CA2352203A1 - Bioassay for identifying estrogen receptor-.beta./.alpha. selective modulators - Google Patents

Bioassay for identifying estrogen receptor-.beta./.alpha. selective modulators Download PDF

Info

Publication number
CA2352203A1
CA2352203A1 CA002352203A CA2352203A CA2352203A1 CA 2352203 A1 CA2352203 A1 CA 2352203A1 CA 002352203 A CA002352203 A CA 002352203A CA 2352203 A CA2352203 A CA 2352203A CA 2352203 A1 CA2352203 A1 CA 2352203A1
Authority
CA
Canada
Prior art keywords
cells
beta
alpha
expression
estrogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002352203A
Other languages
French (fr)
Inventor
Heather Harris
Ramesh A. Bhat
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wyeth LLC
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2352203A1 publication Critical patent/CA2352203A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/723Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Abstract

The present invention provides novel assay methods for identifying compounds that selectively activate estrogen receptors (ER) of the alpha or beta subtype. In particular, the results from two assays, one measuring ER-.beta. activity and the other measuring ER-.alpha. activity are interpreted. The assay measuring ER-.beta. activity uses cells comprising endogenous metallothionein-II as well as a DNA plasmid comprising a polynucleotide encoding human ER-.beta.. The assay monitors expression of metallothionein-I I- mRNA in said cells, wherein the level of metallothionein-II expression is regulated when a potential ligand binds to ER-.beta.. The assay measuring ER - .alpha. activity uses cells comprising ER-.alpha. as well as DNA plasmid comprising a reporter gene linked to an estrogen response element. The assay monitors expression of the reporter gene, wherein the level of reporter gene is regulated when a potential ligand binds to ER-.alpha.. Compounds which modulate activity in one assay but have little or no activity in the other assay are defined as estrogen receptor subtype selective.

Description

BIOASSAY FOR IDENTIFYING ESTROGEN RECEPTOR-~i/oc SELECTIVE
MODULATORS
Technical Field The present invention relates to hormone receptors, and, more particularly to methods for identifying compounds that selectively activate estrogen receptors (ER) of the alpha or beta subtype, as well as a test kit for use in the methods.
Background of the Invention Proteins which regulate gene expression are essential for cell function. A well-studied family of gene regulatory proteins is the steroid hormone receptor superfamily. These receptor proteins enable cells to respond to various hormones by activating or repressing specific genes.
One member of this family is the ER. Estrogens are classically known as important hormones in sexual development and reproductive function. It is well known that estrogens affect cell proliferation and differentiation in target tissues by binding to ERs in target cells. Estrogen replacement therapy is a well established treatment for prevention and/or amelioration of osteoporosis in postmenopausal women (Sagraves, 1995,; Lobo, 1995) because these compounds have been demonstrated to prevent bone loss and fractures in women. Additionally, estrogen replacement therapy has been associated with a decreased mortality from cardiovascular disease. Finally, ongoing studies suggest that estrogens may provide WO 00/37b81 PCT/US99/29856 benefit to the central nervous system with regard to cognitive improvement and a decrease in Alzheimer's disease.
The classic ER, now designated ER-a, exhibits a modular structure with distinct domains involved in ligand binding, receptor dimerization, DNA binding, and transcriptional activation (Green et al, 19861. The nucleotide sequence of the DNA binding domain is conserved among steroid hormone receptors. Kuiper et al ( 1996) exploited this similarity in an attempt to identify additional members of this family. The new member they discovered was recognized to be an ER because the amino acids in the DNA binding domain were almost identical to those of ER-a, , the in vitro translated protein specifically bound [3H]-estradiol with nanomolar affinity and was able to regulate transcription from a simple estrogen response element. This protein has been designated ER-(3 to distinguish it from the previously known form Inow called ER-a). Human and mouse ER-(3 have also been cloned Bhat et.al. 1998; Mosselman, 1996; Pettersson et al, 1997; Tremblay et al, 1997). Following ER-(3's discovery, most work has focused on mapping the distribution of its mRNA in normal and neopfastic tissues, characterizing its binding affinity for a wide variety of ligands, and assessing its interaction with ER-a.
ER-~3 mRNA is detectable by RT-PCR and in situ hybridization in a wide variety of tissues. While its distribution overlaps that of ER-a, some tissues express only one receptor type. For example both receptors are found in the uterus, ovary and pituitary, whereas ER-~3 appears more prominent in the prostate, lung and bladder (Kuiper et al, 1996). When examined by in situ hybridization, further distinctions between receptor distribution can be made. ER-~i mRNA is expressed in the epithelial cells of the rat prostate; ER-a is found in the stromal compartment (Kuiper et al, 1996). In the rat ovary, ER-(3 appears in the granulosa cells; ER-a in the stroma (Shughrue et al, 1996; personal communication). In the rat hypothalamus, ER-~3 but not ER-a, message is expressed in the paraventricular region, whereas both messages are seen in the preoptic area (Shughrue et al, 19961. Interestingly, significant species differences may exist in the relative levels of ER-(3 and ER-a mRNA in certain organs.
For example, ER-(3 mRNA is highly expressed in the rat prostate, whereas more modest levels are detected in the human prostate. ER-/3 predominates over ER-a in the rat prostate (Kuiper et al, 1996; Lau et al, 1998; Enmark et al, 1997), but the levels are more equal in the mouse prostate (Couse et al, 19971.
Several groups have characterized ER expression in tumor samples and cancer cell lines. Most work has been done in the breast where one study reports ER-~i mRNA using RT-PCR in 70% of forty breast biopsy samples (Dotzlaw et al, 1996). No correlation was noted between ER-a and ER-(3 expression. Another study also reports heterogeneity of ER-a/~i mRNA expression in breast tumor samples, with some tumors having only ER-a mRNA and others expressing mRNA for both receptor subtypes (Enmark et al, 1997). Among breast cancer cell lines, MDA MB 231 has only ER-~3 mRNA, and conflicting reports exist about the ER status of MCF-7 cells (Enmark, 1997; Kuiper et al, 1997; Vladusic et al, 1998; R.
Henderson, unpublished observationsl.
Although the figand binding domain of human ER-a and ER-~i are 59% identical at the amino acid level (Enmark et al, 1997), the binding affinity of 17(3-estradiol is quite similar. Some compounds show marked selectivity for either ER-a or ER-~3. Genistein is a phytoestrogen which binds with approximately 10-25 fold higher affinity for ER-(3 (Kuiper 1996, H. Harris unpublished observations). On the other hand raloxifene binds about 20 fold better to ER-a than ER-(3 (H. Harris, unpublished observations).
When ER-a and ER-~i are coexpressed , interaction can be measured by several methods including a mammalian two-hybrid system, glutathione S transferase pulldown and gel shift/supershift assays (Cowley et al, 1997; Pettersson et al, 1997; Ogawa et al, 19981. Since these receptors can heterodimerize, estrogens' effect on tissues containing both receptors may be mediated by a complex interaction between ER-a and ER-Vii.
One valuable tool in determining the function of ER-(3 is the estrogen receptor-a knockout (ERKO) mouse (Lubahn et al, 1993).
Because these mice lack functional ER-a, they can help define the physiological roles of both ER-a and ER-Vii. One study has compared the effectiveness of 17-~i estradiol treatment in ameliorating consequences of artificially induced vascular injury to carotid arteries in wild type and ERKO mice (lafrati et ai, 19971. In both types of mice, pharmacological doses of 17-(3 estradiol suppressed the increase in medial area and smooth muscle cell proliferation seen in the vehicle treated d animals. It 5 is thought that these responses to endothelial denudation may narrow the lumen of the vessel, thus restricting blood flow. Because 17-(3 estradiol was equally effective in ERKO as wild type mice, one interpretation is that ER-a is not necessary for this response. Because ER-(3 mRNA is also expressed in these vessels, perhaps it mediates 17-(3 estradiol's action.
However, direct evidence supporting this hypothesis is lacking. Another example of ERKO mice responding to 17-(3 estradiol replacement was described in a recent poster and abstract (Pan et al, 1997, and personal communication). This study reports a loss of femoral bone mineral density and trabecular bone volume in ERKO mice after ovariectomy and an increase in these parameters upon treatment with 17-~3 estradiol (but not dihydrotestosteronel. Again, based on indirect evidence, a suggestion is made that ER-/3 has a physiological role.
Thus, while certain aspects of ER-~i have been characterized, the art fails to provide methods for identifying ligands which are functionally selective for ER-a or ER-j3. Such an assay would be greatly advantageous to the pharmacological industry to uncover the possible therapeutic applications of the ER subtypes.
Summary of the Invention The present invention provides method for screening a test compound that binds to an ER in a receptor binding assay, wherein said method detects ER-(3 polypeptide-mediated transcription, said method comprising the steps of:
(a) providing a cell which comprises at least one estrogen-induced DNA sequence encoding metallothionein IMT-II) and at least one DNA
sequence encoding ER-p polypeptide, wherein said receptor is transcriptionally active;
(b) contacting said cell with either said test compound which binds ER or a control; and (c) detecting the expression of said MT-II, wherein enhanced expression of said MT-II relative to a control indicates that said test compound has estrogen agonist activity.
This invention further provides a method for screening a test compound that binds to the ER in a receptor binding assay, wherein said method detects inhibition of ER-(3 )- polypeptide-mediated transcription, said methad comprising the steps of:
(a) providing a cell which comprises at least one estrogen-induced DNA sequence encoding metallothionein (MT-II) and at least one DNA
sequence encoding ER-~i polypeptide, wherein said receptor is transcriptionally active;
(b) contacting said cell with one or more estrogen(s) in the presence of the test compound known to bind ER; and (c) and detecting the expression of said MT-II, wherein decreased expression of said MT-II relative to the addition of one or more estrogenls) alone indicates that said test compound has estrogen antagonist activity A method of screening test compounds to identify drug candidates which mimic estrogens' effect on ER-Vii- or ER-a-mediated transcription is further provided wherein said method comprises the steps of:
(a) contacting said test compound with a plurality of:
(i1 first cells comprising at least one DNA sequence encoding metallothionein (MT-II) and at least one DNA
sequence encoding an ER-~3 polypeptide, wherein said receptor is transcriptionally active and (ii) second cells comprising an ERE reporter gene construct, wherein said cells express ER-a polypeptide;
(b) identifying compounds which increase expression of MT-II in said first cells relative to control but have minimal effect on expression of said reporter gene in said second cells, wherein said compounds are considered ER-p selective; or (c) identifying compounds which increase expression of the reporter gene in said second cells relative to control but have minimal effect on expression of MT-II in said first cells, wherein said compounds are considered ER-a selective.
Lastly, the present invention provides a method of screening test compounds to identify drug candidates which inhibit estrogen's effect on ER-(3- or ER-a-mediated transcription, said method comprising the steps of:
(a) contacting said test compound in the presence and absence of one or more estrogen(s) with a plurality of:
(i) first cells comprising at least one endogenous DNA
encoding a metallothionein (MT-II) gene and DNA encoding a ER-~i polypeptide, wherein said receptor is transcriptionally active and (ii) second cells comprising an ERE reporter gene construct, wherein said cells express ER-a polypeptide; and (b) identifying compounds which decrease expression of MT-il in said first cells relative to treatment with one or more estrogen(s) alone but have minimal effect on expression of said reporter gene in said second cells, wherein said compounds are considered ER-(3 selective; or (c) identifying compounds which decrease expression of the reporter gene in said second cells relative to treatment with one or more estrogen(s) alone but have minimal effect on expression of MT-II in said first cells, wherein said compounds are considered ER-a selective.

2fi-08-2000 Brief Descriytion of the Fi4ures Figure 1: RT-PCR amplification of ER-~3 and ER-a a from Saos -2 and LNCaPLN3 cells.
Figure 2: Sequencing gel separating amplified cDNAs and 17-~i estradiol upregulation of fragment 6a.
Figure 3: (A) Nucleotide sequence ISEQ ID No. 12) of the regulated fragment and its alignment with human MT-II, SEQ ID No. 13. (B) Translation of fragment sequence from first methionine to stop codon ('), SEQ ID No. 14 Figure 4: Regulation of MT-II in two cell lines. To determine fold change in mRNA, MT-II signal was normali2ed to that of GAPDH, and compared to the control cells.
Figure 5: Dose response of 17-~3 estradiol regulation of MT-II in Saos-2 cells. Result are shown for four different experiments and the ECso shown for each.
AMENDED SHEET

Figure 6: Time course of metallothionein-II regulation in Saos-2 cells.
Data is normalized with GAPDH and fold change calculated from the vehicle control at time =Ohr.
5 Figure 7: Receptor specificity of MT-II regulation in Saos-2 cells.
Figure 8: Regulation of MT-II in Saos-2 cells by various compounds.
Figure 9: Metallothionein-II regulation in cycloheximide-treated Saos-2 10 cells. (A) Measurement of protein synthesis during treatment with cycloheximide. IB) Whole cell ER binding assay after 8 hours of cycloheximide treatment. (C,DI Metallothionein-II regulation after 8 and 24 hours of treatment respectively.
Figure 10: MT is regulated by estrogens in the rat prostate.
Figure 1 1: Induction of MT-II in the rat prostate requires at least two days of dosing.
Figure 12: Screening strategy for ligands which selectively activate ER-~i and/or ER-a.
Detailed Description of the Invention AMENDED SHEET
The present invention provides an efficient way to screen large numbers of test compounds which selectively activate ERs of the a or (3 subtype. These compounds may have desirable properties for either the treatment or the prevention of various diseases mediated by estrogens, including but not limited to cancers (e.g. breast, ovarian, endometrial, prostate), endometriosis, osteoporosis and cardiovascular and central nervous system diseases.
Definitions (n the present description and claims, the following terms shall be defined as indicated below.
"hER(3L" is defined as the human form of ER-(3 described in Example 1 (the cDNA or its translated protein product).
"Estrogen" is defined as any ligand that can function as an estrogen agonist.
"Estrogen agonist" is defined as a compound that substantially mimics 17-(3 estradiol as measured in a standard assay for estrogenic activity, for example, cellular assays as described in Webb et al. ( 1992).
"Estrogen antagonist" is defined as a compound that substantially inhibits the effect of estrogen agonists as measured in a standard assay for estrogenic activity, for example, cellular as described in Webb et al. ( 1992) WO 00/3?681 PCT/US99/29856 "A functional ER" is defined as a receptor capable of transcriptional activation of endogenous or transfected genes as measured by changes in RNA, protein and/or downstream biological events.
"A nonfunctional ER" is defined as a receptor incapable of transcriptional activation of endogenous or transfected genes as measured by changes in RNA, protein and/or downstream biological events. A "test compound" includes but is not limited to any small molecule compound, peptide, polypeptide, natural product, toxin with potential biological activity.
A "ligand" is intended to include any substance that interacts with a receptor. .
"Transfection" is defined as any method by which a foreign gene is inserted into a cultured cell.
A "reporter" is defined as any substance that can be readily measured and distinguished from other cellular components. The reporter may be the transfected receptor DNA, the transcribed receptor mRNA, an enzyme, a binding protein or an antigen.
A "cell" useful for the present purpose is one which has the ability to respond to signal transduction through a given receptor.
"A "receptor binding assay" is an assay measuring the amount of ligand specifically interacting with a receptor. The ligand can be a radioligand (e.g. conjugated to 3H or 'z51), a fluorescinated ligand (either conjugated to a fluorochrome or possessing inherent fluorescence) or otherwise labeled so as to be detectable.
Description of the Assay The present invention relies on the discovery that the expression of MT-II is selectively regulated by the interaction of a ligand with ER-Vii. .
In the methods of the invention, the regulation of MT-ll activity may be used to provide a screening system that selectively detects both estrogen agonist or antagonist functional activity of a ligand following its interaction with ER-(3.
The methods typically comprise cultured cells that express functional human ER-~i and no, or a diminished amount of ER-a.
Such cells include but are not limited to Saos-2 (ATCC HTB-85) and LNCaPLN3. Preferred cells for this purpose include cells which over-express ER-(3, such as the cells described below which are recombinantly manipulated to over-express ER-Vii. The ER-(3 receptor may be modified in any way, such as in length; these modifications may result in increased ER-(3 selectivity and increased sensitivity of the assay.
One of skill will recognize that various recombinant constructs comprising ER-~i can be used in combination with any cell or line which lacks functional ER-a.
To screen a number of compounds for estrogen agonist activity, cells expressing ER-/3 are exposed to a test compound or a control solution (which is used to dissolve the test compound). The cells can be exposed while either growing in separate wells of a multi-well culture dish or in a semi-solid nutrient matrix. After treatment for a suitable period of time, MT-II mRNA is measured. An estrogen agonist will increase MT-II
mRNA when compared to treatment with the control solution alone.
To screen a number of compounds for estrogen antagonist activity, cells expressing ER-~i are exposed to one or more estrogens in the presence or absence of a test compound. The cells can be exposed while either growing in separate wells of a multi-well culture dish or in a semi-solid nutrient matrix. After treatment for a suitable period of time, MT-11 mRNA is measured. An estrogen antagonist will decrease MT-II mRNA
when compared to treatment with the estrogen solution alone.
Estrogenic or antiestrogenic compounds identified in the assays of the invention can be used in standard pharmaceutical compositions for the treatment of cancer, as components of oral contraceptives, or any other application in which the modulation of estrogen activity is desired. The pharmaceutical compositions can be prepared and administered using methods well known in the art. The pharmaceutical compositions are generally intended for parenteral, topical, oral or local administration for prophylactic and/or therapeutic treatment. The pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration. For example, unit dosage forms suitable for oral administration include powder, tablets, pills, and capsules.Suitable pharmaceutical formulations for use in the present invention arefound in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed. (19851. A variety of pharmaceutical compositions comprising compounds of the present invention and pharmaceutically effective carriers can be prepared.
5 Aaplications The methods and compositions of the present invention can be used to identify compounds that interact with ER-~3 , either to stimulate or to inhibit transcriptional activity . Such compounds include, without limitation, co-activator proteins, as well as estrogens and other steroids, steroid-like 10 molecules, or non-steroid-like molecules that act as agonists or antagonists.
Screening methods can also be used to identify tissue-specific estrogens.
Identification of ER-(3 -interactive compounds can be achieved by cell-free or cell-based assays. In one set of embodiments, purified ER-(3 is contacted with a labeled ligand, such as, e.g., 17-~i estradiol, in the 15 presence of test compounds to form test reactions, and in the absence of test compounds to form control reactions. The labeled moiety may comprise a radiolabel (such as, e.g., 3H or '251) or a fluorescent molecule.
Incubation is allowed to proceed for a sufficient time and under appropriate conditions to achieve specific binding, after which binding of labeled ligand to ER-(3 is measured (by monitoring, e.g., radioactivity, fluorescence, or fluorescence polarization). In one embodiment, the (igand binding domain of ER-(3 produced in E. coli is adsorbed to the wells of a microtiter dish and incubated with f3H]-17(3 estradiol in the absence or presence of test compounds. Alternatively, soluble receptor is incubated with the labeled ligand in the absence or presence of test compounds, and bound ligand is separated from free ligand, either by filtration on glass fiber filters or by using dextran-coated charcoal Whole cell binding assays may also be used in which bound ligand is separated from free ligand by rinsing. Cells used in these assays may either contain endogenous receptor, or may overexpress the receptor subsequent to stable or transient transfection or infection of an ER-j3 cDNA. Non-limiting examples of suitable cells include COS cells, HeLa cells, CHO cells, human umbilical vein endothelial cellsand yeast. Once a compound has been identified as an ER-~ -interactive compound by its binding activity, further in vivo and in vitro tests may be performed to determine the nature and extent of activity, i.e., as an agonist or antagonist (see below).
ER-~i -interactive compounds may also be identified using cell-based assays that measure transcriptional activation or suppression of endogenous or transfected estrogen-responsive genes. For example, agonists (such as, e.g., 17(3 -estradiol) block interleukin-1 (3 induction of endogenous E-selectin in primary human umbilical vein endothelial cells that express ER-/3 . Antagonists (such as, e.g., ICI-182780) block the agonist activity of 17(3 -estradiol. Non-limiting examples of other suitable endogenous estrogen-responsive promoter elements include those that regulate endothelin-1 (ET-1 ); HDL receptor (scavenger receptor type II); and enzymes involved in coagulation and fibrinolysis (such as, e.g., plasminogen activator inhibitor-1 and complement C31. Any promoter element that responds to estrogen may be used as an appropriate target, including, e.g., the NFkB binding site or the apolipoprotein A1 gene enhancer sequence.
In one set of embodiments, appropriate host cells are transfected with an expression vector encoding ER-p and the transfectants are incubated with or without one or more estrogens I in the presence or absence of test compounds. ER-~i activity is assessed by measuring transcriptional activation. This may be achieved by detection of mRNA
(using, e.g., Northern blot analysis, reverse transcriptase polymerase chain reaction, RNase protection assays) and/or by detection of the protein (using, e.g., immunoassays or functional assays). if activation of the target sequence initiates a biochemical cascade, downstream biological events may also be measured to quantify ER-(3 activity. ER-(3 -interactive compounds are identified as those that positively or negatively influence target sequence activation.
In another set of embodiments, appropriate host cells (preferably, mammalian 1 are co-transfected with an expression vector encoding ER-(3 and a reporter piasmid containing a reporter gene downstream of one or more ERE s. Transfected cells are incubated with or without an estrogen in the presence or absence of test compounds, after which ER-(3 activity is determined by measuring expression of the reporter gene. In a preferred embodiment, ER-(3 activity is monitored visually. Non-limiting examples of suitable reporter genes include luciferase, chloramphenicol acetyl transferase (CAT/, and green fluorescence protein.
Preferably, the methods of the present invention are adapted to a high-throughput screen, allowing a multiplicity of compounds to be tested in a single assay. Candidate estrogens and estrogen-like compounds include without limitation diethylstilbesterol, genistein, and estrone. Other ER-(3 -interactive compounds may be found in, for example, natural product libraries, fermentation libraries (encompassing plants and microorganisms), combinatorial libraries, compound files, and synthetic compound libraries.
For example, synthetic compound libraries are commercially available from Maybridge Chemical Co. fTrevillet, Cornwall, UK), Comgenex (Princeton, NJ), Brandon Associates (Merrimack, NH), and Microsource (New Milford, CT). A rare chemical library is available from Aldrich Chemical Company, Inc. (Milwaukee, WI). Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available from, for example, Pan Laboratories (Bothell, WA) or MycoSearch (NC), or are readily produced . Additionally, natural and synthetically produced libraries and compounds are readily modified through conventional chemical, physical, and biochemical means fBlondelle et al., 1996). ER-(3 binding assays according to the present invention are advantageous in accommodating many different types of solvents and thus allowing the testing of compounds from many sources.
Compounds identified as ER-(3 agonists or antagonists using the methods of the present invention may be modified to enhance potency, efficacy, uptake, stability, and suitability for use in therapeutic applications, etc. These modifications are achieved and tested using methods well-known in the art.
Examples The present invention is further described by the following examples. The examples are provided solely to illustrate the invention by reference to specific embodiments. These exemplifications, while illustrating certain specific aspects of the invention do not portray the limitations or circumscribe the scope of the invention.
EXAMPLE 1: CONSTRUCTION OF ER-Q RECOMBINANT ADENOVIRUS
The coding sequence of human ER-/3 was obtained as described in co-owned and co-assigned pending US Serial 08/906,365 entitled "Novel Human Estrogen Receptor j3" filed August 5, 1997, and in Bhat et. al., (19981. Essentially, human testis Poly A+ RNA (1 ~.g, Clontech, Palo Alto CA) was mixed with 0.5 pg oligo dT primer (GIBCO-BRL, Gaithersburg MD) in a total volume of 10 p.l. The mixture was heated at 70°C for 10 minutes, and, after cooling on ice, was supplemented with 500 ~M of each deoxynucleoside triphosphate, 1 X cDNA synthesis buffer, and 10 mM DTT
to a final reaction volume of 20 ~.1. The mixture was incubated at 42°C
for 2.5 minutes and then supplemented with 1-2 units reverse transcriptase (GIBCO-BRL, Gaithersburg MD), after which it was incubated at 45°C for minutes and 50°C for 5 minutes. One-tenth of this mixture (approximately 2 p.11 containing the cDNA template was then used in PCR
amplification of ~R-(3 using forward and reverse primers as described below.
5 The PCR primers designated in Serial No. 08/906,365 (supra) were used to amplify ER-~i sequences in a reaction containing the following components: 2 ~I of the cDNA template described above; 1 X PCR buffer;
200 ~M of each deoxynucleoside triphosphate, 2 units of Hot Tub DNA
polymerase (Amersham, Arlington Heights IL), and 1 pg of each of the 10 forward and reverse primers. The reaction mixture was heated to 95°C
for 2 minutes, annealed at 52°C for 1 minute, and amplified using 36 cycles, followed by an incubation at 72°C for 1.5 minutes.
A fragment of approximately 1500 by in length was produced. The fragment digested with Hindlll and Xbal (which cleave at sites present in 15 the forward and reverse primer sequences, respectively, but not in the main body of the amplified cDNA sequence) and cloned into the corresponding sites of the pcDNA3 expression vector (Invitrogen, Carlsbad CA). This asymmetric cloning strategy places the 5' end of ER-(3 cDNA under the control of the viral CMV promoter in pcDNA3. This clone was designated 20 "truncated hER(3" or hER(3T.
To verify the amino terminal and upstream sequence of human ER(3, two independent approaches were taken, as described below.
(1) 10 p.l of a human ovary 5'-Stretch cDNA library (Clontech, Palo Alto CA) was mixed with 50 ul of 1 X K solution ( 1 X PCR
Buffer (GIBCO-BRL, Gaithersburg MD), 2.5 mM MgCl2, 0.5% Tween-20, 100 ~g/ml Proteinase K), and the reaction mixture was incubated at 56°C
for 2 hours, then at 99°C for 10 minutes. Five p.L of this reaction mixture were then used as template in a nested PCR reaction using the PCR primers designated in Serial No. 08/906,365 (supra). The reaction contained 1 X
Klentaq PCR reaction buffer (40 mM Tricine-KOH, 15 mM KOAc, 3.5 mM
Mg(OAc)2, 75 p.g/ml bovine serum albumin); 0.2 p.M of each dNTP; 0.2 pM
10 of each of the above primers, and 1 unit of Klentaq Polymerase Mix (Clontech, Palo Alto CA). Touchdown PCR conditions were as follows: 5 cycles of 94°C for 2 seconds and 72°C for 4 minutes, followed by cycles of 94°C for 2 seconds and 67°C for 3 minutes.
Excess nucleotides and primers were removed from first round PCR
reactions by purification using Wizard PCR columns (Promega, Madison WI). A second-round PCR reaction was then performed using 2 p.l of the purified first-round reaction mixture using the PCR primers designated in Serial No. 08/906,365 (supra). The PCR reaction and cycling conditions were identical to those employed in the first round. The products were 20 cloned into pCR2.1 (Invitrogen, Carlsbad CA ) and three resulting clones were sequenced. All three clones (designated L1, L2, and L3) contained ER(3 inserts of different lengths, all of which were homologous to ER(3 and to each other.
(2) A Marathon Ready thymus cDNA kit (Clontech, Palo Alto CA) for 5' rapid amplification of cDNA ends (RACE) was also used to isolated ER~i 5' clones. In the first round of a nested PCR reaction, 5 ~I of human thymus Marathon-ready cDNA (Clontech, Palo Alto CA) was used as template and using the primers designated in Serial No. 08/906,365 (supra). The PCR reaction and cycling conditions were identical to those described in (1) above.
Excess nucleotides and primers were removed from the first round PCR reactions by purification over Wizard PCR columns (Promega, Palo Alto CA). A second round PCR reaction was performed using 2 ~I of the purified first round reaction using primers designated in Serial No. 08/906,365 (supra). The second round PCR reaction and cycling conditions were identical to those employed in the first round. The products were cloned into the pCR2.1 vector (Invitrogen, Carlsbad CA) and two clones were sequenced. The two clones contain insert sequences of different lengths that are homologous to ER~3, to each other, and to the sequences isolated from a human ovary cDNA library as described above.
All of the ER(3 sequences isolated by methods (1) and (2) above contained 110 nucleotides corresponding to hER~iT sequences, as well as 228 additional nucleotides at the 5' end.
The hER(3T and 5' end sequences were joined and the resulting cDNA was cloned into pCDNA3 (Invitrogen, Carlsbad CA ) under the control of the cytomegalovirus IE promoter; this expression vector was designated "long hER-Vii" or hER(3L . Full length hER~iL cDNA sequence encodes a polypeptide having 530 amino acid residues.
The hERpL sequence contained an optimal translation initiation sequence CCACC immediately upstream to the initiation codon and the sequence was under the control of the cytomegalovirus IE promoter. The coding sequence of hER~iL was then transferred into an Ad50E1 a vector plasmid containing adenovirus sequences from map unit 0-17 with an deletion of E1 a region between map units 1.4 -9.1 (Davis AR et. al., 1985, Gluzman,Y. et. a1..1982). The hERpL transcription unit in Ad50E1 a plasmid contained cytomegalovirus 1 E promoter, Ad5 tripartite leader, coding sequence of hER~ and SV40 late polyadenylation signal sequences. hER~iL in Ad5 DE1 a plasmid was then linearized with BstEll enzyme and transfected along with Clal A fragment of Ad5 virus with E3 region deletion (80-88 map unitsl into 293 cells (transformed primary human embryonic kidney, ATCC CRL 15731. Viral plaques generated by homologous recombination were isolated, amplified and characterized by restriction DNA analysis and cell lysis assay in A549 cells (human lung carcinoma, ATCC CCL 185). Confirmatory tests indicated that the recombinant Ad5 hER~3L virus contained the expected DNA fragments and was replication defective. The virus was further purified by re-plaguing.
The isolated plaques were amplified, tested and used as a seed stock to generate large amounts of the virus in 293 cells. The virus was titered in 293 cells by plaque assay and the stock contained 1.28 X10 9 PFU/ml.
AMENDED SHEET
EXAMPLE 2: ASSESSMENT OF ENDOGENOUS LEVELS OF ER MRNA IN SAOS-2 Unless otherwise noted, cell culture reagents were obtained from Gibco BRL (Gaithersburg MD). LNCaPLN3 cells were grown in RPMI 1640 medium supplemented with 10% FBS, 2 mM GIutaMAX-1, 100 U/ml penicillin g, and 100 Nglml streptomycin sulfate. Saos-2 cells (ATCC, Manassas VA) were maintained in monolayer culture using McCoy's 5A
medium supplemented with 10% fetal bovine serum (FBS), 2 mM
GIutaMAX-1, 100 U/ml penicillin g, 100 Ng/ml streptomycin sulfate.
RT-PC R
Total RNA was isolated from LNCaPLN3 and Saos-2 cells using TRlzol (Gibco BRL, Gaithersburg MD) according to the manufacturer's directions. The samples were then treated with RNase-free DNase I
(Gibco BRL, Gaithersburg MD1 at 1 unitlNg for 30 minutes at 37°C. RNA
was purified from the reaction using RNeasy columns (Qiagen, Hilden Germany) and amount recovered estimated by UV spectrophotometry.
Reverse transcription reactions were performed on 0.5 Ng of RNA
in a 20 NI reaction. For ER-a the reaction contained 1 x PCR Buffer (Gibco BRL, Gaithersburg MD), 5 mM MgCl2, 1.25 NM ERa-specific reverse primer (5'-CCAGCAGCATGTCGAAGATC-3', SEQ ID No. 1 ), 0.5 NM
GAPDH-specific reverse primer (5'-CACCCTGTTGCTGTAGCCAAATTC-3', SEQ ID No. 21, 0.5 mM dNTPs, 20 units of RNasin (Promega, Madison WI) and 200 units of Superscript II Reverse Transcriptase (Gibco BRL, AMENDED SHEET

Gaithersburg MD). The ER-~3 reaction contained the same components as ER-a with the following exceptions: 2.5 mM MgCl2 and 1.25 NM ER~i-specific reverse primer (5'-GCAGAAGTGAGCATCCCTCTTTG-3', SEQ ID
No. 31. A duplicate reaction which was identical in all reagents except 5 that it did not contain Superscript II Reverse Transcriptase was performed for each sample as a negative control to ensure that the RNA samples were not contaminated by DNA. Reactions were incubated at 42°C for 15 minutes followed by 5 minutes at 99°C and 5 minutes on ice prior to amplification.
10 PCR was initiated by adding 80Ni of master mix containing ER-a-specific forward primer (5'-GGAGACATGAGAGCTGCCAAC-3', SEQ ID
No. 4) or ER-(3-specific forward primer (5'-CAGCATTCCCAGCAATGT
CAC-3', SEQ ID No. 5) and GAPDH-specific forward primer (5'-GACATCAAGAAGGTGGTGAAGCAG-3', SEQ ID No. 6) directly to the 15 20NI reverse transcriptase reaction. Final concentration of reagents in the ER-a 100p1 PCR reaction was as follows: 0.25 NM each ER-specific primer, 0.1NM each GAPDH primer, 1 x PCR Buffer (Gibco BRL, Gaithersburg MD), 0.2 mM dNTPs, 2 mM MgCl2 and 0.5 units of Taq DNA Palymerase (Gibco BRL, Gaithersburg MD1. The ER-(3 reaction 20 contained the same amount of reagents except for 1 mM MgCl2. Two step PCR was carried out in a PE 9600 for 25 cycles as follows: 95°C
for sec, 64°C for 1.5 min. Samples were incubated at 64°C for 10 min after amplification.
AMENDED SHEET

Twenty microliters of each sample were separated using a 1.5%
agarose gel and transferred to Hybond-N + (Amersham Pharmacia, Piscataway NJ) by capillary alkali Southern Blotting in 0.4 N NaOH, 0.6M
NaCI. Blots were pre-hybridized at 42°C for 30 min in Rapid-Hyb buffer (Amersham Pharmacia, Piscataway NJ). Oligonucleotide probes specific for ER-a (5'-TGAACCAGCTCCCTGTCTGCCAGGTTGGT-3', SEQ ID No.
7), ER-p l5'-CCGCATACAGATGTGATAACTGGCGATGGA-3', SEQ ID No.
8) and GAPDH (5'-GCTGTTGAAGTCACAGGAGACAACCTGGT-3', SEQ ID
No. 9) fragments were end-labeled with 32P-y ATP using Polynucleotide Kinase (Gibco BRL, Gaithersburg MD). Probes were added to the blot at 3.0 x 105 CPM/ml and incubated at 42°C for 1 hour. ER and GAPDH
hybridizations were done independently. Blots were washed once in 2x SSC, 0.1 % SDS at room temperature for 15 min then twice in 0.2x SSC, 0.1 % SDS at 42°C for 15 min. Blots were then exposed to film.
Saos-2 cells expressed endogenous ER-~i but not ER-a mRNA when assessed by PCR (Figure 11. As a positive control, GAPDH mRNA was coamplified in these reactions. Whereas both cell lines contained GAPDH
mRNA, only the Saos-2 cells contained ER-Vii. Similar results were obtained for the LNCaPLN3 cells (data not shownl.
AMENDED SHEET

EXAMPLE 3: DISCOVERY THAT ER:f3 UPREGULATES MT-II- IN SAGS-2 AND

Cell culture and infection Because message levels for ER-(3 were iow, Saos-2 and LNCaPLN3 cells were engineered to overexpress hER~3L by transient infection with a recombinant adenovirus (see Example 11 prior to differential display.
LNCaPLN3 and Saos-2 cells were cultured as described above.
Sixteen hours prior to infection, the cells were plated in phenol red-free RPMI 1640 medium supplemented with 10% charcoal/dextran-treated ("stripped") FBS (HyClone, Logan UT1, 2 mM GIutaMAX-1, 100 U/ml penicillin g, 100 Ng/ml streptomycin sulfate. This medium was used for the remainder of the experiment.
Cells were infected with a 1 /20 dilution of an Ad5 hER(3L virus (see Example 1 ) using 2% stripped FBS phenol red-free medium with antibiotics and GIutaMAX-1 for 2 hours at 37° C. Medium containing virus was aspirated and the cells were washed with medium. Fresh medium was added and the cells allowed to recover overnight at 37°C.
The following day, cells were treated with 10 nM 173-estradiol or vehicle for 24 hours and total RNA prepared for differential display using the TRlzol reagent (Gibco BRL, Gaithersburg MD). To remove residual DNA, samples were treated with RNase-tree DNase I (Gibco BRL, Gaithersburg MD) at 1 unit/Ng for 30 minutes at 37°C. RNA was purified from the reaction using RNeasy columns (Qiagen, Hilden Germany) and amount recovered estimated by UV spectrophotometry.

Rapid Analysis of Differential Expression (RADE) Reverse transcription (RT) Following DNase I treatment, six micrograms total RNA was incubated with 1 x RT buffer 125 mM Tris-CI, pH 8.3, 37.6 mM KCI, 3 mM
MgCl2 and 5 mM DTT, from Genhunter, Nashville TN), 20 NM dNTP's (dA, C, G and TTP 2'- deoxynucleoside 5' triphosphates, From Gibco BRL
(Gaithersburg MD), 0.2 NM HT"C (oligonucleotide AAGCTTTTTTTTTTTC, SEQ ID No. 15) in a final volume of 600 NL. This reaction mixture was incubated at 65°C for five minutes to denature secondary structures, followed by a ten minute incubation at 37°C. At this time 30 NI
Superscript II reverse transcriptase (200U/NI, Gibco BRL, Gaithersburg MD) was added to the reaction and incubation proceeded for 1 hr at 37°C. The enzyme was inactivated by heating at 75°C for five minutes.
An aliquot of this reaction was then used for the second strand synthesis by PCR.
Polymerase chain reaction fPCRJ
To 2NI of the RT reaction was added, 1 x PCR buffer ( 10 mM Tris-CI, pH 8.4, 100 mM KCI, 1.5 mM MgClz and 0.001 % gelatin), 2 NM
dNTP's, 15 nM "P dATP (NEN, Boston MA), 1 unit AmpIiTaq DNA
polymerase iPerkin-Elmer, Norwalk CT) and 1NM arbitrary primer 5'AAGCTTGCCATGG-3' for a total reaction volume of 20NI. This reaction mixture was then thermocycled using the following parameters:
92°C for 2 min, 1 cycle AMENDED SHEET

WO 00/3?681 PCT/US99/29856 92°C for 15 sec, 40°C for 2 min, 72°C for 30 sec, 40 cycles 72°C for 5 min Gel alectrophoresis Duplicate samples of PCR products were separated by gel electrophoresis on a 6% denaturing polyacrylamide gel (5.7% acrylamide, 0.3% bisacrylamide, 42% urea and 51 % H20) in 1 x TBE buffer (0.1 M
Tris, 0.09 M Boric Acid, 1 mM EDTA) for three hours at 2000 volts. The gel was then transferred to fitter paper (Schleicher & Schuell, Keene NH), dried under vacuum at 80°C for one hour and exposed to X-ray film for twenty four hours.
One band, designated 6a, was apparently upregulated in both the Saos-2 and LNCaPLN3 cells by 17-~3 estradiol (Figure 2). The developed film was then superimposed over the dried gel and the band's corners were marked using a 22 gauge syringe needle. The gel slice within these boundaries excised with a razor blade and immersed in 100 NI HzO. This sample was boiled in a water bath for fifteen minutes, centrifuged for two minutes and the supernatant solution transferred to a new tube. Added to this sample was 5 NI of 10 mg/ml glycogen, 10NI of 3 M sodium acetate and 450,u1 of 100% ethanol. The sample was mixed, allowed to precipitate overnight at -20°C and centrifuged for ten minutes at 10 g. The supernatant solution was removed, the pellet washed with 200 NI
of 85% ethanol, dried and resuspended in 10 NI H20. A 3 NI aliquot was used in a reamplification PCR reaction in the presence of 1 x PCR buffer, 20 NM dNTP's, 0.2 NM arbitrary primer and 0.2 NM oligonucleotide HT"C
and 2 units AmpIiTaq polymerase using the same cycling parameters as the PCR reaction above. The resulting product was then used as probe in 5 a Northern hybridization assay to confirm regulation and cloned into a bacterial piasmid for sequence analysis.
Fragment cloning Band 6a was cloned using the TA cloning kit (Invitrogen, Carlsbad CA). After lysis, colonies were screened by PCR for the correct insert 10 size. Colonies were lysed in 20 mM Tris-HCI, pH $, 50 mM KCI, 2.5 mM
MgCl2, 0.5% Tween 20 and 100,ug/ml Proteinase K by incubating for 30 min at 56°C then 10 min at 99°C to inactivate the Proteinase K.
Of this reaction 2,u1 was used in a PCR reaction of 20 mM Tris-HCI, pH $, 50 mM
KCI, 2.5 mM MgCl2, 75 NM dNTPs, 375 nM M-13 forward. and reverse 15 primers and 2.5 U of Taq polymerase (Gibco BRL, Gaithersburg MD). The reactions were cycled as follows: 95°C for 30 sec; 64°C for 30 sec;
72°C for 45 sec for 30 cycles. One clone, designated 6a.2, was chosen for sequencing.
Fragment sequencing 20 Clone 6a.2 was sequenced according to the ABI PRISM"" Dye Terminator Cycle Sequencing Ready Reaction Kit with AmpIiTaq DNA
Polymerase using the recommended protocol from Applied Biosystems (Foster City CA). Spin columns (AGTC) were employed to remove unincorporated dye-labeled nucleotides after cycle sequencing. Automated DNA sequencing grade 4.75% polyacrylamide gels were run for all the DNA sequencing samples using ABI 373 DNA sequencers. Sequencing data was edited using Sequence Navigator and assembled using DNAStar (DNAStar, Madison WI). The sequence was trimmed of BADE primers and used in a BLAST search using Millennium Software (Boston MA). A
nucleotide homology search of clone 6a.2 sequence revealed a 98 identity with human MT-II-. Over the coding sequence, however, there were no amino acid mismatches (Figure 3) Confirmation of regulation using Northern blots and 4uantitative reverse transcriptase c~olymerase chain reaction Ic~RT-PCR1:
To confirm the results obtained with the PCR amplification of cellular mRNAs, Northern blots and qRT-PCR were used to assess the effect of 17-(3 estradiol or other compounds on message levels. Unless otherwise noted, all experiments use cells transiently overexpressing hER~3L in response to adenovirus infection as described above, and were treated with compound for 24 hours. In some cases cells transiently overexpressing ER-a were used for comparison. Methods for adenovirus-mediated overexpression of ER-a are identical to those described for ER-(3.
Northern blots Poly A+ RNA was isolated from total RNA using Oligotex mRNA
Isolation kit (Qiagen, Hilden Germany) according to manufacturers instructions. Six micrograms of mRNA or 10 pgs of total RNA were separated on a 1.5% agarose, 0.22 M formaldehyde, 10 mM HEPES, 1 mM EDTA gel. RNA was transferred to Hybond-N (Amersham Pharmacia, Piscataway NJ) by capillary action in 20X SSC (Gibco BRL, Gaithersburg MD ) overnight. After transfer, the membrane was UV-cross-linked and dried at 80°C for 10 min. Northern Blots were pre-hybridized in Rapid Hyb solution (Amersham Pharmacia, Piscataway NJ) for 30 minutes.
The insert from clone 6a.2 was isolated from the plasmid using PCR as described above for colony screen. The PCR product was purified from an agarose gel using Wizard Preps fPromega, Madison WI). Re-amplified RADE fragments for band 6a or fragment from clone 6a.2 were random primer labeled using Redi-prime kit (Amersham Pharmacia, Piscataway NJ) according to manufacturers instructions. Unincorporated nucleotides were removed using a Nap-5 column (Amersham Pharmacia, Piscataway NJ) and incorporation of [32P]-dCTP measured by liquid scintillation counting.
The probes were denatured at 100°C for 10 min and 1.5 x 106 CPM/ml of Rapid-Hyb hybridization solution was added to the membrane.
The blots were hybridized at 65°C for 5 hours and were washed as follows: Once in 2X SSC, 0.1 % SDS at 65° C for 15 min; twice in 0.2X
SSC, 0.1 % SDS at 65°C for 15-30 min. Blots were exposed to film and to a Phosphorlmager screen (Molecular Dynamics, Sunnyvale CA). After probing with 6a RADE fragment or 6a.2 cloned fragment, blots were probed with a cDNA homologous to GAPDH as above. Hybridization signal from 6a.2 fragment was normalized to that of GAPDH on a Phosphorlmager (Molecular Dynamics, Sunnyvale CA) to determine fold induction.
QRT PCR - A MT-II fragment identical to clone 6a.2 except that it contained a 63 by deletion was subcioned into pcDNA.3 (lnvitrogen, Carlsbad CA). RNA was transcribed using the T7 Promoter Large Scale Transcription Kit (Novagen, Madison Wil. After phenol-chloroform extraction and ethanol precipitation the synthesized RNA was quantified and analyzed using UV spectrophotometry and gel electrophoresis.
Reverse transcription reactions were performed on 200 ng and 300 ng of DNased Saos-2 total RNA plus a known amount of MT-II standard RNA in a 20 NI reaction. The reaction contained 1 x PCR Buffer (Gibco BRA, Gaithersburg MD1. 3.75 mM MgCl2, 1.25 NM MT-II-specific reverse primer (5'-GGAATATAGCAAACGGTCAGGGTC-3', SECT ID No. 101, 0.5 mM dNTPs. 1 mM DTT, 20 units of RNasin (Promega) and 200 units of Superscript II Reverse Transcriptase (Gibco BRL, Gaithersburg MD1.
Reactions were incubated at 42°C for 15 minutes followed by 5 minutes at 99°C and 5 minutes on ice prior to amplification PCR was initiated by adding 80NI of master mix containing MT-II
specific forward primer (5'-GGCTCCTGCAAATGCAAAGAG-3', SECT ID
No. 11) directly to the 20NI reverse transcriptase reaction. Final concentration of reagents in the 100NI PCR reaction was as follows: 0.25 NM each MT-II -specific AMENDED SHEET

WO 00/376$1 PCT/US99/29856 primer, 1 x PCR Buffer (Gibco BRL, Gaithersburg MD1, 0.1 mM dNTPs, 1.5 mM MgCl2 and 0.5 units of Taq DNA Polymerase (Gibco BRL, Gaithersburg MD). Two step PCR was carried out in a PE 9600 (Perkin-Elmer, Norwalk CT) for 40 cycles as follows: 95°C for 30 sec, 64°C for 5 1.5 min. Samples were incubated at 64°C for 10 min after amplification.
PCR products were separated and analyzed on a reverse-phase ion-pair high performance liquid chromatography DNASep column (Sarasep, San Jose CA). The elution system was a gradient of acetonitrile in 0.1 M
triethylammonium acetate (Fluka, Ronkonkoma NY) at a flow rate of 0.7 ml/min. The acetonitrile gradient increased from 14.6% to 16.6°~ over 5 minutes. The amount of product from the standard and the native RNA
was determined by UV absorbance detection at 254 nm and signal was analyzed by an on-line integrator. From the chromatograms, the ratio of the area under each peak was used to determine the ratio of the amount 15 of input MT-II standard RNA to the amount of native MT-II message in the Saos-2 RNA.
In LNCaPLN3 cells, the magnitude of MT-II induction by 17-(3 estradiol is approximately 6 fold (Figure 4). In Saos-2 cells, 10 nM 17(3-estradiol upregulates MT-II mRNA as much as 14 fold (Figure 4). This 20 upregulation in Saos-2 cells has been repeated in at least 20 experiments and the range of induction is 3.5-14 fold. The EC5° of this regulation is 4.6 t 2.7 nM as assessed by qRT-PCR (Figure 51. If Saos-2 cells are treated with 10 nM 17(3-estradiol and RNA prepared at different times post treatment, the first discernible increase in MT-II message occurs after 8 hours and the response peaks by 24 hours (Figure 61.
Since the samples prepared for differential display overexpressed hER(3L, , MT-II regulation was assessed in native Saos-2 cells and those 5 overexpressing ERa. As illustrated in Figure 7 , endogenous levels of ER-~3 or overexpressed levels of ER-a were ineffective at mediating 17(3-estradiol's regulation of MT-II.
Other compounds were tested for their ability to upregulate MT-II
in Saos-2 cells. Ten nanomolar diethylstilbesterol (Sigma, St. Louis MO) 10 and 0.1 pM genistein (RBI, Natick MAI both increased MT-II mRNA. One micromolar of the antiestrogen, ICI-182780 (Zeneca, Wilmington DE), completely blocked induction by 17(3-estradiol and genistein but had no effect when given alone (Figure 8). Co-treatment with 1 uM of the antiprogesterone/antiglucocorticoid, RU486 (Ligand Pharmaceuticals, La 15 Jolla CA), did not block regulation by 17(3-estradiol (data not shownl.
EXAMPLE 4: TREATMENT OF SAOS-2 CELLS WITH CYCLOHEXIMIDE
To determine if the increase in MT-II mRNA requires new protein synthesis, cycloheximide was used after hER(3L overexpression and 20 before treatment with 17-(3 estradiol to arrest translation.
Verification of cycloheximide's effect on protein synthesis Cells were plated and infected with hER(3L virus as described above, then pre-treated with 10 ~g/mL of cycloheximide or vehicle for one WO 00/37b81 PCT/US99/29856 hour at 37°C. After this preincubation, 10 pg/mL of cycloheximide, 10 nM 17-(3 estradiol and 50 ~Ci/ml 35S-Methionine (NEN, Boston MA) in methionine-deficient media was added and incubation continued at 37°C
for 8 or 24 hours.Cells were washed 3x in cold PBS and then scraped off plates in 500 ~.I of PBS. . Cells were pelleted and resuspended in 200 ~I of RIPA buffer (150 mM NaCI, 1 % NP-40, 0.5% DOC, 0.1 % SDS, and 50 mM Tris, pH 8.0). Methionine incorporation was measured by TCA
precipitation. Five and ten microliters of each sample were spotted on individual Whatman filters. Filters were boiled in 10% trichloroacetic acid for 10 minutes, then washed 3 times for 10 minutes in deionized water, 3 times for 10 minutes in 95% ethanol and once for 10 minutes in acetone.
The dried filters were placed in scintillation fluid and counted for 1 minute.
Treatment of cells with 17-~i estradiol Cells were plated and infected with hER[3L virus as described above. Cells were pretreated with 10 ug/mL of cycloheximide or vehicle for one hour at 37°C. After this preincubation, 10 nM 17-~i estradiol or vehicle was added and incubation continued at 37°C for 8 or 24 hours.
RNA was isolated from the cells as described above and gene expression evaluated by Northern blot analysisor qRT-PCR as described in Example 3.
Verification of functional ER-(3 protein after cvcloheximide treatment Duplicate cultures were prepared and treated as described in the previous section. After the 8 hour incubation, cells were washed four times with DMEM to remove 17-(3 estradiol. Cycloheximide (10 ug/mL) was added to all samples as was 1 nM (3H]-estradiol. Some cells were co-treated with 0.3 ~,M diethylstilbesterol to estimate nonspecific binding.
After incubation for 2.5 hr at 37°C, cells were washed with DMEM
and lysed with 0.1 % sodium dodecyl sulfate. DPM were measured by liquid scintillation counting.
The MT promoter contains glucocorticoid and metal response elements, but EREs have not been described. It is possible the effect of 17-(3 estradiol on MT-II mRNA expression is indirect. After overexpression of ER-(3, Saos-2 cells were treated with cycloheximide to severely limit new protein synthesis (Figure 9A1. Although comparable amounts of receptor protein were expressed with and without cycloheximide treatment as measured by a whole cell binding assay (Figure 9B), MT-II induction did not occur (Figure 9C,D).
EXAMPLE 5 : TREATMENT OF RATS WITH 17-Q ESTRADIOL
Because 17-(3 estradiol upregulates MT-II in LNCaPLN3 cells, a prostate cancer cell line, we looked for a similar response in the rat prostate.
All animals were treated according to institutional guidelines using approved protocols. Adult (15-19 week, '375g) castrated Sprague-Dawley rats were purchased from Taconic Farms (Germantown NY). Ten days after castration, rats were injected with vehicle, 16 ~.g 17-(3 estradiol, 16 wg diethylstilbesterol (Sigma, St. Louis MO), or 16 ~g 17-(3 estradiol plus 1.6 mg raloxifene (synthesized in-house) subcutaneously once per day for three days. Approximately 24 hr after the last dose, rats were euthanized by COZ asphyxiation and the prostate excised. Total RNA was prepared and analyzed for MT-II mRNA as described above. In another experiment, rats were dosed for 1, 2 or 3 days with 16 ~.g 17-(i estradiol before prostate tissue was collected.
Metallothionein-II mRNA increased five-fold after 17-~i estradiol treatment. A similar response occurred when rats were dosed with diethylstilbesterol, but this upregulation was blocked by coadministration with a 100 fold excess of raloxifene hydrochloride, an estrogen antagonist (Figure 10). Although the initial experiments used tissue from rats dosed for three days, the upregulation of MT-II in the rat prostate is first seen after two days of dosing with 17-(3 estradiol (Figure 11 ).
EXAMPLE 6 : ERE-LUCIFERASE REPORTER ASSAY USING MCF-7 CELLS
Because MT-II regulation in Saos-2 cells is mediated by ER-(3 and not ER-a, another assay was needed to compare selectivity of compounds for these two receptors in cell based assays. MCF-7 is an estrogen-responsive breast cancer cell line and, in our hands, expresses only ER-a as measured by RT-PCR (data not shown). When MCF-7 cells are transiently infected with an ERE--reporter gene construct and treated with 17-~i estradiol, reporter gene activity can be measured.
MCF-7 HTB 22, ATCC , Manassas VA) cells are passaged twice a week with growth medium [D-MEM/F-12 medium containing 10% (vlv) heat-inactivated fetal bovine serum100 U/ml penicillin g, 100 p.g/ml streptomycin sulfate, 2 mM GIutaMAX-1 ]. The cells are maintained in vented flasks at 37oC inside a 5% C02/95% humidified air incubator.
One day prior to treatment, the cells are plated with growth medium at 25,000/well into 96 well plates and incubated at 37oC overnight.
The cells are infected for 2 hr at 37°C with 50 p.l/well of a 1:10 dilution of adenovirus 5-ERE-TK-luciferase (Bodine et. al., 1997) in experimental medium (phenol red-free DMEM/F-12 medium containing 10% (v/v) heat-inactived charcoal-stripped fetal bovine serum, 100 U/ml penicillin g, 100 wg/ml streptomycin sulfate, 2 mM GIutaMAX-1 , 1 mM
sodium pyruvatel. The wells are then washed once with 150 wl of experimental medium. Finally, the cells are treated for 24 hr at 37oC in replicates of 8 wells/treatment with 150 ~I/well of vehicle ( < 0.1 % v/v DMSO) or compound that is diluted > 1000-fold into experimental medium.
After treatment, the cells are lysed on a shaker for 15 min with 25 ~,I/well of 1 X cell culture lysis reagent (Promega, Madison WI). The cell lysates (20 ~1) are transferred to a 96 well luminometer plate, and luciferase activity is measured in a MicroLumat LB 96 P luminometer (EG
& G Berthold, Bad Wildbad Germany) using 100 wl/well of luciferase substrate (Promega, Madison WI). Prior to the injection of substrate, a 1 second background measurement is made for each well. Following the 5 injection of substrate, luciferase activity is measured for 10 seconds after a 1 second delay. After subtracting background subtracts the mean and standard deviation are calculated.
Compounds which induce MT-II in Saos-2 cells and do not stimulate reporter gene activity in MCF-7 cells are thus selective for ER-[3.
10 In addition, results from these two assays can also be used to select a compound that is selective for ER-a as shown in Figure 12.
Discussion Using differential display, it was unexpectedly discovered that ER-~i 15 increases MT-II mRNA in two cell lines treated with 17-~i estradiol. To our knowledge, this is the first gene discovered to be regulated by this new form of the ER. This response has been extensively characterized in the human osteosarcoma cell line Saos-2. A variety of estrogens can upregulate MT-II, and this response is blocked by cotreatment with the 20 estrogen antagonist 1CI-182780. The ECSO for 17-/3 estradiol is approximately 5 nM and this response is mediated by ER-~i acting through as yet unknown proteins.

The action of ER-(3 on MT-II is not a general phenomenon as a survey of over a dozen cell lines failed to reveal this induction in samples other than Saos-2 and LNCaPLN3 cells. However, the fact that MT-II is regulated by 17-[3 estradiol in the rat prostate strengthens the possibility that this is a physiologically relevant response and not a artifact of receptor overexpression in cancer cell lines. We are confident that this prostate response is mediated by the ER for two reasons: A nonsteroidal estrogen (diethylstilbesterol) upregulates MT-II and raloxifene, an estrogen agonist/antagonist, blocks 17-~3 estradiol's action. However, at this time it is not clear if this induction reflects activity of ER-(3 and/or ER-a.
Although ER-~3 is the predominant form of the ER in the rat prostate, ER-a is also present. Current studies are underway to define the receptor type responsible for this in vivo response.
Metallothioneins are low molecular weight, cysteine rich proteins that bind metals such as cadmium, copper and zinc. Although the first metallothionein was discovered over forty years ago (Vallee, 1957), debate continues as to their function. Several proposals have been made and these include protection form metal toxicity, regulation of zinc and copper homeostasis and defense against oxidative stress. Regulation of energy balance has also been implicated because, after reaching sexual maturity, MT (-I and -1I) knockout mice become obese (Beattie et al, 1998).

Recently, studies have detailed how MT may act to regulate zinc homeostasis in cells. Using purified zinc-dependent enzymes such as E.
coli alkaline phosphatase, bovine carboxypeptidase A and sheep sorbitol dehydrogenase two recent publications show how agents in the cellular milieu can facilitate exchange between zinc complexed with MT and (metal depleted) apoenzymes (Jacob et al, 1998; Jiang et al, 1998).
Citrate and glutathione can influence the direction of zinc transfer and thus regulate enzyme activity depending on the redox state of the cell.
Although not an enzyme, the ER also requires zinc for activity, and ER
from MCF-7 cells can reversibly exchange zinc with purified MT in vitro (Cano-Gauci et al, 19961.
A myriad of agents can regulate MT levels, including glucocorticoids and metals such as cadmium (for review see Hamer, 19861. Estrogens are not a classical regulators of MT, but two intriguing papers appear in the literature. First, a two week treatment of female rats with 17-(3 estradiol upregulated a copper binding protein in intestinal mucosa which reduced the amount of copper absorbed into the plasma.
Since the molecular weight of this protein was about 10K, the authors suggest it is MT (Cohen et al, 19791. Recently, MT was identified in a subtractive hybridization screen of uterine mRNAs regulated after a single injection of 17a ethinyl estradiol Rivera-Gonzalez et al, 1998). Although the isoform is not identified, a MT transcript increased three fold between four and eight hours after 17-a ethinyl estradiol stimulation. In addition, it is not clear which receptor type effects this regulation because ER-a, not ER-(3, is the most abundant ER in the uterus (Kuiper at al, 1996;
Couse et al, 1997).
Since the function of MT is only beginning to be elucidated and is completely unknown for ER-~3, understanding the significance of their association is impossible at this time. However, even if the connection between these two proteins is unclear, the observation of regulation can still be exploited to help design an ER-p or ER-a specific ligand. The fact that genistein upregulates MT-II- in Saos-2 cells as well or better than 17~i-estradiol indicates an ER-p-selective compound can effectively direct transcription. In addition, several structurally diverse compounds which bind to ER-(3 can also upregulate this message in Saos-2 cells. When data on a compound's ability to regulate MT-II in Saos-2 cells are coupled with information about how it regulates reporter gene activity in an MCF-7 cell, 15 ER-(3 and ER-a selectivity can be assessed. Thus, when used together, these two assays are tools to help design selective compounds for either ER type. Finally, if the regulation in the prostate can also be shown to be mediated by ER-(3, then the in vivo activity of compounds can be assessed, providing another valuable tool for drug discovery.

REFERENCES
Beattie JH, Wood AM, Newman AM, Bremner I, Choo KHA, Michalska AE, Duncan JS, Trayhurn P. 1998. Obesity and hyperleptinemia in metallothionein (-I and -II? null mice. Proc Natl Acad Sci (USA) 95:358-363.
Bhat RA, Harnish DC, Stevis PE, Lyttle CR, Komm BS. 1998. A novel human estrogen receptor ~3: Identification and functional analysis of additional N-terminal amino acids. J Steroid Molec Biol, in press.
Blondelle SE, Houghten RA. 1996. Novel antimicrobial compounds identified using synthetic combinatorial library technology. Trends Biotechnol 14(21:60-65.
Bodine PVN, Green J, Harris HA, Bhat R, Stein GS, Lian JB, Komm B.
1997. Functional properties of a conditionally phenotypic, estrogen-responsive, human osteoblast cell line. J Cell Biochem 65:368-387.
Cano-Gauci DF, Sarkar B. 1996. Reversible exchange between metallothionein and the estrogen receptor zinc finger. FEBS Letters 386:1-4.

Cohen DI, Illowsky B, Linder MC. 1979. Altered copper absorption in tumor-bearing and estrogen-treated rats. Am J Physiol 23613):E309-15.
Couse JF, Lindzey J, Grandien K, Gustafsson J, Korach, KS. Tissue 5 distribution and quantitative analysis of estrogen receptor-a (ERa) and estrogen receptor-(3 (ERp) messenger ribonucleic acid in the wild type and ERa-knockout mouse. 1997. Endocrinology 138(1 1 ):4613-4621.
Cowley SM, Hoare S, Mosselman S, Parker MG. 1997. Estrogen 10 receptors a and (3 form heterodimers on DNA. J Biol Chem 272(32):19858-19862.
Davis AR, Kostek B, Mason B, Hsiao CL, Morin J, Dheer SK, Hung P.
1985. Expression of hepatitis B surface antigen with a recombinant 15 adenovirus. Proc Natl Acad Sci (USA) 82:7560-7564.
Dotzlaw H, Leygue E, Watson PH, Murphy LC. 1996. Expression of estrogen receptor-beta in human breast tumors. J Clin Endocrinol Metab 82(7):2371-2374.
Enmark E, Pelto-Huikko M, Grandien K, Lagercrantz S, Lagercrantz J, Fried G, Nordenskjold M, Gustafsson J. 1997. Human estrogen receptor-(3 gene structure, chromosomal localization, and expression pattern. J Clin Endocrinol Metab 82(12):4258-4265 Green S, Kumar V, Walter KP, Chambon P. 1986. Structural and 5 functional domains of the estrogen receptor. Cold Spring Harbor Symposia on Quantitative Biology LI:751-758.
Gluzman Y, Reichl H, Solnick D. 1982. Helper-free adenovirus type-5 vectors. In Eukaryotic Viral Vectors, Y Gluzman ed., (Cold Spring Harbor Laboratory), pp 187, 192.
Hamer DH. 1988. Metallothionein. Ann Rev Biochem 55:913-951.
lafrati MD, Karas RH, Aronovitz M, Kim S, Sullivan TR, Lubahn DB, O'Donnell Jr. TF, Korach KS, Mendelsohn ME. 1997. Estrogen inhibits the vascular injury response in estrogen receptor-a-deficient mice. Nature Med 3(5):545-548.
Jacob C, Maret W, Vallee BL. 1998. Control of zinc transfer between thionein, metallothionein and zinc proteins. Proc Natl Acad Sci (USA) 95:3489-3494.

WO 00/3?681 PCT/US99/29856 Jiang L, Maret W, Valee BL. 1998. The glutathione redox couple modulates zinc transfer from metallothionein to zinc-depleted sorbitol dehydrogenase. Proc Natl Acad Sci (USA) 95:3483-3488.
Kuiper GGJM, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA.
1996. Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Proc Natl Acad Sci (USA) 93:5925-5930.Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson, JA. 1997.
Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors a and (3. Endocrinology 138(3):863-870.
Lau K, Leav I, Ho S. 1998. Rat estrogen receptor-a and -(3, and progesterone receptor mRNA expression in various prostatic lobes and microdissected normal and dysplastic epithelial tissues of the Noble rats.
Endocrinology 139( 1 ):424-427.
Lobo R. 1995. Benefits and risks of estrogen replacement therapy. Am J
Obstet Gynecol 173(3):982-989.
Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach, KS. 1993.
Alteration of reproductive function but not prenatal sexual development WO 00/37681 PCT/US99/29$56 after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci (USA) 90:11162-11166.
Mosselman S, Polman J, Dijkema R. 1996. ER(3: identification and characterization of a novel human estrogen receptor. FEBS Letters 392:49-53.
Ogawa S, Inoue S, Watanabe T, Hiroi H, Orimo A, Hosoi T, Ouchi Y, Muramatsu M. 1998. The complete primary structure of human estrogen 10 receptor (3 IhER(3) and its heterodimerization with ER a in vivo and in vitro.
Biochem Biophys Res Comm 243:122-126.
Pan LC, Ke HZ, Simmons HA, Crawford DT, Chidsey-Fink KL, McCurdy SP, Schafer JR, Kimbro KS, Taki M, Korach KS, Thompson DD. Estrogen 15 receptor-alpha knockout (ERKO) mice lose trabecular and cortical bone following ovariectomy. 1997. J Bone Mineral Res 12 (Supplement 1 ):S134.
Pettersson K, Grandien K, Kuiper GGJM, Gustafsson J. 1997. Mouse 20 estrogen receptor [3 forms estrogen response element-binding heterodimers with estrogen receptor-a. Mol Endocrinoi 1 1:1486-1496.

WO 00/37681 PCTlUS99/z9856 Rivera-Gonzalez R, Peterson DN, Tkalcevic G, Thompson DD, Brown TA.
1998. Estrogen-induced genes in the uterus of ovariectomized rats and their regulation by droloxifene and tamoxifen. J Steroid Biochem Molec Biol 64( 1 /2):13-24.
Sagraves R. 1995. Estrogen therapy for postmenopausal symptoms and prevention of osteoporosis. J Clin Pharmacol 35:2s-10s.
Shughrue PJ, Komm B, Merchenthaler I. 1996. The distribution of estrogen receptor-/3 mRNA in the rat hypothalamus. Steroids 61:678-681.
Trembley GB, Tremblay A, Copeland NG, Gilbert DJ, Jenkins NA, Labrie F, Giguere V. 1997. Cloning, chromosomal localization, and functional analysis of the murine estrogen receptor (3. Mol Endocrinol 1 1:353-365.
Vallee BL. 1957. A cadmium protein from equine kidney cortex. J Am Chem Soc 79:4813.
20 Vladusic EA, Hornby AE, Guerra-Vladusic FK, Lupu R. 1998. Expression of estrogen receptor ~i messenger RNA variant in breast cancer. Cancer Res 58:210-214.

Webb P, Lopez GN, Greene GL,. Baxter JD, Kushner PJ. 1992. The limits of the cellular capacity to mediate an estrogen response. Mol Endocrinology. 6(2):157-fi7.

Claims (32)

What is claimed is:
1. A method for screening a test compound that binds to an ER in a receptor binding assay, wherein said method detects ER-.beta. polypeptide-mediated transcription, said method comprising the steps of:
(a) providing a cell which comprises at least one estrogen-regulated DNA sequence encoding MT-II and at least one DNA sequence encoding ER-.beta. polypeptide, wherein said receptor is transcriptionally active;
(b) contacting said cell with either said test compound which binds ER or a control; and (c) detecting the expression of said MT-II, wherein enhanced expression of said MT-II relative to a control indicates that said test compound has estrogen agonist activity.
2. The method of claim 1, wherein the DNA sequence encoding ER-.beta.
polypeptide is incorporated into an adenovirus,
3. The method of claim 2 wherein the adenovirus is a replication-defective Ad5 virus.
4. The method of claim 1, wherein said cells endogenously express ER-.beta. mRNA at a higher rate than ER-.alpha. mRNA.
5. The method of claim 4, wherein said cells are transformed with a recombinant DNA plasmid comprising a polynucleotide encoding a human ER-.beta. operably linked to a suitable promoter, wherein said transformed cells express human ER-.beta. at higher levels than the cells which have not been transformed.
6. The method of claim 1 wherein said cells are Saos-2 or LNCaPLN3 cells.
7. The method of claim 1, wherein said cells do not have a functional ER-.alpha..
8. A method for screening a test compound that binds to an ER in a receptor binding assay, wherein said method detects inhibition of ER-.beta.
polypeptide-mediated transcription, said method comprising the steps of:
(a) providing a cell which comprises at least one estrogen DNA
sequence encoding MT-II and at least one DNA sequence encoding ER-.beta.
polypeptide, wherein said receptor is transcriptionally active;
(b) contacting said cell with one or more estrogens in the presence of the test compound known to bind ER; and (c) detecting the expression of said MT-II, wherein decreased expression of said MT-II relative to the addition of one or more estrogens alone indicates that said test compound has estrogen antagonist activity.
9. The method of claim 8, wherein the DNA encoding ER-.beta.
polypeptide is incorporated into an adenovirus.
10. The method of claim 9 wherein the adenovirus is a replication-defective Ad5 virus.
11. The method of claim 8, wherein said cells endogenously express ER-.beta. mRNA at a higher rate than ER-.alpha. mRNA.
12. The method of claim 11, wherein said cells are transformed with a recombinant DNA plasmid comprising a polynucleotide encoding a human ER-.beta. operably linked to a suitable promoter, wherein said transformed cells express human ER-.beta. at higher levels than the cells which have not been transformed.
13. The method of claim 12 wherein said cells are Saos-2 or LNCaPLN3 cells.
14. The method of claim 13, wherein said cells do not have a functional ER-.alpha..
15. A method of screening test compounds to identify drug candidates which mimic estrogen's effect on ER-.beta.- or ER-.alpha.-mediated transcription, said method comprising the steps of:
(a). contacting said test compound with a plurality of:
(i) first cells comprising at least one endogenous DNA
sequence encoding MT-II and at least one DNA
sequence encoding a ER-.beta. polypeptide, wherein said receptor is transcriptionally active and (ii) second cells comprising an ERE reporter gene construct, wherein said cells express ER-.alpha. polypeptide;
(b) identifying compounds which increase expression of MT-II in said first cells relative to control but have minimal effect on expression of said reporter gene in said second cells, wherein said compounds are considered ER-.beta. selective; or (c) identifying compounds which increase expression of the reporter gene in said second cells relative to control but have minimal effect on expression of MT-II in said first cells, wherein said compounds are considered ER-.alpha. selective.
16. The method of claim 15, wherein the DNA encoding ER-.beta.
polypeptide of said first cells is incorporated into an adenovirus.
17. The method of claim 16 wherein the adenovirus is a replication-defective Ad5 virus.
18. The method of claim 15, wherein said first cells endogenously express ER-.beta. mRNA at a higher rate than ER-.alpha. mRNA.
19. The method of claim 15, wherein said first cells are transformed with a recombinant DNA plasmid comprising a polynucleotide encoding a human ER-.beta. operably linked to a suitable promoter, wherein said transformed first cells express human ER-.beta. at higher levels than said second cells.
20. The method of claim 15 wherein said first cells are Saos-2 or LNCaPLN3 cells.
21. The method of claim 15, wherein said first cells do not have a functional ER-.alpha. and said second cells do not have a functional ER-.beta..
22. The method of claim 15, wherein said second cells were transformed with a DNA polynucleotide comprising an ERE operably linked to a reporter gene.
23. The method of claim 22, wherein said reporter gene is a luciferase gene.
24. A method of screening test compounds to identify drug candidates which inhibit estrogen's effect on ER-.beta.- or ER-.alpha.-mediated transcription, said method comprising the steps of:
(a). contacting said test compound in the presence and absence of one or more estrogens with a plurality of:
(i) first cells comprising at least one endogenous DNA
sequence encoding a metallothionein (MT-II) gene and at least one DNA sequence encoding a ER-.beta.
polypeptide, wherein said receptor is transcriptionally active; and (ii) second cells comprising an ERE-reporter gene construct, wherein said cells express ER-.alpha. polypeptide; and (b) identifying compounds which decrease expression of MT-II in said first cells relative to treatment with one or more estrogens but have minimal effect on expression of said reporter gene in said second cells, wherein said compounds are considered ER-.beta.
selective; or (c) identifying compounds which decrease expression of the reporter gene in said second cells relative to treatment with one or more estrogens but have minimal effect on expression of MT-II in said first cells, wherein said compounds are considered ER-.alpha.
selective.
25. The method of claim 24, wherein the DNA encoding ER-.beta.
polypeptide of said first cells is incorporated into an adenovirus.
26. The method of claim 25 wherein the adenovirus is a replication-defective Ad5 virus.
27. The method of claim 24, wherein said first cells endogenously express ER-.beta. mRNA at a higher rate than ER-.alpha. mRNA.
28. The method of claim 24, wherein said first cells are transformed with a recombinant DNA plasmid comprising a polynucleotide encoding a human ER-.beta. operably linked to a suitable promoter, wherein said transformed first cells express human ER-.beta. at higher levels than said second cells.
29. The method of claim 24 wherein said first cells are Saos-2 or LNCaPLN3 cells.
30. The method of claim 24, wherein said first cells do not have a functional ER-.alpha. and said second cells do not have a functional ER-.beta..
31. The method of claim 24, wherein said second cells were transformed with a DNA polynucleotide comprising an ERE operably linked to a reporter gene.
32. The method of claim 31, wherein said reporter gene is a luciferase gene.
CA002352203A 1998-12-18 1999-12-17 Bioassay for identifying estrogen receptor-.beta./.alpha. selective modulators Abandoned CA2352203A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11279098P 1998-12-18 1998-12-18
US60/112,790 1998-12-18
PCT/US1999/029856 WO2000037681A1 (en) 1998-12-18 1999-12-17 BIOASSAY FOR IDENTIFYING ESTROGEN RECEPTOR-β/α SELECTIVE MODULATORS

Publications (1)

Publication Number Publication Date
CA2352203A1 true CA2352203A1 (en) 2000-06-29

Family

ID=22345864

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002352203A Abandoned CA2352203A1 (en) 1998-12-18 1999-12-17 Bioassay for identifying estrogen receptor-.beta./.alpha. selective modulators

Country Status (9)

Country Link
EP (1) EP1141405A1 (en)
JP (1) JP2002533098A (en)
CN (1) CN1352699A (en)
AU (1) AU760784B2 (en)
CA (1) CA2352203A1 (en)
HU (1) HUP0104719A3 (en)
NO (1) NO20012999L (en)
NZ (1) NZ512347A (en)
WO (1) WO2000037681A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6660468B1 (en) 1998-10-27 2003-12-09 Board Of Trustees Of The University Of Arkansas Vitro and in vivo models for screening compounds to prevent glucocorticoid-induced bone destruction
EP1400810A3 (en) * 1999-04-09 2005-03-02 Karo Bio Ab Estrogen receptors and bone disease
ATE364087T1 (en) 2000-08-08 2007-06-15 Wyeth Corp REGULATION OF EXPRESSION OF EER-7, A MEMBER OF THE LYSYL OXIDASE GENE FAMILY, BY ESTROGEN RECEPTORS
DE10046647A1 (en) * 2000-09-20 2002-03-28 Ludwig Wildt Determining hormonal activity of materials, useful for diagnosis or detecting abuse of anabolics, comprises detecting expression of reporter gene product in transfected cells
ATE313803T1 (en) 2001-11-07 2006-01-15 Schering Ag IN VITRO SCREENING FOR LIGANDS OF THE ESTROGEN RECEPTOR
CN1314966C (en) * 2004-12-03 2007-05-09 湖南农业大学 Detecting method of pig metal sulfur protein
WO2010040446A1 (en) * 2008-10-09 2010-04-15 Merck Patent Gmbh Estrogen and anti-estrogen marker genes
CN109295214A (en) * 2018-11-02 2019-02-01 张磊 Estrogen receptorαgene is related to women eye disease time of origin

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
EP1009807A4 (en) * 1997-09-04 2004-11-03 Univ California DIFFERENTIAL LIGAND ACTIVATION OF ESTROGEN RECEPTORS ER$g(a) AND ER$g(b) AT AP1 SITES

Also Published As

Publication number Publication date
HUP0104719A3 (en) 2004-03-01
CN1352699A (en) 2002-06-05
NO20012999D0 (en) 2001-06-15
HUP0104719A2 (en) 2002-04-29
NO20012999L (en) 2001-08-15
NZ512347A (en) 2003-12-19
JP2002533098A (en) 2002-10-08
AU2187900A (en) 2000-07-12
EP1141405A1 (en) 2001-10-10
WO2000037681A1 (en) 2000-06-29
AU760784B2 (en) 2003-05-22

Similar Documents

Publication Publication Date Title
Levenson et al. The key to the antiestrogenic mechanism of raloxifene is amino acid 351 (aspartate) in the estrogen receptor
Nakshatri et al. Constitutive activation of NF-κB during progression of breast cancer to hormone-independent growth
Young Oestrogen and progesterone action on endometrium: a translational approach to understanding endometrial receptivity
Aronica et al. Stimulation of estrogen receptor-mediated transcription and alteration in the phosphorylation state of the rat uterine estrogen receptor by estrogen, cyclic adenosine monophosphate, and insulin-like growth factor-I.
Thomas et al. Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells
Hyder et al. Identification of functional estrogen response elements in the gene coding for the potent angiogenic factor vascular endothelial growth factor
Katzenellenbogen et al. Molecular mechanisms of estrogen action: selective ligands and receptor pharmacology
Rae et al. GREB1 is a novel androgen‐regulated gene required for prostate cancer growth
Dong et al. Melatonin inhibits mitogenic cross-talk between retinoic acid-related orphan receptor alpha (RORα) and ERα in MCF-7 human breast cancer cells
Wang et al. Identification of a Functional Imperfect Estrogen-Responsive Element in the 5 ‘-Promoter Region of the Human Cathepsin D Gene
Singleton et al. Bisphenol-A and estradiol exert novel gene regulation in human MCF-7 derived breast cancer cells
Marino et al. Estradiol-induced IP3 mediates the estrogen receptor activity expressed in human cells
Hyder et al. Synthetic estrogen 17α-ethinyl estradiol induces pattern of uterine gene expression similar to endogenous estrogen 17β-estradiol
AU760784B2 (en) Bioassay for identifying estrogen receptor-beta/alpha selective modulators
Watanabe et al. Similarities and differences in uterine gene expression patterns caused by treatment with physiological and non-physiological estrogens
CA2160135A1 (en) Method for screening for receptor agonists
WO1996041013A1 (en) Method for screening for receptor agonists and antagonists
Harris et al. Regulation of metallothionein II messenger ribonucleic acid measures exogenous estrogen receptor-β activity in SAOS-2 and LNCaPLN3 cells
Wu et al. Ligand structure-dependent activation of estrogen receptor α/Sp by estrogens and xenoestrogens
EP1009807A1 (en) DIFFERENTIAL LIGAND ACTIVATION OF ESTROGEN RECEPTORS ER$g(a) AND ER$g(b) AT AP1 SITES
Coleman et al. SRA coactivation of estrogen receptor-α is phosphorylation-independent, and enhances 4-hydroxytamoxifen agonist activity
Tekur et al. Expression of RFG/ELE1α/ARA70 in normal and malignant prostatic epithelial cell cultures and lines: regulation by methylation and sex steroids
Castro-Rivera et al. 17β-Estradiol-and 4-hydroxytamoxifen-induced transactivation in breast, endometrial and liver cancer cells is dependent on ER-subtype, cell and promoter context
MXPA01006150A (en) Bioassay for identifying estrogen receptor-&amp;bgr;/&amp;agr;selective modulators
Tsai et al. Forskolin stimulates estrogen receptor (ER) α transcriptional activity and protects ER from degradation by distinct mechanisms

Legal Events

Date Code Title Description
FZDE Discontinued