* BioDetection Systems B.V., Badhuisweg 3, 1031 CM Amsterdam, The Netherlands, and Institute for Environmental Studies, Vrije Universiteit Amsterdam, De Boelelaan 1115, 1081 HV Amsterdam, The Netherlands
1 To whom correspondence should be addressed. Fax: +31-204350757. E-mail: edwin.sonneveld{at}bds.nl
Received August 8, 2004; accepted October 4, 2004
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Key Words: androgen; estrogen; receptor; CALUX; luciferase; bioassay.
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Because of the many possible applications of steroid bioassays, we were interested in developing a panel of assays using the same cellular background, in which the activity of all major classes of steroid hormones can be determined specifically and sensitively. In particular we were interested in generating a selective and sensitive bioassay for androgens, in response to the recent interest in environmental androgens and anti-androgens (ICCVAM, 2003; Kelce et al., 1995
), together with the paucity of good assay systems for this class of hormones.
Androgens are a major class of steroid hormones that have critical roles in the development and maintenance of the male reproductive system and other physiological targets, predominantly in males. Through their anabolic effects, androgens are used to promote muscle strength in athletes and meat quantity in farm animals (Evans, 2004; Meyer, 2001
). It has also been found that environmental chemicals can interfere with androgen action, thereby possibly contributing to disruption of the endocrine system in wildlife and humans (Andersen et al., 2002
; Kelce and Wilson, 1997
). Therefore, identification of androgen active compounds is important in a variety of fields, ranging from pharmacological and clinical screening, food and feed manufacturing, to toxicological monitoring and risk assessment. Traditionally, monitoring strategies focus on two extremes: (1) sophisticated, detailed chemical analysis and (2) determination of biological effects using whole-animal assays and epidemiology. With these two methods a correlation can be made between in vivo or environmental levels of a chemical and the effect seen in organisms (exposure and effect determinations). Rapid advances in molecular biology and biotechnology have allowed identification of mechanisms of action of toxicants and facilitated development of simple assays based on these signaling mechanisms. These assays have the potential to be used as monitoring tools for chemical contaminants interfering with these signaling mechanisms, but also have the potential to replace, in part, animal experimentation for effect determination by offering prescreens to identify chemicals that impact major toxicological endpoints. In the case of androgens, the main mode of signaling is well established.
The effects of androgens in target cells are mediated by the androgen receptor (AR), a member of the nuclear hormone receptor superfamily that also includes receptors for other steroid hormones like progestins and glucocorticoids, retinoids, and thyroid hormones (Mangelsdorf et al., 1995; McKenna and O'Malley, 2002
). AR is a ligand-dependent transcription factor that regulates specific gene expression by binding to specific hormone response elements (HREs) within the regulatory DNA sequences of androgen-responsive genes (Claessens et al., 2001
). The enhancer region of the mouse mammary tumor viral long terminal repeat (MMTV-LTR) promoter is the most widely used enhancer to study AR function, although it was originally isolated as a progesterone and glucocorticoid-responsive enhancer (Di Croce et al., 1999
). This can be explained by the fact that the four inverted repeats of the core sequence 5'-TGTTCT-3' within the MMTV-LTR enhancer are recognized by: AR, glucocorticoid receptor (GR), progesterone receptor (PR), and mineralocorticoid receptor (MR; Glass, 1994
), now classified as the members of the 3C group within the nuclear receptor family (Nuclear Receptors Nomenclature Committee; 1999
). The MMTV promoter also contains several enhancer regions that can be addressed by transcription factors that may respond to other hormonal and cellular stimuli, thereby modulating steroid responses (Aurrekoetxea-Hernandez and Buetti, 2004
; Uchiumi et al., 1998
).
Several stable reporter gene assays have been described for androgens. However, these systems still have several drawbacks, since they either have a low responsiveness, use slowly growing prostatic cell lines, or are not selective in their response because of expression of other nuclear hormone receptors of the C3 class, activating the transfected reporter gene through non-AR-mediated mechanisms (Blankvoort et al., 2001; de Gooyer et al., 2003
; Paris et al., 2002a
; Terouanne et al., 2000
; Wilson et al., 2002
). We decided to generate a new androgen reporter cell line that combines high specificity, sensitivity, and ease of handling. To attain this we selected a cell line, the human bone cell line U2-OS, in which the stably introduced human androgen receptor was highly active, while expression of other C3 class receptors is insignificant. In this line we cotransfected a highly specific reporter construct, containing three HREs and a minimal promoter linked to luciferase, and selected a stable highly responsive clone. The AR CALUX cell line combines rapid growth and levels of high specificity and inducibility so far unreported. We studied its basal response characteristics, as well as its potential to serve in a variety of applications. The AR CALUX cell line is a member of a panel of reporter cell lines with the same cellular background (the U2-OS cell line), allowing efficient and convenient measurement of not only androgen-, but also estrogen-, progesterone-, and glucocorticoid-receptor interacting compounds (Quaedackers et al., 2001
; Sonneveld et al., manuscripts in preparation). Besides describing the characteristics and applications of the AR CALUX cell line, additional data are provided for the ER
CALUX cell line as a complimentary bioassay in the group of CALUX reporter cell lines.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Sera. Fetal calf serum was obtained from Invitrogen (Breda, The Netherlands). A pooled human serum batch was a gift from B. Hendriks-Stegeman (University Medical Centre, Utrecht, The Netherlands). In short, blood from 15 healthy adult volunteers (male/female ratio 8:7) was collected in silica-coated tubes (Capiject, Terumo Medical Corp.). After centrifugation serum was removed, all collected sera were pooled and stored at 20°C.
DNA constructs. A blunt-ended 3050 bp SalI fragment from pSV0-hAR (obtained from A. Brinkmann, Rotterdam, The Netherlands) containing the full-length human androgen receptor (AR) (Brinkmann et al., 1989) was inserted into the blunt-ended XhoI fragment from pSG5-neo (Sonneveld et al., 1998
) containing the neomycin resistance gene, resulting in the expression plasmid pSG5-neo-hAR. An 1800 bp EcoRI fragment from pSG5-hER
(HEGO) (obtained from P. Chambon, Strassbourg, France) containing the full-length human estrogen receptor alpha (Green et al., 1986
) was inserted in the EcoRI fragment from pSG5-neo, resulting in the expression plasmid pSG5-neo-hER
. The reporter construct pMMTVluc was described earlier (Hartig et al., 2002
). The reporter construct 3x HRE-TATA-Luc was constructed as follows: three tandem repeats of ARE oligos AAGCTTAGAACAGTTTGTAACGAGCTCGTTACAAACTGTTCTAGCTCGTTACAAACTGTTCTAAGCTCAAGCTT (Schule et al., 1988
) upstream of the minimal adenovirus E1B TATA promoter sequence (GGGTATATAAT) were inserted in the multiple cloning site of the promoter less luciferase reporter construct pLuc (Folkers et al., 1995
). The reporter construct 3x ERE-TATA-Luc was described earlier (Legler et al., 1999
).
Cell culture. The human osteoblastic osteosarcoma cell line U2-OS (ATCC) was cultured in a 1:1 mixture of Dulbecco's modified Eagle's medium and Ham's F12 medium (DF, Gibco) supplemented with 7.5% fetal calf serum. AR and ER CALUX cells were cultured in DF medium supplemented with 7.5% FCS and 200 µg/ml G418.
Transient transfections. For transient transfections, cells were plated in 24-well tissue culture plates. After culturing for 1 day, cells were transfected with 1 µg reporter plasmid (3x ERE-TATA-Luc, pMMTVluc, or 3x HRE-TATA-Luc), 200 ng SV2-lacZ, and 200 ng expression plasmid (pSG5-neo-hER, pSG5-neo-hPR, pSG5-neo-hGR, or pSG5-neo-hAR) or empty vector DNA (pSG5-neo), using the calcium phosphate coprecipitation method. Luciferase activity was corrected for transfection efficiency by measuring LacZ expression as a result of SV2-lacZ co-transfection (Kalkhoven et al., 1994
).
Establishment of stable AR and ER CALUX cell lines. U2-OS cells were transfected with 3x HRE-TATA-Luc and pSG5-neo-hAR, using calcium phosphate precipitation to generate AR CALUX cells. G418-resistant clones were tested for their response to dihydrotestosterone (DHT). Eight clones showed consequent high response. One of these (clone 568) responding to the lowest concentration of DHT (10 pM) was selected for further investigation. ER
CALUX cells (Quaedackers et al., 2001
) transfected with 3x ERE-TATA-Luc and pSG5-neo-hER
were regenerated in our laboratories, since the original clones showed bell-shaped dose-response curves and relatively high backgrounds, making them less suitable for routine applications (data not shown).
AR and ER CALUX bioassays. AR and ER
CALUX cells were plated in 96-well plates (6000 cells/well) with phenol red-free DF medium supplemented with 5% dextran-coated charcoal-stripped FCS (DCC-FCS; van der Burg et al., 1988
) at a volume of 200 µl per well. Two days later, the medium was refreshed, and cells were incubated with human or fetal serum (010% [v/v]) or the compounds to be tested (dissolved in ethanol or DMSO) in triplicate at a 1:1000 dilution. In case of serum incubation, final serum concentration was 10% (v/v), and lower percentages of the tested sera were supplemented with DCC-FCS. After 24 h the medium was removed, cells were lysed in 30 µl Triton-lysis buffer and measured for luciferase activity using a luminometer (Lucy2; Anthos Labtec Instruments, Wals, Austria) for 0.1 min/well.
Western blotting. Whole-cell extracts were prepared as described previously (Sonneveld et al., 1998). 20 µg of protein was run on an 8% (w/v) SDSpolyacrylamide gel and transferred electrophoretically to nitrocellulose sheets. Membranes were treated with blocking buffer containing 4% (w/v) nonfat powdered milk in TBST (10 mM TrisHCl pH 8.0, 150 mM NaCl, 0.2% (v/v) Tween-20) and then incubated for 2 h with anti-hAR mouse monoclonal antibody Ab-1 (clone AR441) (NeoMarkers, Fremont, CA), diluted at 1:200 in TBST buffer. After washing with TBST, the membranes were immunostained using the ECL Western blotting system (Amersham).
Immunofluorescence. Cells were grown on coverslips and fixed on ice for 15 min with 3.6% (v/v) formaldehyde in ethanol. Subsequently, the cells were washed three times with phosphate buffered saline (PBS), permeabilized with 0.1% Triton X-100/PBS, incubated with 1% (w/v) BSA/PBS, and washed three times with PBS. Cells were incubated with anti-hAR mouse monoclonal antibody Ab-1 (1:80) in 10% (v/v) normal goat serum/PBS for 1 h, washed three times with PBS, incubated with GAM-Cy3 (second) antibody (1:250) in 10% (v/v) normal goat serum/PBS for 1 h, washed three times with PBS, and mounted in Moviol.
Data analysis. Luciferase activity per well was measured as relative light units (RLUs). Fold induction was calculated by dividing the mean value of light units from exposed and nonexposed (solvent control) wells. Luciferase induction as a percentage of maximal DHT (AR CALUX) or E2 (ER CALUX) activity was calculated by setting the highest fold induction of DHT (AR CALUX) or E2 (ER
CALUX) at 100%. When assessing for anti-androgenic effects, the fold induction at the EC50 concentration of DHT was set at 100%. Data are represented as mean values ± SEM from at least three independent experiments, with each experimental point performed in triplicate. Dose-response curves were fitted using the sigmoidal fit (y = a0 + a1/(1 + exp((x a2)/a3)) in GraphPad Prism (version 4.00 for Windows, GraphPad Software, San Diego, CA), which determines the fitting coefficients by an iterative process minimizing the c2 merit function (least squares criterion). The EC50 and 100-times EC50 values were calculated by determining the concentration by which 50 or 100% of maximum activity was reached using the sigmoidal fit equation. The relative transactivation activity (RTA) of each compound tested was calculated as the ratio of maximal luciferase reporter gene induction values of each compound and the maximal luciferase reporter gene induction value of reference compound DHT (AR CALUX) or E2 (ER
CALUX). The transactivation activity of DHT or E2 was arbitrarily set at 100.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
|
|
|
|
|
Typical dose-response curves for several natural as well as synthetic estrogens are shown in Figure 5A. The ER CALUX cells showed high sensitivity toward all estrogens tested, with the following range of potencies (EC50 values): EE2 (8.5 pM), E2 (16 pM), DES (37 pM), estriol (120 pM), estrone (1.0 nM), and 17-alpha-estradiol (1.4 nM) (Table 2). Furthermore, the ER
CALUX cells showed high selectivity toward estrogens, since representative steroids for other hormone receptors (testosterone, progesterone, and dexamethasone) showed no substantial agonistic response (Table 2). Antagonists like raloxifen (IC50 = 0.1 nM), hydroxytamoxifen (IC50 = 0.3 nM), tamoxifen (IC50 = 55 nM), and ICI 164.384 (IC50 = 0.5 nM) repressed E2-induced reporter gene activity (Table 2) consistent with the known anti-estrogenic nature of these compounds.
|
Determination of Estrogens and Androgens in Serum Using AR and ER CALUX Cells
In addition to testing pure compounds (steroids as well as environmentally relevant compounds with endocrine disrupting potency) for estrogenic and androgenic activity, the need to determine steroid bioactivity status in a wide range of pediatric as well as adult clinical conditions is indicated. As a potential clinical application, human serum was applied directly to the AR and ER CALUX bioassays (Fig. 6). Increasing amounts of human serum resulted in increasing luciferase activity (Fig. 6A) in both AR and ER
CALUX cell types, indeed showing the presence of androgenic as well as estrogenic compounds in human serum comparable with plasma levels found in humans, as shown recently by other ER and AR bioassays (Paris et al., 2002b
,c
). The results show that these CALUX bioassays can potentially be used in a clinical setting, thereby potentially having the advantage of demanding only very small serum volumes (maximally 30 µl), making them applicable for pediatric purposes as well. In addition to human serum we also tested fetal calf serum for estrogenic and androgenic activities. As shown in Figure 6B, FCS showed estrogenic activity, but no AR-activating compounds.
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
To generate an androgen reporter line superior to ones currently available, we chose not to use yeast cells, but rather mammalian cells with an origin close to the organism of main concern in the field of endocrine disruption (i.e., fish and mammals, including humans). Yeast-based reporter cells, although convenient in their use (Sohoni and Sumpter, 1998) can have notably different quantitative and qualitative response to hormonally active substances, mainly due to poor transport across the yeast cell membrane, and are therefore not recommended as screening models for endocrine disruptors (ICCVAM, 2003
). Our objective was therefore to construct a mammalian, preferably human, reporter cell line with characteristics superior to the ones available. To avoid interference of signal transduction pathways other than AR-mediated signals, we choose to use a minimal AR-responsive promoter element coupled to a very minimal promoter containing a TATA box only. This approach has been shown to be successful in generation of both in vitro (Legler et al., 1999
; Lemmen et al., 2002
) and in vivo (Legler et al., 2000
; Lemmen et al., 2004
) models for selective measurement of estrogen effects. We show here that this approach can also be successfully used to generate a highly selective androgen reporter cell line in U2-OS cells, the AR CALUX cell line.
Previously, the full length MMTV promoter has been used to generate a number of androgen-responsive reporter cell lines. Although this promoter is quite selective to AR, PR, and GR, it also contains a number of regulatory sites that can be targeted by different agents other than steroids (Ouatas et al., 2002; Spangenberg et al., 1998
; Uchiumi et al., 1998
). MDA-kb2 is a derivative of a human breast cancer cell line named MDA-MB-453, containing such a stably integrated MMTV-luciferase reporter (Wilson et al., 2002
). In addition to responding to androgens, this cell line responds very strongly to glucocorticoids acting through the GR that is present endogenously, making it unsuitable as a selective screening tool. Much better androgen specificity was obtained by stable transfection of human prostatic PC-3 cells with hAR and the MMTV-luciferase reporter, named PALM cells (Terouanne et al., 2000
), CHO-hAR-MMTVluc cells (de Gooyer et al., 2003
), and COS-hAR-MMTVluc cells (Paris et al., 2002c
). So far, the only cell line that uses a simpler reporter construct, thereby avoiding influences by nonsteroidal regulatory pathways is derived from the human breast cancer cell line T-47D, stably transfected with a luciferase reporter under transcriptional control of the PB-ARE2 androgen response element (Blankvoort et al., 2001
). This stable cell line shows additional hormone class specificity, as it mainly responds to progestins, due to the known over-expression of PR in T-47D cells, and relatively low endogenous AR levels (this study, Sonneveld et al., unpublished results; Sutherland et al., 1988
), making it less suitable as a selective screening tool.
Due to the known problems of transcriptional interference between C3 group nuclear receptors, we choose to systematically select a line with an extremely low background activity of PR and GR while supporting an optimal androgen response when the cognate receptor was transiently introduced. This led to selection of the U2-OS cell line, which has the additional advantage of being robust, genetically stable, and of fast proliferation compared to most prostatic cell lines. Through the introduction of a highly selective and responsive reporter gene, we generated the AR CALUX cell line.
Our results with the AR CALUX cell line show that it readily classifies the activities of pure chemicals, including natural and synthetic steroids. The EC50 values obtained with these compounds (partly listed in Table 2) correlate very well with corresponding EC50 values obtained with another established AR reporter cell line, the CHO-hAR-MMTVluc (de Gooyer et al., 2003; van der Burg et al., manuscript in preparation). These data are also consistent with binding affinities to the AR of these chemicals and the in vivo Hershberger assay (van der Burg et al., manuscript in preparation). Not all tested androgens reached the maximal induction level of DHT (Table 2). For example, R1881 only reached a relative transactivation activity of 69% compared to DHT. The reason for this lower maximal response is not clear, but could be due to differences in ligand-dependent AR stabilization as a result of different rates of androgen dissociation and AR degradation (Zhou et al., 1995
), as shown for antiestrogens on ER stability (Gibson et al., 1991
; van den Bemd et al., 1999
). Ligand-dependent AR protein forms with different transactivation capacity as described for R1881 previously (Kuil et al., 1996
) might also be an explanation for the lower maximal induction level of R1881. On the other hand, the synthetic androgen MENT was able to induce a supramaximal response (RTA = 121%). This supra-induction was also observed for genistein and o,p'DDT on ER
(this study; Legler et al., 1999
) and an explanation for this phenomenon could be ligand-dependent differences in the ability of receptor to bind coactivators, such as TIF2 and SCR-1a as shown recently for ER by xenoestrogens (Routledge et al., 2000
).
Weak activation of reporter gene activity was obtained at high concentrations of the strongest synthetic glucocorticoid dexamethasone only, while other high-affinity GR ligands such as hydrocortisone and corticosterone had little or no effect (Table 2). This data correlates with the affinity of these compounds to the GR and their response in the GR CALUX bioassay, with the latter showing EC50 values of 0.5 nM for dexamethasone, 5 nM for hydrocortisone, and 15 nM for corticosterone (Sonneveld et al., manuscript in preparation). Accordingly, GR-mediated activity is insignificant in the AR CALUX bioassay, since only weak effects can be observed with high concentrations of the strongest glucocorticoids. Such activities are very unlikely to be present in chemicals not designed to be glucocorticoids.
While active androgens bind directly to the androgen receptor and induce luciferase activity in AR CALUX cells, androgen precursors need metabolic activation. Androstenedione is a very weak binder to AR (0.1% compared to DHT; van der Burg et al., manuscript in preparation), but is a potent androgen in the AR CALUX bioassay, suggesting the presence of the metabolic enzyme 17ß-HSD type 5 (with 17ß-ketosteroid reductase activity), converting androstenedione to testosterone in AR CALUX cells. DHEA is a very weak transactivator of AR (EC50 > 10 µM; RTA = 11%), indicating absence or low activity of the 3ß-HSD enzyme responsible for the conversion of DHEA to androstenedione. Preliminary PCR data show that 17ß-HSD (type 5), but not 3ß-HSD (type 1 and 2) is expressed in U2-OS cells (data not shown). On the other hand, the precursor progesterone shows induction of luciferase in AR CALUX cells (EC50 = 8.7 µM; RTA = 36%). This could mean that the enzyme CYP17 is present in AR CALUX cells, converting progesterone via OH-progesterone (17-hydroxylase activity) to androstenedione (17, 20 lyase activity). Indeed, PCR experiments showed the expression of CYP17 in U2-OS cells (data not shown). Alternatively, since progesterone can bind AR (2% compared to DHT), this possibly results in direct AR transactivation (van der Burg et al., manuscript in preparation). Cross-talk with PR is not an issue in the bioassay, since the PR specific synthetic ligand Org 2058 did not show activity in the AR CALUX bioassay.
Figure 4 shows that the AR CALUX bioassay readily picks up antagonistic effects of known environmental anti-androgens. Pesticides from the DDT family clearly can antagonize the AR with IC50 values around 1 µM, while the metabolite HPTE was even 30 times more potent than its parental compound methoxychlor, making this compound one of the strongest environmental AR antagonists found so far in the AR CALUX bioassay. The antagonistic activity of vinclozolin (IC50 = 1 µM) is rather potent compared to another AR reporter cell line, the MDA-kb2 cells (IC50 = 10 µM; Wilson et al., 2002). In the latter cell line, metabolites of vinclozolin, M1 and primarily M2, are far more potent than the parental compound, suggesting that in the AR CALUX cells vinclozolin can be metabolized to more active AR antagonistic compounds like M1 and M2.
Compounds, particularly at µM levels or higher, can occasionally nonspecifically repress responses in reporter gene assays. This can be due to overall cytotoxicity, ultimately leading to cell death, but can also be due to more specific effects such as inhibition of protein synthesis or mRNA transcription. In our experience the latter effects precede the more general cytotoxic effects, with overt cell death as the least sensitive parameter. Therefore, controls should be assessing nonspecific repression of reporter gene activity rather than overall cytotoxicity and cell death. Constitutively expressed reporter genes that often are used as controls have the drawback that no bona fide constitutive promoters have been identified so far; therefore the use of these controls should be avoided. In the case of steroid-receptor-mediated responses, the best control for nonspecific inhibition is considered the determination of the effect of the test compound on the reporter gene activation by an excess of high-affinity agonist. This approach worked well with the ligands tested, and all inhibitory responses were reversed by co-incubation with excess DHT, demonstrating the specificity of the response. Squelching of common cofactors by other nuclear (hormone) receptors is a well-known mechanism of interference and might therefore produce false-negative results. An example for this type of mechanism is the interference between PR and ER (Kraus et al., 1995). However, squelching seems not to be prominent in U2-OS derived CALUX bioassays, since they do not express high levels of steroid receptors other than the stably introduced receptor of interest. This is shown by the fact that progestins and glucocorticoids do not interfere with DHT- or E2-induced luciferase activity in the AR and ER
CALUX bioassays, respectively, while androgens do not show reduced E2-induced luciferase activity in the ER
CALUX bioassay (Table 2). Another receptor shown to possess squelching effects with nuclear hormone receptors is the aryl hydrocarbon receptor (AhR). Interference of the AhR ligand TCDD on ER signaling was demonstrated in T-47D cells (ER CALUX bioassay) expressing functional AhR (Legler et al., 1999
; Sonneveld et al., 2003
), but not in U2-OS cells (ER
CALUX bioassay) not expressing AhR (Sonneveld et al., 2003
).
Several environmental contaminants have been shown to activate the estrogen receptor, and if their effects are additive, these may contribute to environmental and human health impacts, most notably feminization of male fish (Gimeno et al., 1996). Remarkably, in contrast to the estrogen receptor that is mostly activated by environmental pollutants, the androgen receptor seems to be prone to antagonism rather than agonism (Paris et al., 2002a
; Sohoni and Sumpter, 1998
; Willemsen et al., 2004
). This point is emphasized by the fact that the pesticides tested in our study (o,p'-DDT, p,p'-DDT, methoxychlor, HPTE) with ER
agonistic activity were found to be also AR antagonists. This phenomenon may contribute to the observed feminization of male fish. The exact reason why the androgen receptor seems to be more readily inhibited rather than activated is unclear. Possibly, a more complex mechanism of activation with interaction between C- and N-terminus and interactions with specific coactivators (Dubbink et al., 2004
; He et al., 2002
) may be involved in the difficulty of AR-binding pollutants leading to activation of this receptor.
The high sensitivity and high selectivity of the AR and ER CALUX bioassays allowed direct measurements in nonextracted biological samples. As a potential clinical application, very low volumes of human serum were applied directly to the CALUX bioassays. The presence of androgens and estrogens in human serum was shown by the AR and ER
CALUX bioassays, respectively, demonstrating that these bioassays can be used to measure low levels of bioavailable hormones directly in very small amounts of human serum (approximally 30 µl), as demonstrated recently for glucocorticoids (Sonneveld et al., manuscript in preparation; Vermeer et al., 2003
). The use of such low volumes makes usage of these CALUX bioassays for human serum even potentially applicable to infants, where low-volume sample taking is desired. In fetal calf serum, high estrogenic activity was measured. For this reason, charcoal-stripped serum is used in the CALUX bioassays to remove all steroids present. However, we could not demonstrate androgenic activity in fetal calf serum, suggesting either the presence of low levels of active androgens in fetal calf serum, or the presence of inactive precursors in the fetal serum which may be converted to active androgens in target tissues.
The established U2-OS-based CALUX systems for estrogens, glucocorticoids, and progestins provide complementary screening systems to the described AR CALUX system (see Table 1 for an overview). Use of these CALUX bioassays will allow the determination of full steroidal activity profiles of compounds using the same cellular background. This has the advantage that maintenance of the cells can be standardized, but also provides several advantages in terms of screening efficacy. Since compounds have different effects on the various steroid receptors effect profiles can be derived, that potentially give more information on the biological risk or benefit, the specificity of the response, and the nature of the biological active components in mixture, as compared with measurement of a single endpoint. In particular, we expect that profiles of compounds generated by CALUX bioassays, either alone or in conjunction with additional endpoints, may be an important step in the prescreening of chemicals, allowing risk ranking and toxicological prioritization and thereby reducing the number of animal tests to be undertaken. Chemical profiles may also be important in the first steps of identification of chemical pollutants in complex mixtures such as food, feed, and environmental matrices. The discriminative power between compounds in an "effect profiling" system will greatly improve by expanding the number of cell lines used. This will, however, lead to increased handling and accordant additional costs. Auto-motion of the handling will therefore be an important future step in an efficient "effect profiling" system. With this in mind, the use of a single robust parent cell line, such as the U2-OS cells, with identical culture and handling conditions greatly facilitates the possibilities for automation.
![]() |
ACKNOWLEDGMENTS |
---|
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Aurrekoetxea-Hernandez, K., and Buetti, E. (2004). Transforming growth factor beta enhances the glucocorticoid response of the mouse mammary tumor virus promoter through Smad and GA-binding proteins. J. Virol. 78, 22012211.
Avances, C., Georget, V., Terouanne, B., Orio, F., Cussenot, O., Mottet, N., Costa, P., and Sultan, C. (2001). Human prostatic cell line PNT1A, a useful tool for studying androgen receptor transcriptional activity and its differential subnuclear localization in the presence of androgens and antiandrogens. Mol. Cell. Endocrinol. 184, 1324.[CrossRef][ISI][Medline]
Bentel, J. M., Birrell, S. N., Pickering, M. A., Holds, D. J., Horsfall, D. J., and Tilley, W. D. (1999). Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells. Mol. Cell. Endocrinol. 154, 1120.[CrossRef][ISI][Medline]
Blankvoort, B. M., de Groene, E. M., van Meeteren-Kreikamp, A. P., Witkamp, R. F., Rodenburg, R. J., and Aarts, J. M. (2001). Development of an androgen reporter gene assay (AR-LUX) utilizing a human cell line with an endogenously regulated androgen receptor. Anal. Biochem. 298, 93102.[CrossRef][ISI][Medline]
Brinkmann, A. O., Faber, P. W., van Rooij, H. C., Kuiper, J. M., Ris, C., Klaassen, P., van der Korput, J. A. G. M., Voorhorst, M. M., van Laar, J. H., Mulder, E., et al. (1989). The human androgen receptor: Domain structure, genomic organization and regulation of expression. J. Steroid Biochem. 34, 307310.[CrossRef][ISI][Medline]
Claessens, F., Verrijdt, G., Schoenmakers, E., Haelens, A., Peeters, B., Verhoeven, G., and Rombauts, W. (2001). Selective DNA binding by the androgen receptor as a mechanism for hormone-specific gene regulation. J. Steroid Biochem. Mol. Biol. 76, 2330.[CrossRef][ISI][Medline]
de Gooyer, M. E., Deckers, G. H., Schoonen, W. G. E. J., Verheul, H. A., Kloosterboer, H. J. (2003). Receptor profiling and endocrine interactions of tibolone. Steroids 68, 2130.[CrossRef][ISI][Medline]
Di Croce, L., Koop, R., Venditti, P., Westphal, H. M., Nightingale, K. P., Corona, D. F., Becker, P. B., and Beato, M. (1999). Two-step synergism between the progesterone receptor and the DNA-binding domain of nuclear factor 1 on MMTV minichromosomes. Mol. Cell 4, 4554.[ISI][Medline]
Dubbink, H. J., Hersmus, R., Verma, C. S., van der Korput, H. A., Berrevoets, C. A., van Tol, J., Ziel-Van Der Made, A. C., Brinkmann, A. O., Pike, A. C., and Trapman, J. (2004). Distinct recognition modes of FXXLF and LXXLL motifs by the androgen Receptor. Mol. Endocrinol. 18, 21322150.
Evans, N. A. (2004). Current concepts in anabolic-androgenic steroids. Am. J. Sports Med. 32, 534542.
Folkers, G. E., and van der Saag P. T. (1995). Adenovirus E1A functions as a cofactor for retinoic acid receptor beta (RAR beta) through direct interaction with RAR beta. Mol. Cell. Biol. 15, 58685878.[Abstract]
Gibson, M. K., Nemmers, L. A., Beckman, W. C., Davis, V. L., Curtis, S. W., and Korach, K. S. (1991). The mechanism of ICI-164,384 antiestrogenicity involves rapid loss of estrogen receptor in uterine tissue. Endocrinology 129, 20002010.[Abstract]
Gimeno, S., Gerritsen, A., Bowmer, T., and Komen, H. (1996). Feminization of male carp. Nature 384, 221222.[CrossRef][ISI][Medline]
Glass, C. K. (1994). Differential recognition of target genes by nuclear receptor monomers, dimers and heterodimers. Endocr. Rev. 15, 391407.[ISI][Medline]
Green, S., Walter, P., Greene, G., Krust, A., Goffin, C., Jensen, E., Scrace, G., Waterfield, M., and Chambon, P. (1986). Cloning of the human oestrogen receptor cDNA. J. Steroid Biochem. 24, 7783.[CrossRef][ISI][Medline]
Hartig, P. C., Bobseine, K. L., Britt, B. H., Cardon, M. C., Lambright, C. R., Wilson, V. S., and Gray, L. E., Jr. (2002). Development of two androgen receptor assays using adenoviral transduction of MMTV-Luc reporter and/or hAR for endocrine screening. Toxicol. Sci. 66, 8290.
He, B., Lee, L. W., Minges, J. T., and Wilson, E. M. (2002). Dependence of selective gene activation on the androgen receptor NH2- and COOH-terminal interaction. J. Biol. Chem. 277, 2563125639.
Interagency Coordinating Committee on the Validation of Alternative Methods (ICVAMM) (2003). ICCVAM evaluation of in vitro test methods for detection of potential endocrine disruptors: Estrogen receptor and androgen receptor binding and transcriptional activation assays. NIH publication no. 034503.
Kalkhoven, E., Kwakkenbos-Isbrücker, L., de Laat, S. W., van der Saag, P. T., and van der Burg, B. (1994). Synthetic progestins induce proliferation of breast tumor cell lines via the progesterone or estrogen receptor. Mol. Cell. Endocrinol. 102, 4552.[CrossRef][ISI][Medline]
Kelce, W. R., Stone, C. R., Laws, S. C., Gray, L. E., Kemppainen, J. A., and Wilson, E. M. (1995). Persistent DDT metabolite p,p'-DDE is a potent androgen receptor antagonist. Nature 375, 581585.[CrossRef][ISI][Medline]
Kelce, W. R., and Wilson, E. M. (1997). Environmental anti-androgens: Developmental effects, molecular mechanisms and clinical implications. J. Mol. Med. 75, 198207.[CrossRef][ISI][Medline]
Kraus, W. L., Weis, K. E., and Katzenellenbogen, B. S. (1995). Inhibitory cross-talk between steroid hormone receptors: Differential targeting of estrogen receptor in the repression of transcriptional activity by agonist- and antagonist-occupied progestin receptors. Mol. Cell. Biol. 15, 18471857.[Abstract]
Kuil, C. W., and Mulder, E. (1996). DNA-binding ability of androgen receptors in whole cells: Implications for the action of androgens and antiandrogens. Endocrinology 137, 18701877.[Abstract]
Legler, J., Broekhof, J. L. M., Brouwer, A., Lanser, P. H., Murk, A. J., van der Saag, P. T., Wester, P., Zivkovic, D., and van der Burg, B. (2000). A novel in vivo bioassay for (xeno-) estrogens using transgenic zebrafish. Environ. Sci. Technol. 34, 44394444.[CrossRef][ISI]
Legler, J., van den Brink, C. E., Brouwer, A., Murk, A. J., van der Saag, P. T., Vethaak, A. D., and van der Burg, B. (1999). Development of a stably transfected estrogen receptor-mediated luciferase reporter gene assay in the human T47D breast cancer cell line. Toxicol. Sci. 48, 5566.
Lemmen, J. G., Arends, R. J., van Boxtel, A. L., van der Saag, P. T., and van der Burg, B. (2004). Tissue- and time-dependent estrogen receptor activation in estrogen reporter mice. J. Mol. Endocrinol. 32, 689701.
Lemmen, J. G., van den Brink, C. E., Legler, J., van der Saag, P. T, and van der Burg, B. (2002). Detection of oestrogenic activity of steroids present during mammalian gestation using ERalpha and ERbeta specific in vitro assays. J. Endocrinol. 174, 435446.
Mangelsdorf, D. J., Thummel, C., Beato, M., Herrlich, P., Schutz, G., Umesono, K., Blumberg, B., Kastner, P., Mark, M., Chambon, P., et al. (1995). The nuclear receptor superfamily: The second decade. Cell 83, 835839.[ISI][Medline]
Meyer, H. H. (2001). Biochemistry and physiology of anabolic hormones used for improvement of meat production. APMIS 109, 18.[ISI][Medline]
McKenna, N. J., and O'Malley, B. W. (2002). Combinatorial control of gene expression by nuclear receptors and coregulators. Cell 108, 465474.[ISI][Medline]
Nuclear Receptors Nomenclature Committee (1999). A Unified Nomenclature System for the Nuclear Receptor Superfamily. Cell 97, 161163.[ISI][Medline]
Ouatas, T., Clare, S. E., Hartsough, M. T., De La Rosa, A., and Steeg, P. S. (2002). MMTV-associated transcription factor binding sites increase nm23-H1 metastasis suppressor gene expression in human breast carcinoma cell lines. Clin. Exp. Metastasis 19, 3542.[CrossRef][ISI][Medline]
Paris, F., Balaguer, P., Terouanne, B., Servant, N., Lacoste, C., Cravedi, J. P., Nicolas, J. C., and Sultan, C. (2002a). Phenylphenols, biphenols, bisphenol-A and 4-tert-octylphenol exhibit alpha and beta estrogen activities and antiandrogen activity in reporter cell lines. Mol. Cell. Endocrinol. 193, 4349.[CrossRef][ISI][Medline]
Paris, F., Servant, N., Terouanne, B., Balaguer, P. Nicolas, J. C., and Sultan, C. (2002b). A new recombinant cell bioassay for ultrasensitive determination of serum estrogenic bioactivity in children. J. Clin. Endocrinol. Metab. 87, 791797.
Paris, F., Servant, N., Terouanne, B., and Sultan, C. (2002c). Evaluation of androgenic bioactivity in human serum by recombinant cell line: Preliminary results. Mol. Cell. Endocrinol. 198, 123129.[CrossRef][ISI][Medline]
Quaedackers, M. E., van den Brink, C. E., Wissink, S., Schreurs, R. H. M., Gustafsson, J. A., van der Saag, P. T., and van der Burg, B. (2001). 4-Hydroxytamoxifen Trans-represses nuclear factor-kB activity in human osteoblastic U2-OS cells through estrogen receptor (ER) alpha, and not through ERbeta. Endocrinology 142, 11561166.
Routledge, E. J., White, R., Parker, M. G., and Sumpter, J. P. (2000). Differential effects of xenoestrogens on coactivator recruitment by estrogen receptor (ER) and ERß. J. Biol. Chem. 275, 3598635993.
Schule, R., Muller, M., Kaltschmidt, C., and Renkawitz, R. (1988). Many transcription factors interact synergistically with steroid receptors. Science 242, 14181420.[ISI][Medline]
Sohoni, P., and Sumpter, J. P. (1998). Several environmental oestrogens are also anti-androgens. J. Endocrinol. 158, 327339.
Sonneveld, E., Jansen, H. J., Riteco, J. A. C., Brouwer, A., and van der Burg, B. (2003). CALUX reporter cell lines as sensitive bioassays for monitoring biological activity of various classes of steroid hormones reveal the repressive effect of TCDD on the estrogen receptor. Organohal. Compounds 60, 263266.
Sonneveld, E., van den Brink, C. E., van der Leede, B. M., Schulkes, R. K., Petkovich, M., van der Burg, B., and van der Saag, P. T. (1998). Human retinoic acid (RA) 4-hydroxylase (CYP26) is highly specific for all-trans-RA, and can be induced through retinoic acid receptors in human breast and colon carcinoma cells. Cell Growth Differ. 9, 629637.[Abstract]
Spangenberg, C., Eisfeld, K., Stunkel, W., Luger, K., Flaus, A., Richmond, T. J., Truss, M., and Beato, M. (1998). The mouse mammary tumour virus promoter positioned on a tetramer of histones H3 and H4 binds nuclear factor 1 and OTF1. J. Mol. Biol. 278, 725739.[CrossRef][ISI][Medline]
Sutherland, R. L., Hall, R. E., Pang, G. Y., Musgrove, E. A., and Clarke, C. L. (1988). Genetic instability and the development of steroid hormone insensitivity in cultured T 47D human breast cancer cells. Cancer Res. 48, 43404347.[Abstract]
Terouanne, B., Tahiri, B., Georget, V., Belon, C., Poujol, N., Avances, C., Orio, F., Balaguer, P., and Sultan, C. (2000). A stable prostatic bioluminescent cell line to investigate androgen and antiandrogen effects. Mol. Cell. Endocrinol. 160, 3949.[CrossRef][ISI][Medline]
Uchiumi, F., Sato, T., and Tanuma, S. (1998). Identification and characterization of a tannic acid-responsive negative regulatory element in the mouse mammary tumor virus promoter. J. Biol. Chem. 273, 1249912508.
van den Bemd, G. J. C. M., Kuiper, G. G. J. M., Pols, H. A. P., and van Leeuwen, J. P. T. M. (1999). Distinct effects on the conformation of estrogen receptor alpha and beta by both the antiestrogens ICI 164,384 and ICI 182,780 leading to opposite effects on receptor stability. Biochem. Biophys. Res. Commun. 261, 15.[CrossRef][ISI][Medline]
van der Burg, B., Rutteman, G. R., Blankenstein, M. A., de Laat, S. W., and van Zoelen, E. J. (1988). Mitogenic stimulation of human breast cancer cells in a growth factor-defined medium: Synergistic action of insulin and estrogen. J. Cell Physiol. 134, 101108.[ISI][Medline]
Vermeer, H., Hendriks-Stegeman, B. I., van den Brink, C. E., van der Saag, P. T., van der Burg, B., van Buul-Offers, S. C., and Jansen, M. (2003). A novel specific bioassay for the determination of glucocorticoid bioavailability in human serum. Clin. Endocrinol. 59, 4955.[CrossRef][ISI][Medline]
Willemsen, P., Scippo, M., Kausel, G., Figueroa, J., Maghuin-Rogister, G., Martial, J. A., and Muller, M. (2004). Use of reporter cell lines for detection of endocrine-disrupter activity. Anal. Bioanal. Chem. 378, 655663.[CrossRef][ISI][Medline]
Wilson, V. S., Bobseine, K., Lambright, C. R., and Gray, L. E., Jr. (2002). A novel cell line, MDA-kb2 that stably expresses an androgen- and glucocorticoid-responsive reporter for the detection of hormone receptor agonists and antagonists. Toxicol. Sci. 66, 6981.
Zhou, Z. X., Lane, M. V., Kemppainen, J. A., French, F. S., and Wilson, E. M. (1995). Specificity of ligand-dependent androgen receptor stabilization: Receptor domain interactions influence ligand dissociation and receptor stability. Mol. Endocrinol. 9, 208218.[Abstract]