micromolar range (Table 1). In intact cells the trialkyltins were approximately 3-fold more potent than the dialkyltins, and the phenyltins were slightly more potent than the butyltins, well correlating with the hydrophobicity of these compounds. It is important to note that the inhibitory potency of organotins is comparable with that of the well-known 11[beta]-HSD inhibitor GA in intact cells.

Dithiothreitol but not glutathione protects 11[beta]-HSD2 from inhibition by organotins. To investigate the molecular mechanism of organotin-induced inhibition of 11[beta]-HSD2, we measured the effect of organotins in the presence or absence of the reducing agent dithiothreitol (DTT) (Figure 1). DTT alone did not significantly alter enzymatic activity. A concentration of 2 mM DTT, added simultaneously with 5 [micro]M of the corresponding organotin compound, restored 70-80% of 11[beta]-HSD2 activity. Upon preincubation of 11[beta]-HSD2 with organotins for 15 min, 50-60% of the enzymatic activity could be restored (not shown), indicating that most but not all of the inhibitory effect was reversible. In contrast to the dithiol DTT, the monothiol glutathione was not able to prevent organotin-dependent inhibition of 11[beta]-HSD2 (not shown).

[FIGURE 1 OMITTED]

We recently demonstrated that dithiocarbamates, another environmentally relevant class of compounds, inhibit 11[beta]-HSD2. Cofactor [NAD.sup.+] partially protected the enzyme, suggesting that covalent modification of the thiol group at [Cys.sup.90] in the cofactor-binding region may be responsible for dithiocarbamate-induced inactivation of 11[beta]-HSD2 (Atanasov et al. 2003). In contrast we found no protective effect of [NAD.sup.+] on organotin-induced enzyme inhibition (not shown), indicating an inhibitory mechanism distinct from that of dithiocarbamates.

Functional analysis of the cysteine residues of 11[beta]-HSD2. We have previously shown that substitution of [Cys.sup.90] by serine leads to abolished protein expression and enzymatic activity (Atanasov et al. 2003). Because [NAD.sup.+] did not protect 11[beta]-HSD2 from organotin-induced inhibition, we analyzed the functional relevance of the remaining eight cysteine residues by mutating them to serines. All nine cysteineto-serine mutants contained a FLAG epitope at the C-terminus, allowing the quantification of the relative protein expression (Figure 2). Five of the mutant enzymes showed significantly reduced expression. No band could be detected for mutant [Cys.sup.228]Ser and only a very weak band for [Cys.sup.90]Ser was detected. As expected, no activity could be measured using lysates from cells transfected with cDNA for either of these two mutant enzymes. Significantly reduced expression was also observed for mutants [Cys.sup.128]Ser and [Cys.sup.188]Ser. When the catalytic activity of these mutant enzymes was adjusted for their reduced expression, an apparent [V.sub.max] comparable to that of wild-type 11[beta]-HSD2 was obtained (Table 2), indicating reduced enzyme stability but intact catalytic activity. For mutant [Cys.sup.264]Ser, which also showed reduced expression, a slightly higher [V.sub.max] Was obtained. Comparison of the kinetic parameters revealed altered kinetic parameters for mutant [Cys.sup.264]Ser, with a 3-fold higher apparent [K.sub.m]. Mutation of [Cys.sup.264] to serine also resulted in a 2-fold increase in the [IC.sub.50] for TBT, suggesting a role for [Cys.sup.264] in the binding of cortisol and in the interaction with organotins with 11[beta]-HSD2.

[FIGURE 2 OMITTED]

Analysis of the mode of organotin-dependent inhibition of 11[beta]-HSD2. We next investigated the effect of preincubation of 11[beta]-HSD2 with either 1.5 [micro]M TPT or 2 [micro]M TBT for 5 or 10 min. Although there was a slight tendency toward increased inhibitory effect upon preincubation with TBT, the changes did not reach significance (Figure 3), in line with reversible inhibition. To further assess the mode of inhibition, we determined the change of apparent [K.sub.m] and [V.sub.max] in the absence or presence of TBT. The apparent [K.sub.m] increased 2- and 3-fold upon incubation with 2 [micro]M and 3 [micro]M TBT (62 [+ or -] 17 nM for the control compared with 137 [+ or -] 24 nM and 197 [+ or -] 33 nM for treated samples, respectively), whereas [V.sub.max] decreased slightly (2.14 [+ or -] 0.32 nmol x [h.sup.-1] x [mg.sup.-1] for the control compared with 1.66 [+ or -] 0.40 nmol x [h.sup.-1] x [mg.sup.-1] and 1.48 [+ or -] 0.48 nmol x [h.sup.-l] x [mg.sup.-1] for treated samples, respectively). These findings suggest a mixed-competitive mode of inhibition, with most of the inhibitory effect being reversible.

[FIGURE 3 OMITTED]

To further test this assumption, we measured the effect of diluting the enzyme-inhibitor complex (EI) (Figure 4). In case of a competitive mode of inhibition, dilution of the EI complex would lead to a decreased inhibitor concentration and reduced competition with the substrate, hence reduced enzyme inhibition. If covalent enzyme modification takes place, the dilution of the El complex would not change the proportion of modified to unmodified molecules, meaning that after the dilution of El there would be no change of the relative inhibition compared with the control. In case of TBT-induced inhibition of 11[beta]-HSD2, a 2-fold and 4-fold dilution of the El complex led to a proportional decrease of the relative inhibitory effect, indicating a transient interaction of the organotin compound with 11[beta]-HSD2.

[FIGURE 4 OMITTED]

Inhibition of 11[beta]-HSD2 in endogenous cell lines. Because organisms are exposed to various sources of organotins and these chemicals undergo dealkylation in vivo, cells in tissues are generally exposed to a mixture of organotins. Therefore, we compared the activity of 11[beta]-HSD2 in intact cells either upon incubation with DBT, TBT, DPT, or TPT alone, at concentrations 50% below their [IC.sup.50] value, or after incubation with a mixture of the four chemicals (Figure 5). Whereas each compound alone reduced 11[beta]-HSD2 activity only slightly, the mixture showed additive inhibitory effects and significantly inhibited enzymatic activity, indicating that the distinct organotins act by the same mechanism. We observed this phenomenon in transfected HEK-293 cells as well as in endogenous cell lines.

[FIGURE 5 OMITTED]

We next determined the potential of TBT to inhibit 11[beta]-HSD2 activity in cell lines derived from tissues with endogenous expression of this enzyme, for example, placenta, renal cortical collecting duct, and colon (Figure 6). In placenta-derived JEG-3 cells and in colon-derived SW-620 cells, the inhibition of 11[beta]-HSD2 by TBT was highly similar to that observed in HEK-293 cells. We observed approximately 2-fold stronger inhibition in renal cortical-collecting duct-derived RCCD-2 cells, with an [IC.sub.50] of 0.83 [+ or -] 0.23 [micro]M for TBT.

[FIGURE 6 OMITTED]

Discussion

Relatively little is known about the molecular targets of organotins despite their wide range of toxic effects and that they can be readily detected in the blood of humans. In this article we describe the organotin-dependent inhibition of 11[beta]-HSD2. Our data suggest that organotins inhibit 11[beta]-HSD2 by a mostly reversible, mixed-competitive mode of inhibition. Comparison of the kinetic parameters obtained from measurements with lysates and intact cells expressing 11[beta]-HSD2 indicates that the trialkyltins enter the cell more easily, which explains their more potent effects in intact cells. However, dialkyltins display equal or even enhanced inhibitory potency in lysates. Organotin-induced inhibition of 11[beta]-HSD2 was prevented by the dithiol DTT but not by the endogenous monothiol glutathione, which sugggests that two cysteine residues in close proximity might be involved in the mechanism of inhibition. This is in contrast to the inhibition of 11[beta]-HSD2 by dithiocarbamates, which irreversibly inhibit the enzyme, probably through covalent modification of a cysteine residue by attachment of a carbamoyl group (Atanasov et al. 2003).

The inhibition of 11[beta]-HSD2 by dithiocarbamates (Atanasov et al. 2003) and organotins seems to involve distinct cysteine residues, as addition of high concentrations of cofactor [NAD.sup.+] partially protected from dithiocarbamates but not from organotins. Site-directed mutagenesis and functional analysis revealed an essential role of [Cys.sup.128], [Cys.sup.188], [Cys.sup.228], and [Cys.sup.264] for enzyme stability and/or catalytic activity in addition to the previously described [Cys.sup.90). To begin to understand the basis for the different activities of these mutant enzymes, we applied a 3-D structural model of 11[beta]-HSD2 (Arnold et al. 2003; Atanasov et al. 2003) to investigate the structure surrounding each of these cysteines (Figure 7). [Cys.sup.48] and [Cys.sup.371] are located in hydrophobic segments thought to associate with cellular membranes in the endoplasmic reticulum. These residues are outside the 260 residue core segment comprising the catalytically active domain in homologs of 11[beta]-HSD2 and were not analyzed further. The 3-D model shows that [Cys.sup.127], [Cys.sup.128], and [Cys.sup.248] do not interact with sites on 11[beta]-HSD2 critical for binding of either [NAD.sup.+], substrate, or the catalytically active [Tyr.sup.232]. Figure 7A shows that these cysteines have few stabilizing interactions with other amino acids and are oriented to the solvent, away from the catalytic pocket.

[FIGURE 7 OMITTED]

As described previously (Atanasov et al. 2003), the thiol group of [Cys.sup.90] has several interactions with amino acids that stabilize [Glu.sup.115] and [Asp.sup.91] (Figure 7B). [Glu.sup.115] has critical hydrogen bonds with the ribose hydroxyl on [NAD.sup.+] that are important in stabilizing binding of the cofactor and in maintaining its orientation to the substrate. Thus, the loss of the stabilizing effects of [Cys.sup.90] on the orientation of [Glu.sup.115] leads to an inactive 11[beta]-HSD2.

Similarly, the thiol group on [Cys.sup.188] stabilizes several amino acids that interact with the pyrophosphate segment of [NAD.sup.+] (Figure 7C). However, like [Cys.sup.90], [Cys.sup.188] is not directly involved in interactions with the cofactor, and it appears that if the thiol group is replaced with a serine hydroxyl group, there remains sufficient stabilization of the structure near the pyrophosphate group in mutant [Cys.sup.188]Ser to retain some catalytic activity.

[Cys.sup.228] is in the loop that precedes the catalytically active [Tyr.sup.232] (Figure 7D). Evidence from solved 3-D structures of 11[beta]-HSD2 homologs, such as 17[beta]-HSD1, clearly shows that this loop stabilizes the position of the nicotinamide ring and the catalytic [Tyr.sup.232] with the steroid substrate to promote catalysis (Breton et al. 1996; Ghosh et al. 1994; Tanaka et al. 1996; Yamashita et al. 1999). The thiol on [Cys.sup.228] stabilizes the backbone oxygen in [Pro.sup.227], which is an important structural amino acid that is also in the loop that positions the catalytic tyrosine, the nicotinamide ring, and the steroid. [Cys.sup.228] also stabilizes [Glu.sup.277], which belongs to a helix in the substrate binding site. Thus, the thiol group on [Cys.sup.228] interacts with amino acids on 11[beta]-HSD2 that are important in substrate binding, which explains the loss of activity of mutant [Cys.sup.228]Ser.

Figure 7D also shows that the thiol group of [Cys.sup.264] has important interactions with [Leu.sup.284], [Ala.sup.285], and [Pro.sup.288], which are part of the helix in the C-terminal region of 11[beta]-HSD2 that likely is important in the substrate binding site, based on analyses of 17[beta]-HSD2 and other homologs (Tanaka et al. 1996) of 11[beta]-HSD2. Our experimental data indicate that the serine hydroxyl group can partially take over the function of the thiol on [Cys.sup.264]. Together, these findings suggest that [Cys.sup.228] and [Cys.sup.264] may be involved in the interactions with organotins, and interference with the function of their thiol groups may be responsible for inactivation of 11[beta]-HSD2 by organotins.

Although the chemical nature of organotin-protein interactions is not completely understood at present, it is believed that most of the properties of organotins are a result of the nature of C-Sn bonds that can be attacked by both nucleophilic and electrophilic reagents (Hoch 2001). Buck et al. recently described the interaction of organotins with the membrane protein stannin (Buck et al. 2003, 2004). A nonapeptide derived from stannin containing the sequence Cys-Trp-Cys was able to dealkylate TMT, followed by binding of the resulting DMT. Buck et al. (2003, 2004) further showed that DTT bound TMT without inducing dealkylation. Stannin efficiently dealkylated organotins with short alkyl side chains with, weak binding observed for TBT and TOT.

11[beta]-HSD2 does not possess a Cys-XaaCys motif, and [Cys.sup.127] and [Cys.sup.128] are unlikely to be the target residues for inhibition by organotins, as replacement of either of these residues did not affect the organotin-dependent inhibition of the mutant enzymes. Replacing [Cys.sup.228] by serine completely abolished enzyme activity, indicating an important functional role of this residue. Mutating [Cys.sup.264] to serine led to a well-expressed enzyme with only slightly reduced catalytic efficiency. This mutant was less sensitive to organotin-dependent inhibition.

The potency of organotins in inhibiting 11[beta]-HSD2 is equal to or greater than that reported for other enzymes involved in steroid hormone metabolism, including cytochrome P450 aromatase (Lo et al. 2003), 5[alpha]-reductases (Doering et al. 2002), and rat testis microsomal 3[beta]-HSD (McVey and Cooke 2003). As mentioned earlier, organotins represent ubiquitous contaminants of the water ecosystem. Although most often present at lower nanomolar concentrations, organotins accumulate in aquatic organisms, with up to 70,000 times higher organotin concentrations in plankton and other organisms than in sea water (Takahashi et al. 1999). As little as 200 g of contaminated shellfish could be sufficient to reach the toxic daily intake of TBT (0.25 [micro]g/kg body weight) (McVey and Cooke 2003). Another major source of organotins for humans is the drinking water in locations where PVC water pipes are used. A study of organotin levels in Canadian drinking water distributed through PVC pipes in 1996 revealed total concentrations of different organotins < 50 ng Sn/L, although in some occasions values > 250 ng Sn/L were detected (Sadiki and Williams 1999). Although the [IC.sub.50] values of organotins for 11[beta]-HSD2 in the present study were in the high nanomolar and low micromolar range, the accumulation of organotins in specific organs and tissues including brain, liver, and kidneys may lead to high local concentrations, as has been found in aquatic organisms. In addition Chicano et al. (2001) provided evidence that organotins are located in the upper part of the phospholipid palisade near the lipid-water interface and affect the degree of hydration of the phospholipid carbonyl moiety. Thus, intracellular organotin concentrations might be highest at the surface of membranes, and 11[beta]-HSD2 might be exposed to concentrations much higher than those in the solution.

The increased glucocorticoid-mediated effects due to the inhibition of 11[beta]-HSD2 are expected to disturb several essential physiologic processes. The effect of TBT and its major metabolite DBT to suppress T-cell-dependent immune functions by causing thymus atrophy has been extensively studied (Krajnc et al. 1984; Seinen et al. 1977a, 1977b; Snoeij et al. 1988; Tryphonas et al. 2004; Vos et al. 1984a, 1990). A single dose of TBT, DBT, or DOT induced dose-dependent reductions in the weight of the thymus, spleen, and lymph node (Seinen et al. 1977b). The effect of TBT was less pronounced and slightly delayed compared with DBT, indicating that in vivo TBT is metabolized to the more toxic DBT (Snoeij et al. 1988). The thymotoxic effects of organotins are completely reversible (Seinen et al. 1977b). A selective inhibition of the proliferation of immature [CD4.sup.-]/[CD8.sup.+] thymocytes by organotins seems to be responsible for the observed depletion of [CD4.sup.+]/[CD8.sup.+] thymocytes, which show a rapid turnover.

11[beta]-HSD enzymes play a pivotal role in regulating proliferation and differentiation in various tissues, 11[beta]-HSD1 generates active glucocorticoids and promotes differentiation, and 11[beta]-HSD2 inactivates glucocorticoids, thereby promoting proliferation. 11[beta]-HSD1 and 11[beta]-HSD2 were both expressed in whole mouse thymus (Moore et al. 2000; Speirs et al. 2004), although the exact subtype-specific expression pattern of 11[beta]-HSD enzymes remains to be determined. In the acute stress response, the high level of glucocorticoids induces thymus involution (Schuurman et al. 1992). Organotin-dependent inhibition of 11[beta]-HSD2 may cause antiproliferative effects on immature thymocytes by increasing locally the ratio of active to inactive glucocorticoids or, alternatively, by increasing systemic glucocorticoid levels. Both organotin-induced inhibition of 11[beta]-HSD2 and thymotoxicity are reversible.

Experiments in mice showed maximal thymocyte apoptosis 8 hr after glucocorticoid administration, followed by full recovery after 18 hr (Ishii et al. 1997). There was a significant depletion of [CD4.sup.+]/[CD8.sup.+] thymocytes. A delayed, dose-dependent apoptosis of thymocytes, reaching maximal effect after 24 hr, was observed when mice were treated with a single dose of the 11[beta]-HSD inhibitor GA (Horigome et al. 1999). Thymocyte apoptosis was induced dose-dependently by corticosterone in vitro. GA alone did not induce apoptosis in vitro, suggesting that elevated corticosterone levels due to inhibition of 11[beta]-HSD2 may cause the apoptosis. Similarly, organotin-dependent inhibition of 11[beta]-HSD2 and subsequent locally enhanced glucocorticoid levels may contribute to the immunotoxicity of these compounds. However, thymus atrophy was also observed in DBT- and TBT-treated adrenalectomized rats (Seinen and Willems 1976; Snoeij et al. 1985), and no extensive cell destruction was observed in DBT- and TBT-treated rats compared with rats treated with very high glucocorticoid concentrations (La Pushin and de Harven 1971). These findings suggest that the immunotoxic effects of organotins are caused by a glucocorticoid-dependent and a glucocorticoid-independent mechanism.

The glucocorticoid-dependent effects caused by organotins may be most critical during pregnancy, where fetal development is highly sensitive to glucocorticoids (Seckl et al. 2000). The lower birth weight and decreased weight gain in the offspring after exposure of organotins during pregnancy is a phenotype also observed as a result of exposure to excessive levels of glucocorticoids. Enhanced glucocorticoid action due to inhibition of 11[beta]-HSD2, which in the placenta protects the fetus from high maternal levels, or due to treatment with synthetic glucocorticoids that cannot be inactivated by 11[beta]-HSD2, such as dexamethasone (Rebuffat et al. 2004), have been associated with reduced birth weight and irreversible changes in the cardiovascular system with complications in later life (Seckl et al. 2000). Thus, inhibition of 11[beta]-HSD2 may be one reason that offspring from pregnant rats treated with organotins show significantly reduced birth weight (Adeeko et al. 2003; Cooke et al. 2004; Crofton et al. 1989).

Conclusions

This work demonstrates the disruption of the 11[beta]-HSD2-dependent inactivation of glucocorticoids by organotins. Various organotin compounds inhibit 11[beta]-HSD2, mainly by a reversible mode of inhibition, and show additive effects. Endogenous glutathione cannot prevent the organotin-induced inhibition of 11[beta]-HSD2, which explains the comparable inhibitory kinetics obtained in experiments with cell lysates and in intact cells. The results suggest that enhanced glucocorticoid concentrations due to disruption of 11[beta]-HSD2 function may contribute to the observed organotin-dependent toxicity in glucocorticoid sensitive tissues such as thymus and placenta. Clearly, additional experiments in vivo must be performed to elucidate the relevance of organotin-dependent interference with glucocorticoid action and its pathophysiologic consequences.

Received 15 April 2005; accepted 14 July 2005.

REFERENCES

Adeeko A, Li D, Forsyth DS, Casey V, Cooke GM, Barthelemy J, et al. 2003. Effects of in utero tributyltin chloride exposure in the rat on pregnancy outcome. Toxicol Sci 74:407-415.

Arnold P, Tam S, Yan L, Baker ME, Frey F J, Odermatt A. 2003. Glutamate-115 renders specificity of human 11[beta]-hydroxysteroid dehydrogenase type 2 for the cofactor NAD(+). Mol Cell Endocrinol 201:177-187.

Atanasov AG, Nashev LG, Schweizer RA, Frick C, Odermatt A. 2004. Hexose-6-phosphate dehydrogenase determines the reaction direction of 11[beta]-hydroxysteroid dehydrogenase type 1 as an oxoreductase. FEBS Lett 571:129-133.

Atanasov AG, Tam S, Rocken JM, Baker ME, Odermatt A. 2083. Inhibition of 11[beta]-hydrexysteroid dehydrogenase type 2 by dithiocarbamates. Biochem Biophys Res Commun 308:257-262.

Baker ME. 2004. Evolutionary analysis of 11[beta]-hydroxysteroid dehydrogenase-type 1, -type 2, -type 3 and 17[beta]-hydroxysteroid dehydrogenase-type 2 in fish. FEBS Lett 574:167-170.

Benediktsson R, Lindsay RS, Noble J, Seckl JR, Edwards CR. 1993. Glucocorticoid exposure in utero: new model for adult hypertension. Lancet 341:339-341.

Bhosle NB, Garg A, Jadhav S, Harjee R, Sawant SS, Venkat K, et al. 2004. Butyltins in water, biofilm, animals and sediments of the west coast of India. Chemosphere 57:897-907.

Breton R, Housset D, Mazza C, Fontecilla-Camps JC. 1996. The structure of a complex of human 17[beta]-hydroxysteroid dehydrogenase with estradiol and NABP+ identifies two principal targets for the design of inhibitors. Structure 4:905-915.

Buck BA, Mascioni A, Cramer CJ, Veglia G. 2004. Interactions of alkyltin salts with biological dithiols: dealkylation and induction of a regular [beta]-turn structure in peptides. J Am Chem Soc 126:14400-14410.

Buck B, Mascioni A, Que L Jr, Veglia G. 2003. Dealkylation of organotin compounds by biological dithiols: toward the chemistry of organotin toxicity. J Am Chem Soc 125: 13316-13317.

Chicano JJ, Ortiz A, Teruel JA, Aranda FJ. 2001. Organotin compounds alter the physical organization of phesphatidylcholine membranes. Biochim Biophys Acta 1510:330-341.

Coelho MR, Bebianno MJ, Langston WJ. 2002. Routes of TBT uptake in the clam Ruditapes decussatus. II: Food as vector of TBT uptake. Mar Environ Res 54:193-207.

Cooke GM, Tryphonas H, Pulido O, Caldwell D, Bondy GS, Forsyth D. 2004. Oral (gavage), in utero and postnatal exposure of Sprague-Bawley rats to low doses of tributyltin chloride. Part 1: Toxicology, histopathology and clinical chemistry. Food Chem Toxicol 42:211-220.

Croffon KM, Dean KF, Boncek VM, Rosen MB, Sheets LP, Chernoff N, et al. 1989. Prenatal or postnatal exposure to bis(tri-n-butyltin)oxide in the rat: postnatal evaluation of teratelogy and behavior. Toxicol Appl Pharmacol 97:113-123.

Djelidi S, Beggah A, Courtois-Coutry N, Fay M, Cluzeaud F, Viengchareun S, et al. 2001. Basolateral translocation by vasopressin of the aldosterone-induced pool of latent Na-K-ATPases is accompanied by [[alpha].sub.1] subunit dephosphorylation: study in a new aldosterone-sensitive rat cortical collecting duct cell line. J Am Soc Nephrol 12:1805-1818.

Doering DD, Steckelbroeck S, Boering T, Klingmuller D. 2002. Effects of butyltins on human 5[alpha]-reductase type 1 and type 2 activity. Steroids 67:859-887.

Drake A J, Walker BR, Seckl JR. 2005. Intergenerational consequences of fetal programming by in utero exposure to glucocorticoids in rats. Am J Physiol Regul Integr Comp Physiol 288: R34-R38.

Evans S, Nicholson GJ. 2000. The use of imposex to assess tributyltin contamination in coastal waters and open seas. Sci Total Environ 258:73-80.

Fait A, Ferioli A, Barbieri F. 1994. Organotin compounds. Toxicology 91:77-82.

Fent K. 1996. Ecotoxicology of organotin compounds. Crit Rev Toxicol 26:1-117.

Fent K. 2003. Ecotoxicologicar problems associated with contaminated sites. Toxicol Left 140-141:353-365.

Gadd GM. 2000. Microbial interactions with tributyltin compounds: detoxification, accumulation, and environmental fate. Sci Total Environ 258:119-127.

Ghosh D, Wawrzak Z, Weeks CM, Duax WL, Erman M. 1994. The refined three-dimensional structure of 3[alpha],20[beta]-hydroxysteroid dehydrogenase and possible roles of the residues conserved in short-chain dehydrogenases. Structure 2:629-640.

Hasan Z, Zimmer L, Woolley D. 1984. Time course of the effects of trimethyltin on limbic evoked potentials and distribution of tin in blood and brain in the rat. Neurotoxicology 5:217-244.

Hoch M. 2001. Organotin compounds in the environment--an overview. Appl Geochem 16:719-743.

Horigome H, Horigome A, Homma M, Birano T, Oka K. 1999. Glycyrrhetinic acid-induced apoptosis in thymocytes: impact of 11[beta]-hydroxysteroid dehydrogenase inhibition. Am J Physiol 277:E624-E630.

Ishii T, Nakamura M, Yagi H, Soga H, Kayaba S, Gotoh T, et al. 1997. Glucocorticoid-induced thymocyte death in the murine thymus: the effect at later stages. Arch Histol Cytol 60:65-78.

Kozak M. 1989. Context effects and inefficient initiation at non-AUG codons in eucaryotic cell-free translation systems. Mol Cell Biol 9:5073-5080.

Krajnc El, Wester PW, Loeber JG, van Leeuwen FX, Vos J6, Vaessen HA, et al. 1984. Toxicity of bis(tri-n-butyltin)oxide in the rat. I: Short-term effects on general parameters and on the endocrine and lymphoid systems. Toxicol Appl Pharmacol 75:363-386.

La Pushin RW, de Harven E. 1971. A study of glucocorticosteroid-induced pyknosis in the thymus and lymph node of the adrenalectomized rat. J Cell Biol 50:583-597.

Lindsay RS, Lindsay RM, Edwards CR, Seckl JR. 1996a. Inhibition of 11-[beta]-hydroxysteroid dehydrogenase in pregnant rats and the programming of blood pressure in the offspring. Hypertension 27:1200-1204.

Lindsay RS, Lindsay RM, Waddell B J, Seckl JR. 1996b. Prenatal glucocorticoid exposure leads to offspring hyperglycaemia in the rat: studies with the 11[beta]-hydroxysteroid dehydrogenase inhibitor carbenoxolone. Diabetolegia 39:1299-1305.

Liu JY, Jiang GB. 2002. Survey on the presence of butyltin compounds in chinese alcoholic beverages, determined by using headspace solid-phase microextraction coupled with gas chromatography-flame photometric detection. J Agric Food Chem 50:6683-6687.

Lo S, Allera A, Albers P, Heimbrecht J, Jantzen E, Klingmuller D, et al. 2003. Dithioerythritol (DTE) prevents inhibitory effects of triphenyltin (TPT) on the key enzymes of the human sex steroid hormone metabolism. J Steroid Biochem Mol Biol 84:569-576,

McVey M J, Cooke GM. 2003. Inhibition of rat testis microsomal 3[beta]-hydroxysteroid dehydrogenase activity by tributyltin. J Steroid Biochem Mol Biol 85: 99-105.

Moore XL, Hoong I, Cole TJ. 2000. Expression of the 11[beta]-hydroxysteroid dehydrogenase 2 gene in the mouse. Kidney Int 57:1307-1312.

Odermatt A. 2004. Corticosteroid-dependent hypertension: environmental influences. Swiss Med Wkly 134:4-13.

Odermatt A, Arnold P, Stauffer A, Frey BM, Frey FJ. 1999. The N-terminal anchor sequences of 11[beta]-hydroxysteroid dehydrogenases determine their orientation in the endoplasmic reticulum membrane. J Biol Chem 274: 28762-28770.

Penninks A, Kuper F, Spit B J, Seinen W. 1985. On the mechanism of dialkyltin-induced thymus involution. Immunopharmacology 10:1-10.

Rebuffat AG, Tam S, Nawrocki AR, Baker ME, Frey BM, Frey F J, et al. 2004. The 11-ketosteroid 11-ketodexamethasone is a glucocortieoid receptor agonist. Mol Cell Endocrinol 214:27-37.

Sadiki Al, Williams DT. 1999. A study on organotin levels in Canadian drinking water distributed through PVC pipes. Chemosphere 38:1541-1548.

Schuurman H J, Van Loveren H, Rozing J, Vos JG. 1982. Chemicals trophic for the thymus: risk for immunodeficiency and autoimmunity. Int J Immunopharmacol 14:369-375.

Schweizer RA, Atanasov AG, Frey BM, Odermatt A. 2003. A rapid screening assay for inhibitors of 11[beta]-hydroxysteroid dehydrogenases (11[beta]-HSD): flavanone selectively inhibits 11[beta]-HSD1 reductase activity. Mol Cell Endocrinol 212:41-49.

Seckl JR, Cleasby M, Nyirenda MJ. 2000. Glucocorticoids, 11[beta]hydroxysteroid dehydrogenase, and fetal programming. Kidney Int 57:1412-1417.

Seinen W, Vos JG, van Krieken R, Penninks A, Brands R, Hooykaas H. 1977a. Toxicity of organetin compounds. Ill: Suppression of thymus-dependent immunity in rats by di-n-butyltindichloride and di-n-octyltindichloride. Toxicol Appl Pharmacol 42:213-224.

Seinen W, Vos JG, van Spanje I, Snoek M, Brands R, Hooykaas H. 1977b. Toxicity of organotin compounds. II: Comparative in vivo and in vitro studies with various organotin and organolead compounds in different animal species with special emphasis on lymphocyte cytotoxicity. Toxicol Appl Pharmacol 42:197-212.

Seinen W, Willems Ml. 1976. Toxicity of organotin compounds. I: Atrophy of thymus and thymus-dependent lymphoid tissue in rats fed di-n-octyltindichloride. Toxicol Appl Pharmacol 35:63-75.

Snoeij NJ, Penninks AH, Seinen W. 1988. Dibutyltin and tributyltin compounds induce thymus atrophy in rats due to a selective action on thymic lymphoblasts. Int J Immunopharmacol 10:891-898.

Snoeij NJ, van Iersel AA, Penninks AH, Seinen W. 1985. Toxicity of triorganotin compounds: comparative in vivo studies with a series of trialkyltin compounds and triphenyltin chloride in male rats. Toxicol Appl Pharmacol 81: 274-286.

Speirs HJ, Seckl JR, Brown RW. 2004. Ontogeny of glucocorticoid receptor and 11[beta]-hydroxysteroid dehydrogenase type-1 gene expression identifies potential critical periods of glucocorticold susceptibility during development. J Endocrinol 181:105-116.

Stewart PM, Krozowski ZS. 1999. 11[beta]-Hydroxysteroid dehydrogenase. Vitam Horm 57:249-324.

Stewart PM, Regerson FM, Mason JI. 1995. Type 2 11[beta],-hydroxysteroid dehydrogenase messenger ribonucleic acid and activity in human placenta and fetal membranes: its relationship to birth weight and putative role in fetal adrenal stereidogenesis. J Clin Endocrinol Metab 80:885-890.

Takahashi S, Tanabe S, Takeuchi II, Miyazaki N. 1999. Distribution and specific bioaccumulation of butyltin compounds in a marine ecosystem. Arch Environ Contain Toxicol 37:50-61.

Tanaka N, Nonaka T, Tanabe T, Yoshimoto T, Tsuru D, Mitsui Y. 1996. Crystal structures of the binary and ternary complexes of 7[alpha]-hydroxysteroid dehydrogenase from Escherichia coil Biochemistry 35:7715-7730.

Tryphonas H, Cooke G, Caldwell D, Bondy G, Parenteau M, Hayward S, et al. 2004. Oral (gavage), in utero and post-natal exposure of Sprague-Dawley rats to low doses of tributyltin chloride. Part II: Effects on the immune system. Food Chem Toxicol 42:221-235.

Vos JG, de Klerk A, Krajnc El, Kruizinga W, van Ommen B, Rozing J. 1984a. Toxicity of bis(tri-n-butyltin)oxide in the rat. II: Suppression of thymus-dependent immune responses and of parameters of nonspecific resistance after short-term exposure. Toxicol Appl Pharmacol 75:387-408.

Vos JG, De Klerk A, Krajnc El, Van Loveren H, Rozing J. 1980. Immunotoxieity of bis(tri-n-butyltin)oxide in the rat: effects on thymus-dependent immunity and on nonspecific resistance following long-term exposure in young versus aged rats. Toxicol Appl Pharmacol 105: 144-155.

Vos JG, van Logten M J, Kreeftenberg JG, Kruizinga W. 1984b. Effect of triphenyltin hydroxide on the immune system of the rat. Toxicology 29:325-336.

Yamashita A, Kato H, Wakatsuki S, Tomizaki T, Nakatsu T, Nakajima K, et al. 1999. Structure of tropinone reductase-II complexed with NADP+ and pseudotropine at 1.9 A resolution: implication for stereo specific substrate binding and catalysis. Biochemistry 38:7630-7637.

Atanas G. Atanasov, (1) Lyubomir G. Nashev, (1) Steven Tam, (2) Michael E. Baker, (2) and Alex Odermatt (1)

(1) Department of Nephrology and Hypertension, Department of Clinical Research, University of Berne, Berne, Switzerland; (2) Department of Medicine, University of California, San Diego, La Jolla, California

Address correspondence to A. Odermatt, Department of Nephrology and Hypertension, Department of Clinical Research, University of Berne, Freiburgstrasse 15, 3010 Berne, Switzerland. Telephone: 41-31-6329438. Fax: 41-31-632-9444. E-mail: alex.odermatt@dkf.unibe.ch

We thank N. Farman (INSERM, Faculte X-Bichat, Paris, France) for providing the RCCD-2 cells, S. Andersson (University of Texas Southwestern Medical Center, Dallas, Texas) for the gift of 17[beta]-HSD1 and 17[beta]-HSD2 plasmids, and J. Vos (National Institute of Public Health and the Environment, Biltoven, the Netherlands) for helpful discussion. We also thank H. Jamin for excellent technical support.

A.O. is a Cloetta Research Fellow supported by grants from the Swiss National Science Foundation (3100A0-100060 and NRP50 "Endocrine Disruptors" 4050-066575) and the Swiss Cancer League (OCS-01402-08-2003). M.E.B. and S.T. were supported by National Institute of Health grants DK41841 and HL004791.

The authors declare they have no competing financial interests.

COPYRIGHT 2005 National Institute of Environmental Health Sciences
COPYRIGHT 2005 Gale Group

Return to Estrone
Home Contact Resources Exchange Links ebay