“w,- - _. . «If, ,.. 2.0 9F. . "7 er, "mm? Cu. 5- . , 4 v . v ‘. ‘ :2; :51 . . 1., :12?» 1. At .\ 1.“..4 3.. ‘ , . r .1... : . . . ...s . 5rd 1.::‘ p... .1 1—3.“ . , Vaw‘ v. a. I‘lv .b,lx. fir. fivmvrn mm“? 3.... z. IAF . 4.4.6.. ‘ :2! f 1 z , fl. .. .. i .t... ‘r’. .u . mam.» .gguq .YnV $331.33? .,a... . :1: «.1 .. z. . «M E: . . inf?! l, l. ‘ miuwufl Hyman :2 3g .5. V: I. in“?! 9. . .1. 91...”..21. 2: II .Wnfli‘l THESIS 4 LIBRARY 200« 7’71 Michigar‘. State 5" “’0‘ University This is to certify that the dissertation entitled Functional Analysis of Estrogen Receptor-alpha mRNA Splicing Variants presented by Aliccia Berg Bollig has been accepted towards fulfillment of the requirements for Ph.D. . Physiology degree in Mm profissor il 11, 2 Date Apr 003 MS U i: an Affirmative Action/Equal Opportunity Institution 0-12771 PlACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 c:/CIRCIDataDue.p65op.15 FUNCTIONAL ANALYSIS OF ESTROGEN RECEPTOR-a mRNA SPLICING VARIAN TS By Aliccia Berg Bollig A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Physiology 2003 ABSTRACT FUNCTIONAL ANALYSIS OF ESTROGEN RECEPTOR-a mRNA SPLICING VARIANTS By Aliccia Berg Bollig Analysis of mRN A prepared from a variety of estrogen responsive cells and tissues has established that ERa mRNA is expressed as a mixture of transcripts. This heterogeneity results largely from a pattern of alternative mRN A splicing that gives rise to a family of correctly processed and exon—skipped ERa mRNAs. Although variant forms of ERa derived by alternative splicing have been identified in all ERa expressing tissues and cell lines examined and their expression levels are well documented in published reports, little is understood regarding their function. We have reconstructed ERor cDNAs representing the single exon-skipped variants ERAEZ through ERAE7 in order to analyze their activities in a well defined cell transfection system. The aims of this thesis are two-fold. First, it addresses the hypothesis that certain ERor mRN A splicing variants retain many aspects of wt ERa biochemical function; and secondly it addresses the hypothesis that EROL splicing variants alter gene expression. Biochemical analysis of ERa mRN A splicing variants has identified two exon- skipped variants that share significant residual function compared to their full-length counterpart, wt ERa. The variants with sequence deletions corresponding to exon 3 (ERAE3) or exon 5 (ERAES) are observed to display normal nuclear uptake and retain the ability to interact with transcriptional co-regulators. Furthermore, the variant ERAE3, like wt ERa, binds ligand. These properties confer on ERAE3 and ERAES the ability to inhibit the activity of some ERa-responsive promoters through an interplay with wt ERa signaling, and potentially to stimulate the transcription of other genes. Results from this study suggest that potential targets for transcriptional activation by these ERa splicing variants include the extracellular matrix protease, human collagenase gene, and the human IGF-l gene. The promoters of these genes lack consensus DNA-binding sites for ERa and appear to be regulated indirectly by ERor isoforms through interactions with other transcription factors such as AP-l. That these genes are implicated in tumorigenesis suggests that ERa splicing variants may have an influence on cell growth and cancer progression. The initial discovery of ERa mRNA splicing variants challenged any previous understanding of ER function in gene transcription and cell signal transduction. That understanding is further complicated by the present study which contributes to evidence suggesting that ERa splicing variants may have a significant impact on the ability of the breast and other tissues to respond to estrogens and other regulatory signals. To my father iv ACKNOWLEDGMENTS I am grateful to the members of my graduate committee—Drs. Susan Conrad, Kathy Gallo, Sandra Haslam, Donald Jump, and Richard Miksicek—for their helpful suggestions throughout the course of this work. I am especially grateful to Dr. Miksicek who is both my respected mentor and friend. Thank you Mary Morrison and Dr. David Ankrapp for your comradeship and insights regarding this project. I sincerely enjoyed learning from and working with both of you in the lab. TABLE OF CONTENTS Page List of Figures ......................................................................................................... viii Key to Abbreviations ................................................................................................ x I. Introduction .......................................................................................................... 1 II. Literature Review ................................................................................................ 4 1. Molecular Mechanisms of Estrogen Receptor Action ............................. 4 1.1 Multiple genes ............................................................................ 5 1.2 Non-genomic actions of estrogen ............................................... 9 1.3 Structural and functional domains of ERa ................................ 12 1.4 Factors influencing ERtx activity: Coactivators ......................... 18 1.5 Factors influencing ERor activity: Corcpressors ........................ 33 1.6 Antiestrogens and partial agonists ............................................. 39 1.7 Regulation of ERa function by phosphorylation ....................... 43 2. Nonconcensus Estrogen Enhancer Elements .......................................... 47 3. Identification of ERa mRNA Splicing Variants ..................................... 51 4. Activity of ERa Splicing Variants .......................................................... 55 III. Biochemical Analysis of ERa mRNA Splicing Variants ................................. 59 1. Introduction ............................................................................................. 59 2. Results ................................................................................................... 61 2.1 Measurement of the DNA-binding activity of the ERa mRN A splincing variants .................................................... 62 2.2 ERAE3, like wt ERa, binds ligand ............................................ 63 2.3 Subcellular localization of ERa splicing variants ..................... 64 2.4 Characterization of the transactivation function of ERa splicing variants on the vitellogenin ERE ................... 65 2.5 Dimerization and co-regulator binding properties of ERAE3 and ERAES ......................................................... 67 3. Discussion. ............................................................................................. 83 vi IV. ERa Splicing Variants Lacking Exons 3 or 5 Display Promoter-Specific Transcriptional Effects Through Nonconsensus ERES ................................ 92 1. Introduction ............................................................................................. 92 2. Results ................................................................................................... 95 2.1 Like wt ERa, ERAE3 activates the ovalbumin gene promoter ...................................................................... 96 2.2 Both ERAE3 and ERAES activate the Coll(-73)Luc reporter ........................................................... 98 2.3 ERAES activates the human [OF -1 promoter ........................... 101 4. Discussion. ............................................................................................ 118 V. Summary and Conclusions ............................................................................... 132 VI. Materials and Methods .................................................................................... 139 1. Expression Vectors ................................................................................ 139 2. Cell Culture, Transfection and CAT Assays ......................................... 139 3. Ligand Binding Analysis ....................................................................... 141 4. DNA Binding Assays ............................................................................. 141 5. Immunoblot Analysis ............................................................................. 142 6. In Vitro Protein-Protein Interaction Assays ........................................... 143 7. Immunohistochemical and Cytochemical Analysis ............................... 144 VII. List of References ........................................................................................... 146 vii LIST OF FIGURES Figure Page 1. ERa functional domains .................................................................................... 16 2. Model of nuclear receptor ligand-binding domain ....................................... i ...... 17 3. Diagram of SRC-le fimctional domains ............................................................ 29 4. Schematic of the SMRT functional domains ..................................................... 38 5. Depiction of ERa activation of consensus and nonconsensus regulatory elements ............................................................................................ 50 6. Comparison of ERor mRNA Splicing variants and wt ERa structure ................ 54 7. ERa mRNA splicing variant immunoblot analysis ........................................... 69 8. ERE-binding by wt ERor and EROL splicing variants ......................................... 71 9. Ligand-binding capacity of wt ERoc and ERoc splicing variants ........................ 72 10. Binding of nitrile THC by wt ERor and ERAE3 ................................................. 73 11. Scatchard analysis of E2 binding by wt ERa and ERAE3 .................................. 74 12. Cellular localization of wt ERa and ERa splicing variants .............................. 76 13. Activation of an ERE by wt ERa and ERa splicing variants ............................ 77 14. Inhibition of wt ERa activity by ERAE3 and ERAES ....................................... 79 15. In vitro binding of ERa isoforms and SRC-le fragments ................................. 81 16. In vitro binding of ERa isoforms and SMRT .................................................... 82 17. Depiction of ERa C-terminus secondary structure ............................................ 86 18. Activation of the Ovalbumin promoter by ERa isoforms ................................. 106 viii 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. Effect of c-jun overexpression on vaalb-CAT activated expression .............. 107 In vitro binding of c-jun and c-fos with wt ERa ................................................ 108 Activity of wt ERa and ERa splicing variants on pColl(-73)Luc ..................... 109 Effect of c-jun overexpression on pColl(-73)Luc activated expression ............. 110 Conditions of treatment for ERAE3 activation of pColl(-73)Luc ...................... 112 Activity of wt ERor and ERa splicing variants on p(AP1)3TK-CAT ................ 114 Activity of wt ERor and ERa splicing variants on the IGF-l promoter ............. 115 ERAES activation of IGF-I promoter fragments ............................................... 117 Transactivation Model 1: Coactivator recruitment ........................................... 127 Transactivation Model 2: Kinase activation ..................................................... 129 Transactivation Model 3: Corepressor neutralization ....................................... 131 ix AD 1 AD2 AF 1 AF 2 AP-l AR CAT CREB CBP/p300 Coll DBD E2 EGF ER ERa ERB ERAEZ ERAE3 ERAE4 ERAES ERAE6 ERAE7 ERE GR GRIP] GST IGF-l JN K LBD Luc MAPK Mab-l 7 mRN A N-CoR NLS Ovalb PMA PR PPAR KEY TO ABBREVIATIONS SRC-l activation domain-1 SRC-l activation domain-2 N-terminal transactivation function-l C-terminal transactivation function-2 Activator Protein 1, representing homo- and heterodimers of jun and fos Androgen Receptor Chlorarnphenicol Acetyl Transferase, a reporter gene Cyclic AMP response element binding protein CREB binding protein, a family of 300 kDa transcription coactivators -73 to +63 promoter fragment from the Collagenase gene DNA—Binding Domain 17B-Estradiol Epidermal Growth Factor Estrogen Receptor Estrogen Receptor-0t Estrogen Receptor-[3 Exon 2-skipped variant of Estrogen Receptor-alpha Exon 3-skipped variant of Estrogen Receptor-alpha Exon 4-skipped variant of Estrogen Receptor-alpha Exon 5-skipped variant of Estrogen Receptor-alpha Exon 6-skipped variant of Estrogen Receptor-alpha Exon 7-skipped variant of Estrogen Receptor-alpha Estrogen Receptor Element Glucocorticoid Receptor Glucocorticoid Receptor Interacting Protein-1 Glutathione S-Transferase Insulin-Like Growth Factor-1 c-Jun NHz-terminal Kinase Ligand-Binding Domain Luciferase, a reporter gene Mitogen Activated Protein Kinase ER—specific Monoclonal antibody messenger Ribonucleic Acid Nuclear Receptor Corepressor Nuclear Localization Signal -1342 to +7 promoter fragment from the chicken Ovalbumin gene Phorbol 12-Myristate, 13-Acetate Progesterone Receptor Peroxisome Proliferator-Activated Receptor RIP 1 40 RIP 1 60 SMRT SRC- 1 Tam TBP TIFl TIP 2 TR VDR wt ERoc Retinoic Acid Receptor Receptor Interacting Protein-140 Receptor Interacting Protein-160 Silencing Mediator of Retinoic Acid and Thyroid Hormone Receptor Steroid Receptor Coactivator-1 4-hydroxytamoxifen TATA Binding Protein Transcriptional Intermediary Factor-l Transcriptional Intermediary Factor-2 Thyroid Hormone Receptor Vitamin D Receptor 595 amino acid wild-type Estrogen Receptor-alpha xi I. Introduction Analysis of messenger RNA (mRN A) prepared from a variety of estrogen- responsive cells and tissues has established that estrogen receptor-a (ERa) mRN A is typically expressed as a mixture of transcripts. This heterogeneity results largely from a pattern of alternative mRNA splicing that gives rise to a family of correctly processed and exon-skipped variant ERa mRNAs (Gotteland et al., 1995; Hu et al., 1996; Miksicek et al., 1993; Murphy et al., 1997). Although there is no consistent ratio of relative expression, wild-type (wt) and variant ERa transcripts are coexpressed in all ERa-positive cell lines and tissues examined (Castles et al., 1995; Friend et al., 1997; Gotteland et al., 1995; Hu et al., 1996; Kuiper et al., 1996). There has been extensive analysis at the RNA level of the pattern of expression and abundance of ERor splicing variants, yet limited information is available on their functional activity. ERor mRN A comprises sequences from eight coding exons and is translated to yield a protein with discrete functional domains (for review, see Tsai and O’Malley, 1994). Like other nuclear receptors, ERa is a modular protein in that individual domains are capable of demonstrating autonomous function within receptor mutants, as well as when they are introduced into heterologous fusion proteins (Kumar et al., 1987; Tsai and O’Malley, 1994). One of the major aims of this thesis is to examine the ftmction of ERa splicing variants from the vantage point of what is known about the functional organization of wt ERa. To test the hypothesis that the mRNA splicing variants retain wt ERor activities, the function of six ERor splicing variants that arise by the deletion of one of its internal exons (ERAEZ through ERAE7, where the deleted exon is indicated numerically) was characterized. Biochemical analysis demonstrates that, the omission of a particular exon results in the loss of ERa ftmction ascribed to that exon. The deletion of an exon is also observed to disrupt activities attributed to other exons and to bestow novel function on the receptor isoform. Most notably, none of the exon-skipped variants effectively promotes gene expression from a consensus estrogen response element (ERE). The traditional view of estrogen action dictates that hormone binding by nuclear localized ER elicits a conformational change in the receptor allowing it to dimerize, recruit transcription coactivators, bind DNA at an ERE and activate target genes (Tsai and O’Malley, 1994). More recently a novel pathway for regulation of transcription by ERa has been described to involve cooperation of the receptor with AP-l factors such as c-jun and c-fos (Gaub et al., 1990; Uht et al., 1997). An important distinction of this nonclassical pathway for ERa action is that functional domains within the receptor that are crucial for promoting gene expression from an ERE appear to be dispensable for ERa activity on AP-l directed promoters (Gaub et al., 1990; Uht et al., 1997). This suggests a potential role for ERoc splicing variants in regulation of nonconsensus ERES. The second major aim of this thesis is to identify a transcription regulatory role for ERa variants. More specifically, it investigates whether ERa variants are transcriptionally active on promoters lacking a consensus ERE which have previously been shown to be, or are predicted to be, estrogen responsive. This thesis comprises seven chapters beginning with a brief topic introduction and a statement of its major aims and hypotheses. Chapter H provides a review of relevant literature to summarize the discovery and analysis of ERa splicing variants in estrogen responsive tissues and cell lines. Chapter II also reviews the literature that defines our current understanding of gene regulation by ERa. Chapter III details the research project which focuses on the biochemical characteristics of ERa splicing variants compared to wt ERa. This project tests the hypotheses that selected splicing variants retain certain aspects of wt ERa function such as DNA and ligand binding, cellular localization, and protein-protein interactions with steroid receptor co-regulators. The research on ERa variant regulation of transcription is described in chapter IV. This research project tests the hypothesis that ERa splicing variants positively regulate gene transcription and identifies promoters that may be induced by specific ERa splicing variants. Chapter V offers a summary and completes a context of relevance for the work described in the chapters preceding it. The final chapters, VI and VII, provide an outline of materials and methods and a list of references, respectively. II. Literature Review 1. Molecular Mechanisms of Estrogen Receptor Action Estrogen exerts many biological activities by influencing gene activity in estrogen sensitive cells. This genomic response is imparted when estrogen binds a ligand-activated transcription factor, the estrogen receptor (ER). Binding of estrogen to the ER elicits a change in receptor conformation that allows the receptor to bind to DNA and enhance gene expression from the promoters of regulated genes (Kumar and Chambon, 1988; Tsai and O’Malley, 1994). ER-induced gene expression supports cell growth and differentiation in many tissues. For example, it has a role in the remodeling and development of the male and female reproductive systems (Curtis Hewitt et al., 2000; Haslam, 1987; Hess et al., 1997), influences brain and cardiovascular function (Kuroski de Bold, 1999; Toran-Allerand et al., 1999), and it has a role in regulating bone growth and maintenance (Compston, 2001; Grumbach, 2000). A nuclear receptor for estrogen was first characterized in rat uterus in 1966 when specific, high affinity binding of radiolabeled l7B-estradiol (E2) to cell extracts was shown (Tofi and Gorski, 1966). Shortly thereafter, a general description of a mechanism of steroid hormone action was proposed. Jensen and colleagues described a two-step mechanism that is initiated when estrogen binds and activates a cytoplasmic receptor (Jensen et al., 1968). The activation event is not specifically defined; however, this model continues to explain that the activated hormone-receptor complex is then translocated to the nucleus and binds chromatin to cause a change in mRN A expression. Since this mechanism of steroid action was first proposed, research has corrected and elaborated upon the details of this model so that we now know that the ER is associated primarily with the cell nucleus in an unliganded state (King and Greene, 1984; Welshons et al., 1984), and that estrogen is an allosteric effector for the receptor. Ligand-bound ER forms homodimers which bind to DNA at a specific nucleotide sequence called an estrogen response enhancer element (ERE) for which the Xenopus vitellogenin A2 promoter ERE sequence (5 'GGTCACA-GTGACC-3 ') serves as a paradigm (Klein-Hitpass et al., 1988; Kumar and Chambon, 1988). Continued research has taken the current understanding of ER-directed gene expression beyond simply correcting the details of Jensen’s model. Investigators are now confronted by a regulatory mechanism with unforeseen complexity and new uncertainties. 1.1 Multiple genes The transcriptional effects of estrogens are mediated by two closely related receptor isoforms, ERa and ERB, each encoded by a separate gene. The human ERa gene, which was first cloned by Chambon and colleagues in 1986 (Green et al., 1986), is localized to the long arm of chromosome 6 (6q23) where it spans approximately 450 Kb of genomic sequence (Gosden et al., 1986; Manasce et al., 1993). The location of the more recently described human ERB gene has been mapped to chromosome 14 (more specifically, l4q22-24) (Enmark et al., 1997; Mosselman et al., 1996). Analysis of mRNA has determined that there is a wide tissue distribution for both ERa and ERB, and mRNA splicing variants for each isoforrn have been identified (Chu and Fuller, 1997; Enmark et al., 1997; Mosselman et al., 1996; Murphy et al., 1997; Poola et al., 2002; Price et al., 2001; Stabile et al., 2002). The variety of ERor splicing variants include mRNA variants with a deletion of one exon, including exons 2, 3, 4, 5, and 7, as well as combinations of exons (Miksicek et al., 1993; Murphy et al., 1997). Single exon- skipped variants of ERB include mRNA isoforms missing exons 2, 3, 4, 5, and 6 (Chu and Fuller, 1997; Poola et al., 2002). Variants of ERB also include receptors expressing an insert of 54 nucleotides within the ligand binding domain and another ERB mRNA splicing variant that harbors both the exon 3 deletion and the 54 base pair insert has also been reported (Price et al., 2001). The ERa and ERB cDNAs share a great deal of identity, particularly in the sequences corresponding to those regions of the receptor that bind ligand and DNA (59 percent and 97 percent, respectively) (Enmark et al., 1997). The resulting proteins share similarities in structure and in their function to regulate transcription. Both ERa and ERB homodimerize in the presence of E2 (Ogawa et al., 1998). In vitro and in viva binding assays suggest that ligand-bound ERa and ERB are colocalized within the nucleus and form heterodimers (Matsuda et al., 2002; Ogawa et al., 1998). The mouse homologs of ERa and ERB are shown to bind E2 with similar affinity (although a slightly lower average dissociation constant is reported for ERB) (Tremblay et al., 1997). Moreover, Ez-liganded ERa and ERB dimers bind and activate a consensus ERE with similar effectiveness. This transcriptional activity can be blocked by treatment with antagonists [CI 164, 384 and 4- hydroxytamoxifen (tamoxifen) (Mosselman et al., 1996; Paech et al., 1997). Tissues that are sensitive to estrogen generally express both isoforms of the receptor; however, the mRNAs are often present at unequal ratios and some tissues offer examples of ERa- and ERB— differential expression (Enmark et al., 1997, Mosselman et al., 1996). Human ERa and ERB mRNA are coexpressed in many tissues, including uterus, breast, lung, kidney, ovary and prostate. Examples of cells that express only one of the receptor isoforms include human granulosa cells which express only ERB mRNA and leydig cells of the human testis which only contain ERa (Enmark et al., 1997). Comparing the ratio of expression in tissues that coexpress the two isoforms Enmark et al. report that ERa is more abundant than ERB in the human uterus and ERB predominates in the breast, however quantitative analysis of ERoc and ERE mRNA in normal and breast cancer tissues suggests that ERa is the dominant transcript in tumors and ERB expression is down regulated during carcinogenesis (Enmark et al., 1997; Gustafsson and Warner, 2000; Iwao et al., 2000). A study of relative protein levels in benign and malignant breast biopsies by western blotting confirms that ERB is more abundant than ERa in the normal human mammary gland samples. Although very little ERa was detected in the normal mammary gland, it was strongly detected in invasive tumor samples (Gustafsson and Warner, 2000). Additional distinctions between ERa and ERB are revealed when fertility and breast development are examined in mice with disrupted genes. ERa knockout mice are infertile and, compared to wild-type mice expressing both ERa and ERB, ERB knockout mice have decreased fertility and fewer and smaller litters (Krege et al., 1998; Lubahn et al., 1993). In ERa knockout mice uterine weight is reduced compared to wild-type littermates and the uteri of adult ERa knockout mice do not undergo cyclic changes in response to treatment with ovarian steroid hormones (Couse et al., 1995; Korach 1994; Lubahn et al., 1993). In contrast, the uteri of adult ERB knockout mice resemble the wild-type phenotype (Krege et al., 1998; Lubahn et al., 1993). Embryonic and fetal development of the mammary gland occurs in the absence of either receptor, however, in ERor knockout mice postpubertal ductal growth is absent. Like wild-type counterparts, ERB knockout mice develop a normal ductal structure that fills the entire fat pad (Couse et al., 2000; Gustafsson and Warner, 2000). Together with the relative abundance of receptor isoforms observed in normal and malignant breast biopsies these findings suggest that ERa and ERB each offer a unique contribution to estrogen responsiveness in the breast. Further consideration and additional studies that consider the relative ratio of expression of ERa and ERB may offer greater insight into both normal breast development and the advancement of cancer. Notable differences between ERa and ERE are also evident when their capacity to effectively activate various promoters is compared. In agreement with earlier studies, Paech and coworkers have shown that with E2 treatment ERa and ERB were equally effective at activating an ERE-directed reporter gene construct in HeLa cells (an ER-negative human cancer cell line of uterine origin). Treatment with antiestrogens raloxifene, tamoxifen and ICI 164384 did not support this expression. When their activities were compared on an AP-l response element treatment with raloxifene, tamoxifen and ICI 164,384 induced gene expression from an AP-l -directed reporter gene in HeLa cells cotransfected with ERB. E2 treatment of ERB-expressing cells had no effect. Distinctively, E2 treatment induced ERa-dependent gene expression while antiestrogen treatment had relatively little effect on AP-l reporter gene activity in cells cotransfected with ERa ( Paech et al., 1997) 1.2 Non-genomic actions of estrogen While most of the biological effects of estrogens involve programmed changes in gene expression that are mediated by receptors located within the nucleus, rapid responses to estrogens have also been described that appear to originate at the cell membrane and to be independent of gene transcription (for review see, Coleman et al, 2001). These signaling events are considered to be non- genomic and can be initiated by estrogen analogues that do not traverse the cell membrane. BSA-conjugated B; does not enter the cell yet has been shown to bind specifically at the cell membrane and within minutes (a time frame too rapid to involve gene transcription) can cause an increase in intracellular signaling molecules such as cAMP, calcium and inositol-l ,4,5-triphosphatc (1P3) in many cell types (Aronica et al., 1994; Coleman et al., 2001; Fiorelli et al., 1996; Guo et al., 2002, Improta-Brears et a1 ., 1999). The putative membrane receptor for estrogen has not been fully characterized, however a study using ER-negative Chinese hamster ovary (CHO) cells to transiently express ERa and ERB demonstrated that a small percentage of BRO: and ERB were translocated to the membrane (Razandi et al., 1999). Studies using membrane preparations from these ERa- and ERB-expressing CHO cells indicate that membrane localized ERa and ERB are coupled with G- proteins. Treatment with E2 stimulated Ca2+-dependent mitogen-activated protein kinase (MAPK) activity and Gaq-dependent production of 1P3, both effects were blocked by co-treatment with the ER antagonist ICI 182, 780 (Improta-Brears et. al., 1999; Razandi et al., 1999). Rapid stimulation of PKC activity by estrogen in primary cultures of female rat growth plate chondrocytes is also attributed to a membrane localized receptor and does not involve alteration of gene expression (Sylvia et al., 2000). Sylvia and colleagues demonstrated that treatment of cultures with 17B-E2 produced a dose- dependent increase in PKC activity which was mediated by G-protein-coupled phospholipase C. PKC activity was unaltered by treatment with the ER agonist 10 diethylstilbesterol (DES) and co-treatment with [Cl 182, 780 did not block the effect of 17B-E2, suggesting that Ez-dependent PKC activity in these cultures may not be directed by either of the classical ERor or ERB receptors (Sylvia et al., 2000). Increased release of the cell signaling molecule nitric oxide (NO) is stimulated in vascular endothelial cells when Ez-liganded ERa activates phosphatidylinositol-3-OH kinase (PI3-K). P13-K activates protein kinase B/Akt (PKB/Akt) which subsequently activates endothelial nitric oxide synthase (eNOS) (Hisamoto et al., 2001; Rameh and Cantley, 1999; Simonicini et al., 2000). ERa- associated PI3-K activity is ligand-dependent and sensitive to antiestrogen treatment. Co-transfection experiments using CHO cells determined that E;- stirnulated PKB/Akt activity in these cultures was dependent on transfected ERor. Notably, E2 treatment did not enhance PKB/Akt activity in cells transfected with ERB (Hisamoto et al., 2001). Studies reported by Simonicini et al., suggest that the activation of PI3-K by ERor is in part the effect of recruitment and binding of PI3-K by ERor localized in the cytosol. ERor interaction with the PI3-K regulatory subunit p850r is ligand-dependent and not supported by ICI 182, 780 binding. Consistent with its inability to activate PI3-K, ERB did not complex with p850: (Simoncini et a], 2000). Unlike ER-dependent PI3-K activation, both ERa and ERB mediate the activation of MAPK in a number of cell types including the human breast tumor cell line, MCF7, in the presence of E2 (Improta-Brears et al., 1999; Migliaccio et al., 1996) However, although co-treatment with ICI 182, 780 or tamoxifen inhibits ll ERor-directed MAPK activation, ERE-directed activation is stimulated by antiestrogen treatment (Wade et al., 2001 ). This is not an entirely unique effect as the ERor-dependent increase in intracellular cAMP in breast cancer and uterine cell lines was also found to be stimulated by both estrogens and antiestrogens (Aronica et al., 1994). Further complicating our understanding of ER regulation is the knowledge that CAMP acts as a second messenger which signals a PKA-dependent binding of ERor and cyclin D1, an association which signals ligand-independent activation of ERE—directed gene transcription (Lamb et al., 2000). 1.3 Structural and functional domains of ERor To understand the mechanisms by which ERor transactivates a promoter it is useful to break the receptor down and describe its individual functional domains. Like other nuclear receptors, BRor is a modular protein that has discrete functional domains capable of demonstrating autonomous activity when they are expressed in receptor mutants as well as when they are introduced into heterologous fusion proteins (for review, see Beato and Sanchez-Pacheco, 1996; Kumar et al., 1987). ERor is translated from mRNA with eight coding exons (Ponglikitmongkol et al., 1988). An N-terminal transactivation function (AF 1) encoded by exon 1 and a portion of exon 2 is thought to promote gene expression by interacting with and recruiting other transcription factors (e.g., nuclear receptor coactivators) to the promoters of responsive genes (Endoh et al., 1999; Kobayashi et al., 2000; Metivier et al., 2001; Metivier et al., 2000; Metzger et al., 1995; Ofiate et al., 1998; Sathya et 12 al., 2002; Tremblay et al., 1999; Watanabe et al., 2001; Webb et al., 1998). Also, AF 1 directly binds, and may thereby stabilize, basal transcription factors TFIIB, TATA-binding protein (TBP), and the TBP-associated factor TAFn30 (Ing et al., 1992; Jacq et al., 1994; Sadovsky et al., 1995). Derived fiom exons 2 and 3 is a centrally located zinc-finger motif (commonly referred to as the DNA-binding domain or DBD) that is essential for sequence-specific DNA binding and transcriptional activation through a consensus ERE (Kumar et al., 1987). A receptor dimerization interface is also located within this domain, specifically between the first two cysteines of the second zinc finger (Schwabe and Rhodes, 1991). Within the region encoded by exon 4 are the nuclear localization signals (NLS) and a hinge region that allows for receptor conformation flexibility (Tsai and O’Malley, 1994; Ylikomi et al., 1992). A ligand binding domain (LBD) confers regulatory function to the receptor and is encoded by the C-terminal exons 4 through 8 (Kumar et al., 1986). The ERor LBD structure resembles the crystal structures of other nuclear receptor LBDs (Brzozowski et al., 1997; Nolte et al., 1998; Tanenbaum et al., 1998). An arrangement of receptor a-helices numbered H3 to H12 contribute to the ERor LBD structure. The ligand-binding pocket is formed principally by H3, H6, H8, and H12. In the unliganded state H3, H6 and H8 form the interior cavity and H12 extends away from this domain. When E2 binds the cavity it makes direct contact with critical interior residues and effects a conformational change that causes H12 to fold and contact H3, H5 and H11. As depicted in Fig. 2, this orientation of H12 bars the cavity entrance and locks the hormone in the binding 13 pocket (Brzozowski et al., 1997; Chen et al., 1996). Within the LBD are binding sites for heat shock proteins such as Hsp90, Hsp40, Hsp56, and Hsp70 which form a heterologomeric complex with ER. Heat shock proteins bind to the nascent ER and in concert with accessory proteins (e. g. p23, hip, hop, and CyP40) assemble to form a dynamic multicomponent complex which promotes proper protein folding and functions to maintain the mature receptor in a conformation which facilitates receptor interaction with hormones (Howard et al., 1990, Kimmins et al., 1999). The region encoded by exons 4 through 8 also includes additional regulated determinants for subunit dirnerization, and a well-characterized C-tenninal transactivation function (AF 2) (Kumar and Chambon, 1988). In the ligand-bound state, AF 2 adopts a conformation that supports the association of ERor with factors that are known to cooperate with the receptor to promote gene transcription (e.g., coactivators or other transcription factors) (Nolte et al., 1998; Tanenbaum et al., 1998). With this association, ERor may act to enhance gene expression by ushering additional transactivators to ERE-containing promoters. When the receptor is unliganded or binds an antagonist, AF 2 does not bind transcription promoting factors and may instead interact with proteins (generally referred to as corepressors) that repress transcription (Nagy et al., 1999). Unlike AF 2 activity, the N-terminal AFl activity is considered to act independently of ligand binding (Tora et al., 1989); i.e., it is constitutively active and its function is generally not influenced by the binding of antagonists that are otherwise shown to hinder AF 2 activity (Berry et al., 1990; Metivier et al., 2001). The two activation domains function independently l4 when introduced into heterologous fusion proteins and may fimction independently in the context of the full-length receptor on certain promoters in a cell type-specific manner (Berry et al., 1990; Tora et al., 1989); yet it is clear that for many estrogen- sensitive promoters and cell types full transcriptional activity is the sum total of the activities of both transactivation domains (Berry et al., 1990; McInery et al., 1996; Sathya et al., 2002; Tora et al., 1989). 15 Hinge Region AF‘l _ 1 38 116 184 263 302 549 595“ V l I [080 NLS w\' A B Fig. 1. A representation of ERor functional domains. The ER functional domains and A-F regions are arranged as they reside along the length of the receptor. Relative positioning of the nuclear localization signal (NLS), DNA-binding domain (DBD), ligand binding domain (LBD), AF 1 , and AF 2 domains are depicted. l6 Fig. 2. Three dimensional model of a nuclear receptor ligand-binding domain with and without bound estradiol. Left, in the unliganded state helix 12 (H12) extends away from the opening of the ligand-binding cavity which is formed by helices H3, H5, and H11. Right, when estradiol binds to the cavity it effects a conformation change that causes H12 to fold and contact H3, H5, and H11. This folded orientation of H12 bars the cavity entrance and locks the hormone (shown in green) in the binding pocket. From L. P. Taylor and H. Akil. NRR Graphics Library, http://biocheml . basic—sci. georgetown. edu/nrr/glib. htmI 1.4 Factors influencing ERor activity: Coactivators Binding of estradiol to ERor elicits a conformational change that promotes receptor dirnerization and translocation to an ERE in the nucleus of ERor-expressing cells. However, the expression of ER and the accessibility of an ERE in a target cell are not the sole requirements for an efficient genomic response. The process of hormone binding and the resulting change in conformation also promotes an interaction with proteins that enhance or deter ERor activity. The profile of ERor transcriptional activity has been recently modified to incorporate a role for two new classes of proteins, coactivators and corepressors. Coactivators function to enhance the activity of ligand-bound nuclear receptors, and corepressors associate with nuclear receptors in the unliganded state to repress activity (Nagy et al., 1999). The existence of a coactivator-type protein was first suspected when investigators observed a squelching of ERor activity in cells overexpressing the receptor (Meyer et al., 1989). Transcriptional interference suggested that the receptors were competing for a limiting factor that was fundamental to receptor activity. This notion was ultimately reinforced when proteins were identified that were shown to directly bind to ERor and enhance ERE-directed reporter gene expression (Cavailles et al., 1994; Ofiate et al., 1995; Voegel et al., 1996). The nature of coactivator involvement in nuclear receptor-mediated transcription was firrther elucidated when over expression of a dominant negative isoform of a single coactivator was shown to effectively block ERor-enhanced reporter gene expression (Ofiate et al., 1995). In 1994 Malcolm Parker and colleagues described two murine l8 proteins of 160 and 140 kDa (referred to as receptor interacting proteins, RIP160 and RIP140, respectively) that associated directly with mouse ERa in the presence of E2 (Cavailles et al., 1994). Later it was shown that these particular proteins enhanced ERor-directed transcription in reporter gene expression experiments (Cavailles et al., 1995; Voegel et al., 1996). Since these proteins were characterized, the list of coactivators has grown significantly to include, among others, a group of “pl 60” nuclear receptor coactivators (NCOAI , NCOA2, and NCOA3), the CBP/p300 and P/CAF proteins, and the multirneric SWI/SNF and TRAP/DRIP/ARC complexes (Freedman, 1999; Janknecht and Hunter, 1996; McKenna et al., 1999; Muchardt and Yaniv, 1993; Naar et al., 1999; Rachez et al., 1999). The p160 coactivators from several species have been cloned and characterized by a number of laboratories, giving rise to a confusing nomenclature. For consistency, NCOAI (also called SRC-l , RIP160, and TIFl) will be referred to by its more common name, SRC-l , while NCOA2 (TIF2/GRIP1/SRC-2) and NCOA3 (ACTR/AIBl/RACB/p/CIP) will be referred to as TIF2 and ACTR, respectively. In general, coactivators enhance nuclear receptor transcriptional activity in the presence of an agonist and do not associate with antagonist-bound receptor. Coactivator frmction requires recruitment to the promoter by the receptor or by other DNA-binding transcription factors. The growing list of coactivators and similarities in their behavior suggest that they are functionally redundant proteins. However, continued research hints that there are preferences among the various nuclear receptors (NR5) for individual coactivators (Glass and Rosenfeld, 2000; 19 Ding et al., 1998; Kalkhoven et al., 1998; Voegel et al., 1996). There are three classes of coactivators distinguished by the nature of their intrinsic activities which appear to be critical for their role in nuclear receptor- directed gene transcription (for review, see Glass and Rosenfeld, 2000). The first class of coactivators includes mammalian homologues of the yeast SW12/SNF2 factor named BRGl and hBrm (Laurent et al., 1993; Khavari et al., 1993; Muchardt and Yaniv 1993). SWI/SNF functions as a multiprotein coactivator complex and has DNA-stimulated ATPase activity (Laurent et al., 1993; Owen-Hughes et al., 1996). SWI/SNF complexes are recruited to promoter elements by interactions with sequence specific DNA-binding proteins (including ER). With the hydrolysis of ATP they effect a local remodeling of chromatin structure that facilitates binding of additional transcription regulatory factors and correlates with enhanced gene transcription (Chiba et al., 1994; Kingston and Narlikar, 1999; Laurent et al., 1993). A second class of coactivators are known to possess a histone acetyltransferase (HAT) functional domain. Examples of coactivators with HAT activity include SRC-l , ACTR, P/CAF, and CBP/p300. As with the SWI/SNF complex, translocation of these coactivators to DNA allows their intrinsic HAT activities function, remodeling chromatin and facilitating the binding of additional transcriptional regulatory proteins to specific DNA sequences (Freedman, 1999; Ogryzko et al., 1996; Spencer et al., 1997; Wolffe, 1994; Yang et al., 1996c). The third class of coactivators is characterized by the TRAP/DRIP/ARC complex. TRAP/DRIP/ARC subunits assemble to form a multiprotein composite coactivator 20 that binds both nuclear receptors and other coactivators (Fondell et a1, 1999; Glass and Rosenfeld, 2000; K0 et al., 2000; Nair et al., 1999; Rachez et al., 1999). It is unclear how TRAP/DRIP/ARC functions to enhance NR—dependent transcription. The capacity for TRAP/DRIP/ARC to interact with NRs and potentially recruit other coactivators may in part define their role in NR-directed gene transcription. The TRAP/DRIP/ARC complex does not have intrinsic HAT activity, however, it associates with other coactivators including CBP/p300 which do have HAT activity (Ko et al., 2000). The p160 coactivators (SRC-l , TIF2, and ACTR (also referred to as AIBl, RAC3, or p/CIP) share similarities in structure, activation function, and receptor binding properties (Freedman, 1999) and have received much attention in the literature. To date, the relationship between coactivator and ERor activity is best understood within the context of SRC-l , which was the first nuclear receptor accessory factor reported to efficiently stimulate the AF 2 activity of ERor (Ofiate et a1 ., 1995). In a study designed to assess direct interaction between coactivators and nuclear receptors, researchers observed varying affinities for interaction between the nine nuclear receptors and two coactivators examined (Ding et al., 1998). Differences are especially notable in the in vitro preference for TIF2 binding by the androgen receptor (AR) and progesterone receptor (PR) compared to the relatively weak affinity these receptors exhibited for SRC-l (Ding et al., 1998). Comparing TIF2 and SRC-l activities, results fi'om reporter gene experiments suggest that TIF2 21 enhances transactivation by AR, ER, and PR, but not by the glucocorticoid receptor (GR), retinoic acid receptor (RAR), thyroid hormone receptor (TR) and vitamin D receptor (V DR), whereas SRC-l associates with and stimulates the AF 2 activity of all nuclear receptors (Ding et al., 1998; Nolte et al., 1998; Ofiate et al., 1995; Voegel et al., 1996). In more comprehensive studies SRC-l activity was tested on promoters responsive to transcription factors other than nuclear receptors. Coexpression of SRC-l does not affect cyclic AMP response element-binding protein (CREB) activity on a CREB-responsive promoter in reporter gene assays unlike the CBP/p300 coactivator. Therefore, although SRC-l has been shown to enhance transcription by nuclear receptors, it is not a general coactivator for all inducible transcription factors (Freedman et al., 1999; Nestin et al., 2000; Oflate et al, 1995). In contrast, CBP/p300 has been shown to bind both NRS and CREB, allowing it to function as a signal integrator (Kamei etal., 1996) Three human SRC-l isoforms have been cloned (Hayashi et al., 1997; Kalkhoven et al., 1998). The isoforms are encoded by the same gene and have an identical amino acid sequence up to residue 1385. The SRC-la C-terminus extends forty-two amino acids longer than SRC-le and the last fourteen C-terminal amino acids of SRC-le (1385-1399) are not conserved in SRC-la (Kalkhoven et al., 1998). SRC-lq differs fiom SRC-la by deletion of one C-terminal glutamine residue (amino acid 1386 of SRC-la) (Hayashi et al., 1997). It has been shown that SRC- la, SRC-le and SRC—lq function similarly and all three share a capacity to bind NRs and enhance NR-induced reporter gene transcription (Hayashi et al., 1997; 22 Kalkhoven et al., 1998; Ma et al., 1999; Ofiate et al., 1995). Two reports suggests that SRC-le may be more effective than SRC-la and SRC-lq in coactivating NR transcription (Hayashi et al., 1997; Kalkhoven et al., 1998). Binding and transactivation studies suggest, and studies of co-crystal structures strengthen the argument, that the conformational change induced by an agonist is required for formation of an active nuclear receptor/SRC-l complex (Darimont et al., 1998; Ding et al., 1998; Nagy et al., 1999; Nolte et al., 1998; Ofiate et al., 1995). The necessity for SRC-l activity in vivo is exemplified in studies of ovariectomized mice with a disrupted SRC-l gene. Xu and coworkers measured a decrease in estrogen-induced uterine growth in ovariectomized SRC-l - null mice compared to heterozygous mutants and wild-type counterparts (Xu et al., 1998). These authors also report that the endocrine feedback control system and mammary gland proliferation and differentiation in response to estrogen and progesterone treatment are disrupted in homozygous SRC-l mutants (Xu et a1 ., 1998). It is important to stress that, although stunted, there was uterine and mammary gland development in these SRC-l -mutant mice with hormone (estrogen and progesterone) treatment and that both male and female homozygous mutants were fertile (Xu et al., 1998). Results fiom these studies suggest that SRC-l is important for steroid hormone-mediated gene transcription, since mice lacking SRC- 1 exhibit partial resistance to E2 treatment. Conversely, these results also substantiate the notion that one coactivator may, to some extent, compensate for another. 23 As noted above, coactivator-like accessory factors were first hypothesized when receptor activity squelching was observed in transient transfection studies (Meyer et al., 1989). SRC-la over expression is shown to reverse the squelching of PR—directed gene expression by cotransfected ligand-bound ERor in some contexts, in a dose-dependent manner (Lopez et al., 1999; Ofiate et al., 1995). This observation is tempered by reports of other efforts to study the limiting influence of SRC-l which indicate that over expression of SRC-l may itself attenuate reporter gene expression (Lopez et al., 1999). Studies suggest that the degree of coactivator influence on receptor activity is cell type-specific, and under certain transfection conditions using a range of exogenous coactivator expression levels, the presence of too little or too much coactivator may be equally ineffective at potentiating receptor activity or alleviating receptor activity squelching (Cavailles et al., 1995; Kamei et al., 1996; Lopez et al., 1999). These results led some researchers to investigate whether or not SRC-l can titrate out another factor that in turn becomes limiting. In separate transient transfection experiments it has been shown that p300/CBP over expression can enhance cooperative SRC-l/ERa activity, and under different conditions, it can alleviate SRC-l attenuation of ERor-directed reporter gene expression (Kamei et al., 1996; Lopez et al., 1999; Smith et al., 1996). When considering the mechanism by which SRC-l enhances ERa activity it is necessary to reiterate that evidence from binding studies suggests that SRC-l acts by interacting directly with ERa and is recruited by the receptor to the promoter where it modulates gene expression. This puts the coactivator at the scene of 24 transcription initiation, but it does not fully explain how this complex enhances receptor activity. Two modes of activity are thought to contribute to SRC-l activity. First, SRC-l associates with other coactivators (e.g., CBP/p300) (Yao et al., 1996) and it directly binds the basal transcription factors TFIIB and TBP (Takeshita et al., 1996). It may in effect be recruiting other necessary factors to the ERor-responsive promoter for efficient transcription and acting to stabilize the transcription initiation complex. Secondly, SRC—l is shown to have HAT activity, a function that, as described earlier, correlates with remodeling of the chromatin structure and increased gene transcription (Spencer et al., 1997; Wolffe, 1994). An (Jr-helical receptor-binding motif with the sequence LXXLL is conserved among many of the coactivators and is sufficient for ligand-dependent nuclear receptor binding (Heery et al., 1997). Site-directed mutagenesis of residues in this motif reduce or abolish interaction with nuclear receptors (Heery et al, 1997; Kalkhoven et al., 1998). Three of these sequences are localized to the central region of all SRC-l isoforms and one additional LXXLL motif is present in the divergent C-terminus of SRC-1a(Kalkhoven et al., 1998). Each of them contributes to interaction with ERor, but mutational studies demonstrate that maintaining the integrity of just the second of the three central motifs is sufficient for in vitro ERor binding. This observation, and reports which suggest that there are different affinities among a variety of receptor/coactivator complexes that interact via this binding sequence, indicate that motif location and the context of the residues surrounding this motif may influence receptor binding preference (Ding et al., 1998; 25 Kalkhoven et al., 1998; Voegel et al., 1996). The binding of SRC-l can be mapped to a conserved C-terminal (Jr-helix in the LBD of nuclear receptors (Danielian et al., 1992; Darimont et al., 1998; Nolte et al., 1998; Shiau et al., 1998; Tanenbaum et al., 1998). When a nuclear receptor binds its cognate ligand, receptor helix 12 folds to expose a surface to accommodate SRC-l binding. Crystal structures of the LBDs of multiple nuclear receptors, including ERor, indicate that helix 12 extends outwardly from the body of the unliganded receptor. When the receptor binds estrogen, helix 12 is tightly packed against the LBD and makes direct contact with the estrogen molecule (Darimont et al., 1998; Nolte etal., 1998; Shiau et al., 1998; Tanenbaum et al., 1998). This conformation brings a conserved glutamic acid and lysine residue to the receptor surface in an orientation that allows them to form hydrogen bonds with the amino and carboxy-terminal ends, respectively, of the contacting SRC-l LXXLL motif. The length of the LXXLL helix is accommodated by a hydrophobic clefi which forms between these specified glutamic acid and lysine residues (Darimont et al., 1998; Nolte et al., 1998; Shiau et al., 1998). Additional contacts occur between coactivators and helices 4 and 5 of the LBD. Coactivator binding is impaired by mutations in this region, and binding of antagonists such as raloxifene to ERor do not support the appropriate conformational change to expose the receptor SRC-l docking site (Barettino et al., 1994; Brozozowski et al., 1997; Danielian et al., 1992; Durand et al., 1994). Co-crystal structure analysis indicates that the two AF 2 domains of a nuclear receptor dimer contact separate LXXLL motifs of a single 26 SRC-l coactivator molecule (Nolte et al., 1998). In addition to the well characterized C-terminal interaction, SRC-l also associates with an N-terminal region of ERa. In most contexts, the synergistic activity of AF 1 and AF 2 is required for maximal Ez-stimulated activity on an ERE; i.e., the full-length receptor, with both AF 1 and AF 2 intact, activates ERE-directed gene transcription more effectively in many cell types than mutated counterparts lacking one of the activation functions (McInery et al., Metivier et al., 2001; 1996; Tora et al., 1989). The first convincing data that SRC-l interacts with both AF 1 and AF 2 came from experiments in which truncation mutants corresponding to the AF 1 - containing N-terminus and AF2-containing C-terminus of ERor were expressed in the presence of SRC-l (Kraus et al., 1995). When both receptor mutants were simultaneously expressed, the effect on reporter gene activation was synergistic in the presence of SRC-l compared to either receptor mutant expressed individually alone or in combination with SRC-l (McInery et al., 1996). Considering that it has been shown that SRC-l binds the ERor N-teminus in vitro (Metivier et al., 2001; Ofiate et al., 1998), it is plausible that SRC-l and perhaps other coactivators, as well, are simultaneously binding both the N- and C-terminus mutants and thereby integrating the ligand-independent AF 1 fimction and Ez-induced AF 2 activity to fully activate the reporter gene in these studies. This model has received considerable experimental support and is believed to account for much of the cell- type specificity that has been observed for AF] function (Benecke et al., 2000; Kobayashi et al., 2000; Metivier et al., 2001). 27 As mentioned above, members of the p160 class of coactivators have an acetyltransferase catalytic activity and multiple nuclear receptor interaction motifs. They also have intrinsic activation domains that correlate with binding sites for other coactivators (Koh et al., 2001; Ofiate et al., 1998; Sheppard et al., 2001). The centrally located SRC-l activation domain 1 (ADl) binds the homologous CBP and p300 factors (Sheppard et al., 2001). Each of these are themselves coactivators with intrinsic HAT activity that interact with other activating proteins such as P/CAF (Ogryzko et al., 1996; Yang et al., 1996c). A C-terminal activation domain (AD2) within the p160 coactivators binds coactivator-associated arginine methyltransferase 1 (CARMl) (Koh et al., 2001). Recruitment of CARMI to the transcription intitiation complex by a nuclear receptor coactivator, and its ability to synergistically enhance steroid receptor-mediated transcription in the presence of coexpressed pl 60 coactivator, renders CARMl a secondary cofactor (Chen et al., 1999; Koh et al., 2001). The same mutation that eliminated the ability of CARMl to methylate histone H3 in vitro also reduced CARMl ’s ability to enhance transcriptional activation in reporter gene experiments (Chen et al., 1999). It appears that CARMl ’s ability to selectively methylate histone H3 coincides with enhancement of steroid receptor-mediated transcription. 28 1| % i b;% , -ADz - 1 399 aa 570-780 989-1 240 781 -988 1 241 -1 399 Fig. 3. Diagram of SRC-le functional domains. The location of the nuclear receptor (NR) binding domains are indicated by hatched boxes. The activation function domains are represented by black boxes. The SRC- 1 e amino acid coordinates are indicated under the diagram. 29 The knowledge that some coactivators possess different activities does little to help our understanding of why there are so many factors that frmction as NR coregulators. Many coactivators share the same functional domains (e.g., both SRC- 1 and ACTR have HAT activity) and furthermore it is unclear why multiple coactivator activities are even necessary. These uncertainties will be addressed when the mechanism by which coactivators potentiate NR-directed transcription is better understood. In a review of coregulator function, Glass and Rosenfeld describe three models to explain how multiple coactivators might have a role in regulating NR transactivation (Glass and Rosenfeld, 2000). The first model proposes that various coregulatory complexes are recruited in a sequential fashion such that one coactivator may initially bind a NR and subsequently be exchanged for another. The binding of each successive coactivator may present a different activity that is required in a particular chronological order to effectively enhance transcription. A second model speculates that various coactivators are simultaneously recruited by NRs to a promoter. This model suggests that proteins from each of the functionally distinct coactivator classes may act in concert to modulate gene transcription. The final model considers that the same gene may be responsive to the assembly of a variety of coactivator complexes. Multiple signal transduction events may independently effect the expression of the same gene to varying degrees by causing different coactivators, coordinated in either a simultaneous or sequential manner, to be recruited to the target gene promoter. A better understanding of how cofactors and NRs effect transcription may be 30 advanced with a greater appreciation that promoters are organized into nucleosomes and that chromatin structure contributes to regulation of promoter activity. Transactivation studies using the mouse mammary tumor virus (MMTV) promoter emphasize this point. The MMTV promoter is packaged into a regular array of nucleosomes which suppresses basal transcription when the MMTV promoter is organized into replicating chromatin (Bresnick et al., 1992). GR binding to elements in this nucleoprotein associated promoter induces a chromatin remodeling event that allows secondary transcription factors to bind, thereby activating transcription. When the MMTV promoter is transiently transfected into cells it adopts a less organized structure that allows secondary transcription factors to bind constitutively. Compared to the chromatin template, basal activity for the transfected promoter is relatively higher and a smaller degree of induction by liganded GR is observed (Archer et al., 1992; Fragoso et al., 1998). Like the MMTV promoter, the nucleoprotein structure of the genomic p82 promoter has also been mapped (Sewack and Hansen, 1997). The p82 promoter contains an ER binding enhancer site and is transcriptionally upregulated by liganded ER. Using a chromatin irnmunoprecipitation (ChIP) assay Sewack et. a1. demonstrate specifically that the acetylation of two histones within the p82 chromatin array is induced with estradiol treatment in ER expressing cells. Moreover, they correlate this acetylation with DNAse 1 hypersensitivity and promoter activation (Sewack et al., 2000). Rather than simply knowing that acetylation and nuclease sensitivity is increased generally, this experimental method 31 allows researchers to address the question of which nucleoproteins are acetylated and directs their attention to where any subsequent disruptions may occur within the targeted promoter nucleosome structure. ChIP assays have also been used to demonstrate that both the coactivator BRGl , and the basal transcription factor TBP, interact with the p82 promoter chromatin in an ER—dependent manner (DiRenzo, et. al., 2000; Sewack et al., 2000). The association of BRGl with the promoter complex plays a significant role in the positive regulation of the p82 gene. From studies with the p82 promoter we know that coactivation of ER by SRC-l requires BRGl (Sewack et al., 2000). Results from studies investigating GR regulation of the MMTV promoter offer further insight into BRGl function in NR transactivation. Liganded-GR recruits BRGl to the MMTV promoter where the presence of both GR and BRGl is required for recruitment of the secondary transcription factor, nuclear factor 1 (NFl). BRGl subsequently directs the ATP-dependent dissociation of GR and BRGl from the chromatin template (Fletcher et al., 2000). It is unclear whether NFl remains associated with the MMTV promoter after the dissociation of GR and BRGl , however, it is clear that the association of GR and BRGl with the chromatin is a dynamic process. Corroborating this observation, McNally et al. report that liganded-, fluorofore-labeled GR undergoes continuous exchange between being bound to and dissociating from chromatinized MMTV DNA regulatory elements in living cell cultures (McNally et al., 2000). 32 1.5 Factors influencing receptor activity: Compressors In contrast to coactivators, corepressors are a class of proteins that interact with unliganded nuclear receptors and repress transcription (for review, see Horowitz et al., 1996). Two of the better characterized examples of corepressors are the ubiquitously expressed, homologous proteins SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) and N-CoR (nuclear receptor corepressor) (Chen et al., 1995; Horlein et al., 1995). Biochemical studies have determined that SMRT and N-CoR have multiple conserved silencing domains at their N-terminus and distinct C-terminal receptor interaction domains (Li et al., 1997; Nagy et al., 1999; Perissi et al., 1999; Web et al., 2000). Their role in regulating transcription is especially apparent when the mechanism for gene silencing by the RAR and the TR is considered. Unlike ER, class H nuclear receptors such as RAR and TR form heterodimers with RXR and localize to their cognate DNA response elements as unliganded heterodimers. In this state they strongly repress basal expression from promoters containing TR and RAR enhancer elements (Baniahmad et al., 1992). This transcriptional silencing effect of class H receptors is mediated by the interaction of unliganded receptor with corepressors (Chen et al., 1995; Horlein et al., 1995). Basically, ligand binding causes corepressor dissociation fiom the DNA-bound receptor complex, encourages coactivator interaction, and ultimately triggers gene transcription (Chen et a1 ., 1995; Horlein et al., 1995; Nagy et al., 1999; Perissi et al., 1999). The suggestion that the SMRT and N-CoR proteins are transcriptional corepressors that facilitate repression 33 by unliganded TR and RAR is supported by protein-protein interactions and transient transfection experiments with expression vectors for the GAL4 DBD—fused SMRT and N-CoR proteins. SMRT and N-CoR complex with unliganded RAR/RXR or TR/RXR heterodimers that are associated with their cognate DNA enhancer elements (Chen et al., 1995; Horlein et al., 1995). A repression of basal transcription from a GAL4-responsive reporter gene in cells over expressing GAL4 DBD-fused SMRT or N-CoR C-terminally truncated mutants demonstrates that SMRT and N-CoR have multiple independent silencing function domains with activity that can be conferred on heterologous fusion proteins (Chen et al., 1995; Hdrlein et al., 1995). The silencing domains correspond with binding sites for Sin3A/B proteins (Li et al., 1997). Sin3 acts as a scaffold protein that links SMRT and N-CoR with basal transcription factors and histone deacetylases to form a multisubunit repressor complex (Heinzel et al., 1997; Nagy et al., 1997; Wong and Privalsky, 1998) Reducing the availability of Sin3 and its associated histone deacetylase RPD3 by antibody blocking correlates with abolition of N-CoR- dependent transcriptional repression (Heinzel et al., 1997). In a similar role to that of histone acetylases, deacetylases may influence transcription by affecting DNA structure. DNA wraps around histone proteins to form nucleosomes, and nucleosomes give shape to chromatin (Wu and Grunstein, 2000). Acetylation destabilizes nucleosomes by causing DNA to be less tightly associated with histones. Enhanced transcriptional activation parallels increased histone acetylation (Bartsch et al., 1996; Wolffe, 1994). On the other hand deacetylation stabilizes the 34 nucleosome structure and decreased transcriptional activity is generally associated with increased deacetylation (Burke and Baniahmad, 2000; Chen and Townes, 2000) The location and structure of nuclear receptor interaction domains found in coactivators are conserved in SMRT and N-CoR. An I/LXXII motif, which resembles the coactivator LXXLL motif, dictates the sites of nuclear receptor interaction with corepressor (Nagy et al., 1999; Perissi et al., 1999; Webb et al., 2000). The C-terminal half of SMRT contains two I/LXXII motifs, and the C- terminus of N-CoR has three motifs that contribute to receptor binding (Nagy et al., 1999; Perissi et al., 1999; Webb et al., 2000). Since the nuclear receptor interaction domains conserved in SMRT and N-CoR resemble those described for coactivators, researchers speculated that the domains for coactivator and corepressor binding to nuclear receptors may overlap. The I/LXXII motif is believed to have an amphipathic (rt-helical structure with a non-polar surface on one side that, like the coactivator L)O(LL motif, could be a site for hydrophobic interaction with NRS (Feng et al., 1998, Mak et al., 1999; Nolte et al., 1998). Studies found that point mutations of specific amino acids at or near receptor residues critical for coactivator binding located in receptor helices 4 and 5 also disrupt corepressor binding (F eng et al., 1998; Nagy et al., 1999; Perissi et al., 1999). However, carboxy terminal truncation of RAR or TR, which deletes helix 12 and results in a receptor mutant that shows reduced coactivator binding, exhibits enhanced interaction with SMRT (Li et al., 1997). Further truncation of the C-terminus that removed helices 11 and 35 12 of TR and RAR reduced SMRT interaction with the receptor implicating sequences within helix 11 in corepressor binding. It appears that corepressors bind nuclear receptors at multiple sites and, to some extent, share similar points of contact with coactivators; yet a clear distinction arises when receptor helix 12 is considered. To recapitulate, coactivator binding is severely impaired by mutations in helix 12 (Collingwood et al., 1997), but corepressors still bind receptor helix 12 mutants (Li et al., 1997). Moreover, additional studies suggest that corepressors do not efficiently dissociate from helix 12-mutant nuclear receptors (Chen et al., 1996). Unlike class 11 nuclear receptors (such as RAR and TR), which form unliganded RXR heterodimers that bind DNA, class I nuclear receptors (i.e., steroid receptors, including ERor) only bind DNA as ligand-activated homodimers. In the ligand-bound state, ERor assumes a conformation that promotes coactivator interaction and ERor-directed gene transcription (Darimont et al., 1998; Ding et al., 1998; Nagy et al., 1999; Nolte et al., 1998; Ofiate et al., 1995). Researchers have also shown that ERor binds SMRT both in the absence or presence of ligand (Smith et al., 1997), but it is unclear what consequence this may have on transcriptional signaling. Recent reports suggest that one consequence of corepressor binding may explain the mixed agonist/antagonist nature of tamoxifen, which under some circumstances inhibits ERor activity and at other times acts as a stimulatory agent (Katzenellenbogen et al., 1996; Smith et al., 1997). Whether tamoxifen acts as an agonist or antagonist when bound to the receptor may be dictated by the relative abundance of coactivators and corepressors in a particular cell. Using a reporter 36 gene transfection assay in yeast cells, Graham and coworkers demonstrated that when the coactivator L7/SPA is coexpressed with tamoxifen-liganded ERor the agonist activity of tamoxifen was enhanced (Graham et al., 2000). L7/SPA is shown to interact with NRS and enhance partial agonist activity of antagonists bound to PR, GR and ERor (Jackson et. al., 1997). Similar increased activation was observed when TIF2 or SRC-l and ERor were coexpressed with an ER—responsive reporter gene construct in HeLa cells (Webb et al., 1998). 37 $01 302 R101 RIDZ 1 134 475 743 1495 aa Fig. 4. Schematic of the SMRT functional domains. The diagram shows where functional domains are roughly positioned along the length of the 1495 amino acid SMRT protein. The hatched boxes represent receptor interaction domains (RID) and black boxes specify silencing domains (SD). 38 1.6 Antiestrogens and partial agonists Estrogens are critical for the normal development and firnction of physiological processes in a variety of tissues, however, estrogen is also implicated in the promotion of pathological processes such as breast and uterine cancer (Gustafsson and Warner, 2000; Lavinsky et al., 1998; Lawson et al., 1999; Speirs et al., 1999). Awareness of estrogen related pathologies has made the ER a target for endocrine therapy and spawned the development of selective estrogen receptor modulators (SERMS) which bind ER and influence its activity. Initial research focused on the ability of estrogen-like compounds to disrupt the proliferative effect estrogen has on tissues such as the breast and uterus. Now SERMS are also investigated to assess their potential to mimic the positive effects estrogen has on physiological parameters such as bone growth and maintenance, and cardiovascular and neurological fitness (Dechering et al., 2000). Ideally, SERMS would offer the beneficial estrogen effects in some tissues while lacking or inhibiting the undesirable estrogen effects in other tissues. Speculation that further investigation of SERMS may disclose a cancer preventative agent has prompted even more research (King et al., 2001). The SERM classification catagorizes a group of compounds that range from very effective estrogen receptor agonists to pure antiestrogens which interact with both ERor and ERB to differentially regulate their function in control of cell growth and signaling (Gustafsson and Warner, 2000; Katzenellenbogen et al., 2000). Antiestrogens are competitive antagonists of ER activity. They occupy the ERor and 39 ERB LBD to the exclusion of agonists and inhibit the ER transactivating function. Many ER—binding compounds have been synthesized and confirmed to have an antagonistic nature. This array of antiestrogens effect inhibition of ER transactivation to varying degrees that are shown to be cell type- and promoter- specific (Berry et al., 1990; McDonnell et al., 1995: Yang et al., 1996a; Yang et al., 1996b). Antiestrogens are divided into two basic classes: pure antiestrogens and partial, or mixed, antiestrogens. The related compounds ICI 164,384 and [CI 182,780 exemplify pure antiestrogens. In vivo studies indicate that [Cl 182,780 inhibits uterine and mammary cell growth and consistently blocks estrogen-induced, ERor-directed transcription of an ERE reporter gene (Bowler et al., 1989; Nuttall et al., 2000; Wakeling et al., 1991; Wijayaratrre et al., 1999). The chemicals raloxifene and tamoxifen are examples of mixed antiestrogens. As a relatively recently developed drug, raloxifene has received comparatively less attention in the literature than other antiestrogens; however, a few reports on raloxifene studies offer a great deal of insight into the character of both pure and mixed antiestrogen activity. Raloxifene inhibits an estradiol growth response in breast and uterine cells; conversely, it has a stimulating effect on bone growth and remodeling (Draper et al., 1996; Huston, 1999). Administration of raloxifene stimulates TOE-[33 expression in the femur of ovariectomized rats and cotransfection studies indicate that raloxifene induces ERor-dependent activation of the TGF-B3 promoter (Yang et al., 1996a; Yang et al., 1996b). Although we were 40 unable to confirm their results in HeLa cells, Yang et al. report that cotransfected ERor strongly activates a reporter gene construct driven by the TGF-B3 promoter in human osteosarcoma MG63 cells treated with raloxifene. The TGF-B3 promoter lacks a consensus ERE and was not activated by E; treatment in these studies. Moreover, in a reversal of roles, cotreatrnent with E2 antagonized raloxifene action (Yang et al., 1996b). A comparison of the crystal structures of the E2- and raloxifene-bound ERor LBD indicates that the receptor undergoes a different conformational change upon binding of structurally dissimilar ligands (Brzozowski et al., 1997). When raloxifene settles into the LBD, a portion of the raloxifene molecule is not accommodated by the cavity (it protrudes from the pocket) and prevents H12 from folding into the body of the LBD as it does when estrogen binds (Brzozowski et al., 1997, Ekena et al., 1997). These observations provide direct evidence that the binding of structurally distinct compounds effects distinct changes in receptor conformation. Further research suggests that this difference modulates the ability of nuclear receptors to recruit co-regulators (Lavinsky et al., 1998; Smith et al., 1997). Like raloxifene, tamoxifen is a mixed antiestrogen and acts as an agonist (to varying degrees), or antagonist of ER-induced gene expression in a cell type- and promoter-specific manner (Katzenellenbogen et al., 1996, Montano et al., 1998). Tamoxifen antagonizes estrogen stimulated growth of breast cancer cells in culture and is ofien used successfully in cancer treatment to cause grth cessation and regression of ER-positive breast tumors (Furman et al., 1992; Lamy et al., 2002). 41 The duality of tamoxifen action is evident in the treatment of some breast cancer patients for whom it is observed that tamoxifen antagonizes the proliferative effects of estrogen in the breast, yet encourages the cell proliferation that introduces an increased risk for carcinogenesis in the uterus (Juneja et al., 2002; Decensi, et al., 2002; Shang and Brown, 2002). Activation of the quinone reductase (QR) gene by tamoxifen-liganded ERor and ERB provides another example of the agonist activity of tamoxifen. QR is an enzyme that decreases the generation of hydroxyl radicals in a cell by reducing quinones. This may be a factor in defining tamoxifen’s putative role as an anti- cancer agent. Tamoxifen regulation of the QR promoter shows complete reverse pharmacology such that ER directed QR expression is suppresed by E; in breast cancer cells (Montano et al., 1998). From these examples it appears that the promoter, tissue-type and receptor isoform all play a role in SERM signaling. Tamoxifen binding supports dimer formation and receptor DNA binding (Cheskis et al., 1997). As noted above, ER has two distinct transactivation domains. The C-terminal AF 2 is regulated by conformation of the LBD. AF 1 at the N- terminus is not directly regulated by ligand. Both contribute to the total activity of the estrogen-bound full-length receptor on an ERE (Benecke et al., 2000; kabayashi et al, 2000; Metivier et al., 2001; Tora et al., 1989). When tamoxifen binds to ERor, in contrast to E2 binding, the resulting LBD conformation impairs AF 2 activity; however, AF 1 activity remains intact (Berry etal., 1990; Brzozowski et al., 1997). Therefore, with tamoxifen bound, a receptor may dimerize and translocate to a 42 promoter and effect transcription via a functional AF 1 domain. Notably, although ERor and ERB share a great deal of homology, their sequence is most divergent at the N-terminus (Enmark et al., 1997). Perhaps by affecting how a receptor interacts with cofactors, any N-terminal structural difference between ERor and ERB may distinguish the effect tamoxifen binding has on the receptors (Berry et al., 1990; Lazennec et al., 2001). Furthermore, differential expression of receptor isoforms may partially explain tissue specific responses to tamoxifen treatment (Gustafsson and Warner, 2000; Shang and Brown, 2002). Evidence is building to suggest that the relative amounts and profile of corepressor and coactivator expression may also be a contributing factor. A decreased level of N-CoR expression correlates with the observation of tamoxifen-resistant growth in both cultured human breast cancer cells and in mouse breast tumors (Lavinsky et al., 1998). In transient transfection studies, over expression of SRC-l is reported to enhance tamoxifen-stimulated transcription in a dose—dependent manner (Smith et al, 1997). Conversely, over expression of SMRT or N-CoR reduced tamoxifen-stimulated transcription (Lavinsky et al., 1998; Smith et al., 1997). Consistent with the notion that the selective interaction and relative levels of co-regulators may influence the activity of tamoxifen-liganded receptor, in vitro binding studies have shown that both corepressors and coactivators can bind tamoxifen-liganded receptor (Smith et al., 1997). 1.7 Regulation of ERor function by phosphorylation Steroid hormone receptors, including ERa, are substrates for a variety of 43 kinases and phosphatases. The state of phosphorylation of specific receptor residues affects receptor function (Arnold et al., 1997; Joel et al., 1998). Unliganded receptors are basally phosphorylated and become hyperphosphorylated following association with hormone. This increased phosphorylation observed with agonist binding is not supported by the binding of receptor antagonists (Denner et al., 1990; Denton et al., 1992; Orti et al., 1989). In vitro phosphorylation of purified ERor by Src family tyrosine kinases (e.g., p60"’src and p56'°") on tyrosine 537 correlates with maximum estrogen binding capacity and ERE association (Arnold et al., 1997, Arnold et al., 1995b). In vitro dephosphorylation by protein-tyrosine phophataselB (PTPlB), or a phenylalanine substitution of this residue significantly decreases ERor honnone—binding capacity (approximately 90 percent) and also inhibits DNA binding (Arnold et al., 1997, Arnold et al., 1995b). Results from studies designed to investigate intracellular localization of phosphorylated ER in Ez-treated MCF-7 cells (an ER—positive breast carcinoma cell line) demonstrate that ninety percent of the MCF-7 ER population is nuclear localized and phosphorylated at tyrosine 537. The smaller percentage of ER found in the MCF-7 cytosolic fraction is not phosphorylated at tyrosine 537 (Arnold et al., 1995b). Increased affinity of ERor for its ERE and enhanced activation of an ERE- directed reporter gene construct by ERor is found to occur with phosphorylation of serine 167 (Arnold et al., 1995c; Frddin and Gammeltoft, 1999; Joel et al., 1998). Serine 167 phosphorylation is regulated by a mitogen-activated protein (MAP) kinase signal transduction cascade. Serine 167 is directly phosphorylated by a p90 kDa ribosomal S6 kinase (RSK; also known as MAPK-activated protein kinase-1, MAPKAP-Kl) (F rodin and Gammeltofi, 1999; Joel et al., 1998). RSK is activated when it is phosphorylated by the MAPK, extracellular signal-regulated kinase (ERK) (Blenis, 1993; Joel et al., 1998). RSK and ERK phosphorylate both cytosolic and nuclear localized proteins (Frodin and Gammeltofi, 1999). Over expression of either RSK or ERK enhances endogenous ER-mediated transcription of an ERE- driven reporter gene construct in Ez-treated MCF-7 cells (Joel et al., 1998). Serine 118 is located in the ER A/B region and is also phosphorylated by ERK. Phosphorylation of serine 118 by ERK correlates with enhanced ERor gene activation in cotransfection studies (Kato et al., 1995). Kato and coworkers observed increased expression from an ERE-driven reporter gene construct in ER- negative COS-1 cells when ERK was overexpressed with either ligand-bound wt ER or a truncated receptor containing receptor regions A-C (including the AF 1 and DBD domains). This suggests that phosphorylation of serine 118 modulates AF 1 activity. Furthermore, these authors report that coexpression of either Ras or its substrate Raf (MAPK kinase kinase) enhanced activity of the AF 1 -containing truncated mutant (Kato et al., 1995). Some studies suggest that ER and growth factor receptor signal transduction pathways may cooperate to transduce the cell response to growth factors such as IGF-l , TGFor, and EGF (Bunone et al., 1996; Ignar-Trowbridge et al., 1996; Newton et al., 1994). The ERK kinase pathway that converges on the ER is activated by membrane-associated receptor tyrosine kinases, examples of which 45 include receptors for the growth factors EGF and IGF -1 (for review, see Pearson et al., 2001). Receptor tyrosine kinases phosphorylate and activate Ras which in turn activates Raf. MAPKK or MEK is subsequently phosphorylated and activated by Raf. The substrate for MEK is ERK (Pearson et al., 2001; Frddin and Gammeltofi, 1999). TGFa similarly acts via a parallel MAP kinase cascade which through phosphorylation of p38 MAPK induces phosphorylation of ERK and RSK (F rodin and Gammeltofi, 1999). The Treatment of cells with IGF-l, EGF or TGFor stimulates hyperphosphorylation of ERor and is reported to result in both enhanced ERa-dependent transcription in cotransfection assays and cell growth (Bunone et al., 1996; Ignar-Trowbridge et al., 1996; Kato et al., 1995; Newton et al., 1994). 46 2. Non-consensus Estrogen Enhancer Elements Recent reports have identified an increasing number of genes with promoters lacking an ERE that show ERor-enhanced gene expression. For these promoters, transcriptional stimulation by ERor appears to be mediated by an indirect mechanism, involving synergistic protein-protein interactions with known transcription factors including AP-l and SP-l (Gaub et al., 1990; Krishnan et al., 1994) The expression levels of the RARor, cathepsin D and c-fos genes are upregulated by Ez-liganded ERor. The ERor enhancer element maps to a GC-rich Spl binding motif in the promoter region of these genes (Duan et al., 1998; Krishnan et al., 1994; Sun et al., 1998). Porter and colleagues demonstrate that ERor and Spl directly bind in solution (Porter et al., 1997). Using a gel mobility shift assay, another report confirms that both ERor and Spl complex with a putative Spl/ERor response element in the c—fos promoter (Duan et al., 1998). Collectively, these studies demonstrate that the E2 responsiveness of the RARor, cathepsin D, and c-fos promoters in transient transfection studies requires the presence of ERor, Spl , and the identified Spl binding motif (Duan et al., 1998; Krishnan et al., 1994; Sun etaL,1998) AP-l describes the ubiquitous jun/fos family of transcription factors whose activity is crucial for the efficient expression of a wide variety of genes. As an important downstream target for the MAPK signaling cascades, AP-l is a central 47 player in mediating the effects of serum and growth factors on cellular proliferation (Boyle et al., 1991; Westwick et al., 1994). A variety of estrogen-responsive genes have been described that lack a palindromic ERE, but instead contain one or more consensus AP-l elements (5 '-TGAG/CTCA-3 '), with or without a degenerate ERE or ERE half-site (5'-GGTCA-3' or 5’TGACC-3'). Examples of such genes include ovalbumin, which is induced by E; in chicken oviduct cells (Tora et al., 1988), and the rat insulin-like growth factor-1 (IGF-l) gene whose expression is stimulated by E; in the uterus of ovariectomized-hypophysectomized rats and in cultured rat osteoblast cells (Ernst and Rodan, 1991; Murphy and Ghahary, 1990). An AP-l enhancer motif identified in the chicken IGF-l promoter is essential for E2 and phorbol ester-stimulated gene transcription (U mayahara et al., 1994). Phorbol ester serves to stimulate increased AP-l activity by directly activating PKC, which in turn activates Ras to initiate a MAP kinase cascade that sequentially includes the activation of MEKK (MAPK kinase kinase), c-jun N-terminal kinase kinase (JNKK) and c-jun N-terminal kinase (JN K). JN K ultimately phosphorylates and thereby activates AP-l (Boyle et al., 1991; Bokemeyer et al., 1996; Vuong et al., 2000; Westwick et al., 1994). Like the rat IGF-l gene, the chicken ovalbumin promoter does not have an ERE and the ERor regulatory element maps to a critical AP-l site (Gaub et al., 1990; Tora et al., 1988). That this effect is mediated by AP-l is supported by the observation that the synergistic induction of ERor-dependent expression by E2 and phorbol ester cotreatrnent is further enhanced by cotransfection with c-fos or c-jun 48 (Gaub et al., 1990). Reporter gene cotransfection studies with expression vectors for AP-l isoforms and ERor in HeLa cells indicate that a similar mechanism regulates the human collagenase promoter (Webb et al., 1995). The minimal region of the collagenase promoter reported to be responsive to tamoxifen-liganded wt ERor (and to a variety of ERa mutants) harbors a critical AP-l element and lacks a consensus ERE. The activity of ERor on the collagenase promoter was enhanced with AP-l (c- jun or c-fos) over expression (Webb et al., 1995). Furthermore, evidence that ERor regulation converges with AP-l-directed gene transcription is provided by results from protein binding assays indicating that c-jun is able to bind to wt ERor in vitro (Webb et al., 1995). 49 cell nucleus GGTCAnnnTGACC TGAGICTCA TGAGICTCA Fig. 5. A diagram depicting wt ERor transcriptional activation of consensus and nonconsensus regulatory elements. The left side of the figure represents a receptor dimer acting through a consensus ERE to transactivate gene expression. The receptors in the lower middle and on the right are cast in a nonclassical transactivating role on a consensus AP-l motif (TGAG/CTCA). In this role it is unclear if ER directly binds AP-l factors (e.g., jun and fos) or indirectly interacts with them through association with coactivators that bind AP-l factors. 50 3. Identification of ERor mRNA Splicing Variants Numerous variant ERor cDNAs have been cloned and sequenced from a variety of breast tumors and established tumor cell lines (Castles et al., 1995: Gotteland et al., 1995; Miksicek et al., 1993; Murphy et al., 1997). The most common variants harbor a precise deletion of one or more of the internal exons that contribute to the structure of the mature ERor protein, suggesting that they arise as a result of imprecise splicing of the primary ERor mRNA transcript. ERor cDNAs with deletions corresponding to exons 2, 3, 4, 5, and 7 have been identified, along with a large number of more complex variants (Castles et al., 1995: Gotteland et al., 1995; Miksicek et al., 1993; Murphy et al., 1997). These basic variants are referred to as ERAE2 through ERAE7, where the deleted exon is indicated numerically. While there is no consistent ratio of relative expression, wt and variant ERor transcripts are apparently coexpressed in all ERa-positive cell lines and tissues (Castles et al., 1995; Gotteland et al., 1995; Hu et al., 1996). Quantitation of individual variants shows that each variant generally represents a minority of ERor mRN A; however, as a population, splicing variants can constitute 50 percent or more of the total ERor mRNA (Castles et al., 1995; Erenburg et al., 1997; Gotteland et al., 1995; Murphy et al., 1997). While most efforts to evaluate expression levels of individual variants have focused on mRNA, Traish et al. report that of 29 human breast tumors they analyzed, 45 percent contained ER protein with a defect in the DNA-binding domain. Using gel mobility shift analysis and site-directed 51 monoclonal antibodies these authors also report that 3 of the 29 tumors contained ER protein which had defects in both the N-terminal A/B region and the DNA- binding domain (Traish et al., 1995). Quantitation of ER variant transcripts has revealed that the population of ERor transcripts is mixed and varies among the tissues and cell types examined; furthermore, wt ERor is usually the major transcript expressed. However, some studies indicate that the presence of a single splicing variant can sometimes exceed wt ERa in amount. Quantitation of mRNA transcripts in the BT-20 breast cancer cell line shows that ERAES is the major ERor isoform expressed in these cells (Castles et al., 1993; Hall et al., 1990). ERAES comprises 68 percent of the ERor mRNA population while wt ERor measures only 8 percent. Results from immunoblot analysis agree with mRNA analysis and demonstrate that ERAES is the predominant ER protein expressed in BT-ZO cells (Castles et al., 1995). The receptor status in the BT-2O cancer cell line, which was considered to be ER- negative by ligand-binding analysis, raises the notion that the ratio of variant and wt ERor expression may affect hormone responsiveness and cancer progression. In this context, analysis of wt ERor and ERAES mRN A in various stocks of the human breast tumor cell line MCF-7 showed that an elevated ratio of ERAES compared to wt ER mRNA correlated with the ability of some MCF-7 sublines to grow independently of estrogenic stimulation (Klotz et al., 1995). Studies also indicate that, while ERAEB tends to be underrepresented in 52 breast tumors and tumor cell lines, it appears to constitute 50 percent or more of ERor mRNA in both stromal fibroblasts and epithelial cells isolated fi'om reduction mammoplasty specimens (Erenburg et al., 1997). This leads to speculation that higher relative abundance of ERAE3 may be a feature of normal breast tissue, where it may slow cell growth relative to breast tumor cells. Examination of the growth of MCF-7 cells stably transfected with ERAE3 supports this notion. Estrogen stimulated the ability of the parental cell line to form colonies in soft agar. Reduced colony formation was observed for ERAE3 expressing clones (Erenburg et a1 ., 1997) 53 DNA Hormone wildtype N c 66.6kDa AF‘I NLS AF2 I VII VIII Exon Structure r l .11.": IV 'V .V| ' Zi III IIIIII ensez n—i-c:_r§-1_::i;jj:j::j}-c 17.0 kDa ERAE3 N———-" f ' ' ' ' c 62.3kDa ERAE4 N——-.t‘ 1W0 54.1 kDa. ERAES n—Icmc j;:-::::::r-c 41w» ERAEG N MyIIIZG-C 53.0 kDa ERAE7 N We ri-c 52.2 kDa Fig. 6. Comparison of ERor mRNA splicing variants and wt ERor structure. The locations of the various functional domains of ERa are depicted along with the exon sequence from which they are derived. The variants are referred to by deleted exon. The size and molecular weight of each of the variants are predicted from the translational reading frame of the sequenced cDNA clones. Dashed lines indicate regions of the major open reading frame of the full-length ERor protein that are missing from each variant. The nuclear localization signal is circled. The regions encompassing the DNA- and ligand-binding domains are marked by darkened and hatched boxes, respectively. The AF 1 domain and AF 2 core domains are indicated where they reside within the N- and C-termini of the wild-type receptor. 54 4. Activity of ERor Splicing Variants While there has been extensive analysis of the pattern of expression and abundance of ERor splicing variants, limited information has been available until recently on their functional activity. Early reports indicated that ERAES can support weak, cell type-dependent activity (Chaidarrrn and Alexander, 1998; Fuqua et al., 1991; Rea and Parker, 1996). Also, when tested on an ERE, both ERAEB and ERAES are dominant negative receptor forms in the presence of wt ERor (Ohlsson et al., 1998; Wang and Miksicek, 1991). Taken together these two observations suggested to us that splicing variants may play a role in regulating gene transcription. Fuqua and colleagues reported that ERAES (which contains the AF 1 domain, but lacks AF 2 and the regulatory functions imparted by the LBD), is constitutively active in promoting transcription from an ERE in a heterologous yeast reporter gene assay (Fuqua et al., 1991). These authors also described that over expression of ERAES in a stably transfected breast cancer cell line supported greater proliferation compared with control cells, as well as imparting a tamoxifen-resistant phenotype (Fuqua and Wolf, 1995). In the human osteosarcoma cell line U2-OS, coexpression of ERAES significantly enhances ERE-directed reporter gene expression induced by wt ERor (Chaidarun and Alexander, 1998). The presence of a constitutively active receptor variant able to exert a mitogenic effect in breast tumor cells in the absence of E2 or in the presence of tamoxifen was thought to be an appealing explanation for 55 aquired antiestrogen resistance observed in previously responsive breast tumors and cell lines (Wiseman et al., 1993; Wolf and Jordan, 1994). However, this model is now challenged by conflicting observations that ERAES and closely related, genetically engineered ERor mutants do not efficiently induce transcription from an ERE reporter when transiently transfected into the ER-negative HeLa or chicken embryo fibroblast (CEF) cell lines, or promote proliferation in stably transfected breast tumor cells (Kumar and Chambon, 1988; Rea and Parker, 1996). A constitutive transcriptional activity has also been reported for the ERAE4 variant. When transfected into the ER-negative Chinese hamster ovary CHO k1 cell line, ERAE4 was observed to induce an ERE-containing reporter gene construct in a hormone-independent manner (Pasqualini et al., 2001). Like reports that describe ERAES to be a constitutively active variant, the significance of this finding is called into question by results from another study which suggests that ERAE4 does not bind DNA in vitro and ftuthermore does not transactivate an ERE in human choriocarcinoma JEG3 cells (Koehorst et al., 1994). Aside from the potential for ERor variants to activate a promoter, the mRN A splicing variants ERAE3 and ERAES are reported to have an inhibitory effect on wt ERor transactivating activity in cotransfection studies (Ohlsson et al., 1998; Wang and Miksicek, 1991). A 70 percent inhibition of transcriptional activation by E;- liganded wt ERor on an ERE-driven reporter gene was observed in HeLa cells when ERAE3 and wt ERa were coexpressed (Wang and Miksicek, 1991). In the ER- 56 negative nontumorigenic human breast cell line HMT-3522S 1 , coexpression of an equal amount of ERAES also significantly inhibited stimulation of an ERE reporter construct by wt ERor (Ohlsson et al., 1998). Increasing the ratio of transfected variant to wt ERor demonstrates that the repression of wt ERor by ERAE3 and ERAES is dose-related and becomes nearly complete when the variants are present in sufficient excess over the intact receptor (Ohlsson et al., 1998; Wang and Miksicek, 1991). This observation has physiological significance in the case of cells such as the BT-20 breast tumor cell line that predominantly express one of these splicing variants. Additionally, stable over expression of ERAE3 in MCF-7 cells to levels seen in normal mammary epithelial cells dramatically reduced the ERor- dependent expression of endogenous p82 mRN A and anchorage independent growth of MCF-7 cells (Erenburg et al., 1997). ERAES retains both the DBD and AF 1 domains and, based on what we know of the modular character of wt ERor, presumably also retains the functions ascribed to these domains. With the DBD and AF 1 intact, ERAES may also retain the ability to bind an ERE or interact with co-regulators, thereby possessing the capacity to transactivate genes or disrupt wt ERor activity by competing for DNA or coactivator binding. The fact that ERAE3 lacks an intact DBD must account, at least in part, for its inability to direct gene induction through an ERE, even though it retains fully functional AF 1 and AF 2 domains. Furthermore, ERAE3 inhibition of wt ERor activity on an ERE-driven reporter gene may be a result of its ability to 57 prevent wt ERor from binding to an ERE (Miksicek et al., 1993; Wang and Miksicek, 1991). The limited observations described above hint at a functional role for some of the ERor mRNA splicing variants. Considering the prevalence of exon-skipped ERor variants it is clear that further study is warranted to more thoroughly evaluate their activities, particularly regarding their capacity to regulate gene transcription. 58 III. Biochemical Analysis of ERor mRNA Splicing Variants Published in the journal Molecular Endocrinology (Bollig and MIkSicek, 14(5): 634-649, 2000) 1. Introduction Numerous variant ERor cDNAs have been cloned and sequenced from breast tumors and established tumor cell lines (Castles et al., 1995; Gotteland et al., 1995; Miksicek et al., 1993; Murphy et al., 1997). The most common variants harbor a precise deletion of one of the internal exons from the eight that contribute to the structure of the mature ERor protein, suggesting that they arise as a result of imprecise splicing of the primary ERor mRNA transcript. Like other nuclear receptors, ERor is a modular protein in that individual domains demonstrate autonomous function (Beato and Sanchez-Pacheco, 1996; Kumar et al., 1987). Based on mutational studies, it can reasonably be assumed that the exclusion of a particular exon will predictably result in a receptor variant lacking the function ascribed to that exon. Additionally, it is probable that the loss of a particular exon will result in unpredictable functional deficits or perhaps even bestow a novel function on the variant receptor. This study examines the function of wt ERor splicing variants from the vantage point of what is known about the functional organization of wt ERor. Concurrently, the process of examining splicing variants, like mutational studies, improves our understanding of wt ERor firnction. 59 Following are results from experiments designed to assess receptor capacity to translocate to the nucleus, bind to DNA, bind ligand, participate in protein complexes, and promote gene transcription. 60 2. Results Cytomegalovirus (CMV) promoter-driven ERor cDNA expression vectors representing the exon-skipped variants ERAE2 through ERAE7 have been constructed in order to enable their functional characterization in a well defined cell transfection system. This assembled pool of ERor splicing variants also includes the hypothetical receptor ERAE6, even though this variant is not readily identified in vivo. Figure 6 diagrams the ERor mRN A splicing variants examined, showing the positions of deleted exons and their consequences with respect to protein structure. Deletion of exon 2, 5, 6, or 7 all cause a frame-shift mutation resulting in premature termination of translation, thereby generating a diverse class of C-terminally truncated receptor forms. Omission of either exon 3 or 4 does not disrupt the mRN A reading flame, but produces a receptor protein with an internal deletion. Transient expression in Cos7 cells demonstrates that each of these variants translates to a stable protein able to accumulate to readily detectable levels within transfected cells (Fig. 7). The C037 cell line is an ER-negative SV40 large T- antigen transformed cell line derived from the simian kidney cell line CV-l. Expression of SV40 large T-antigen allows for replication of our CMV-derived expression vectors which contain the SV40 origin of replication (Gluzrnan 1981). Based on immunoblot analysis with an N-terminal monoclonal antibody (Mab-17), which recognizes an epitope within exon 1 common to all of the variants (Neff et al., 1994), we observe that the mobility of the six variant proteins is consistent with 61 their predicted molecular weights. No immunoreactivity is observed in mock transfected cells, confirming the specificity of the Mab—l 7 antibody. 2.1 Measurement of the DNA-binding activity of the ERor splicing variants Efficient DNA binding by ERor requires the c00peration of several functional elements within this protein, including the centrally located DBD and a ligand- inducible subunit dirnerization motif located near the C-terminal end of the LBD (Kumar and Chambon, 1988; Kumar et al., 1987). Since all of the ERor splicing variants sustained deletions within various regions of this protein, it was of interest to systematically assess the DNA-binding ability of each variant. For this purpose, gel mobility shift assays were performed using extracts prepared from Ez-treated, transiently transfected Cos7 cells. Extracts were incubated with a 32P-labeled oligonucleotide (AGGTCACAGTGACCT) containing a consensus ERE from the Xenopus vitellogenin A2 promoter. As expected, variants that harbor a deletion within the DBD (ERAE2 and ERAE3) are completely unable to recognize the ERE (Fig. 8, lanes 5—8). Less predictably, the loss of exons contributing to the LBD also result in a strong defect in ERE recognition (Fig. 8, lanes 9-16). For ERAES and ERAE7, however, this appears to be a quantitative defect in DNA binding. The addition of the monoclonal antibody, Mab-17, to the binding reactions consistently results in the recovery of weak DNA binding by ERAES (Fig. 8, lane 12). Presumably, the role of the bivalent antibody is to stabilize the interaction of receptor subunits withtheir palindromic binding site, mimicking the function of the 62 missing dirnerization motif present within the C-terminus of the LBD. These results suggest the possible existence of cell-specific constituents that perform a similar function in viva (e.g., co-regulators) and may account for the variable activity of ERAES and related constructs in different cell types (Chaidarun and Alexander, 1998; Fuqua et al., 1991; Rea and Parker, 1996). We have also observed the formation of a complex between ERAE7 and labeled ERE probe (Fig. 8, lane 16). Overall, however, the relative weakness of DNA-binding observed in these studies raises serious questions about the extent to which any of the variants, including ERAES and ERAE7, are able to recognize and bind to a consensus ERE, in viva. Furthermore, that ERAE7 binds an ERE in vitro has little transcriptional relevance in light of the observation that ERAE7 is not translocated to the nucleus when expressed in Cos7 cells (see below). 2.2 ERAE3, like wt ERor, binds ligand To test the ability of the ERor mRNA splicing variants to bind hormone, we performed a saturation binding assay on whole-cell extracts from Cos7 cells transiently transfected with wt ERor or the ERor variants. Only wt ERor and ERAE3 were able to bind 3H-labeled E2, whereas all of the remaining variants demonstrated no specific ligand binding (Fig. 9). The individual deletion of exons 2, 4, 5, 6, and 7 effectively eliminates all, or a significant portion, of the LBD (see Fig. 6), consistent with their loss of hormone binding. These results were confirmed using an in viva ligand-binding assay in which the binding of a fluorescent estrogen analog was 63 visualized in cells cultured on glass cover slips. Cos7 cells transiently expressing the individual variants or wt receptor were treated with the fluorescent ligand, nitrile tetrahydrochrysene (nitrile THC) (Miksicek et al., 1995). Only those cultures transfected with wt ERor or ERAE3 were observed to stain with this ligand. In both cases, staining was localized tightly within the nucleus. This suggests that among the variants examined, wt ERor and ERAE3 exclusively bind ligand and in both cases, the receptors are translocated normally to the nucleus of expressing cells (Fig. 10). The affinities of ERAE3 and wt ERa for 3H-labeled 132 were compared with Scatchard analysis. The measured dissociation constants were 0.66 nM for wt ERor and 0.79 nM for ERAE3 (Fig. 11). 2.3 Subcellular localization of ERor splicing variants To more carefully assess the subcellular localization of ERor splicing variants, including those that fail to bind ligand, Cos7 cells were transiently transfected with expression vectors encoding wt ERor or individual variants. These receptors were detected in transfected cells by indirect immunofluorescence staining (using the Mab-17 monoclonal antibody) and confocal microscopy. Similar to wt ERor, ERAE3 and ERAES localize to the nuclei of transfected cells, although ERAES showed perinuclear as well as nuclear staining (Fig. 12). These results are consistent with the fact that both ERAE3 and ERAES retain a NLS immediately downstream of the DBD (Tsai and O’Malley, 1994; Ylikomi et al., 1992). Subcellular localization studies have also been completed for the exon 2, 4, 6, and 7 deletion variants. Each of these proteins can be readily detected in transfected cells, but they all possess dramatic defects in nuclear targeting (Fig. 12). Nuclear targeting of wt ERor is governed, in large part, by a tripartite karyophilic signal present within exon 4 (Ylikomi et al., 1992). Loss of this signal is therefore consistent with the cytoplasmic pattern of distribution of mutants such as ERAE2 and ERAE4, both of which lack protein corresponding to exon 4 sequences. Inappropriate presentation or folding of this signal must account for the defects in nuclear localization seen with ERAE6 and ERAE7, since the NLS is retained in these variants. Based on their subcellular distribution, we would predict that only ERAE3 and ERAES, like wt ERor, would have the potential to exert nuclear effects, such as modulating gene transcription. Moreover, the inability of the cytoplasmic variants ERAE2, ERAE4, ERAE6, and ERAE7 to dimerize with wt ERor (see below) predicts that their subcellular distribution will not be influenced by coexpression with the nuclear isoforms (wt ERor, ERAE3, and ERAES). 2.4 Characterization of the transactivation function of ERor splicing variants on the vitellogenin ERE HeLa cell cotransfection experiments designed to assess the transcriptional activity of individually expressed ERor splicing variants have failed to demonstrate any significant ability of variant receptors to support gene activation through an 65 ERE, with the possible exception of the ERAES variant, which is reported to display a low level of constitutive transcriptional activity on an ERE—driven reporter in some, but not all, cell types examined (Chaidarun and Alexander, 1998; Fuqua et al., 1991; Rea and Parker, 1996). In our hands none of the variants were effective transcriptional activators of an ERE-containing promoter. ERAES repeatedly showed only modest constitutive activity (5 percent of wt ERor induction) on an ERE-directed reporter plasmid cotransfected into HeLa cells (see Fig. 13). It is important to recognize that the tissues and cell lines that express these variants also express wt ERor. It has previously been reported that the ERAE3 variant acts as a dominant negative mutant when it is coexpressed with wt ERor in HeLa cells treated with B; (Wang and Miksicek, 1991). In the human breast epithelial cell line HMT-352281, ERAES has also been shown to disrupt transactivation by agonist-bound wt ERor of an ERE reporter gene (Ohlsson et al., 1998). To clarify whether this is a frmction unique to these variants, we completed a series of experiments to test whether the remaining exon-skipped ERor variants also support transcriptional inhibitory effects. When examined in a HeLa cell cotransfection assay in which the expression of pERE-TK-CAT was driven by E;- bound wt ERor, a 5-fold molar excess of any of the splicing variants lacking exons 2, 4, 6, or 7 failed to inhibit the Ez—dependent induction of CAT gene expression by intact receptor. In agreement with previously published results, the ERAE3 aned ERAES variants both demonstrated a dominant inhibitory activity at all molar ratios 66 tested (Fig. 14). Coupled with the observation that equal amounts of plasmid DNA support similar levels of variant expression (see Fig. 7), it appears that ERAE3 and ERAES are approximately equivalent in their inhibitory activity in HeLa cells. 2.5 Dimerization and co-regulator binding properties of ERAE3 and ERAES We next questioned whether a direct interaction between the variants and factors responsible for ERE—directed transcription might explain the inhibitory effects of ERAE3 and ERAES on wt ERa activity; specifically, we tested for a direct interaction of ERAE3 and ERAES with wt ERor and SRC-le. The C-terminus of wt ERor and fragments of the SRC-le protein were expressed as fusion proteins with glutathione S-transferase (GST) and attached to glutathione-Sepharose beads. Binding assays with GST fused to the C-terminus ofwt ERor (GST-AF2) and ”s- methionine labeled in vitro translated receptor demonstrate that ligand-bound wt ERor and ERAE3, but not ERAES, dimerize with the LBD of ERor in solution (Fig. 15A). In experiments with GST-SRC-le fragments, both ERAE3 and ERAES were observed to bind to regions of the coactivator that also bind wt ERor (Fig. ISB). ERAE3 and wt ERor bind the SRC-le fiagments comprising amino acids 579-780 and 989-1240 and do so only in the presence of E2. In contrast, binding of ERAES to the 989-1240 amino acid fragment is constitutive (Fig. 15B). It is previously reported that wt ERor binds corepressors (Smith et al., 1997). In order to further evaluate the relationship between co-regulators and the splicing 67 variants we tested whether ERAE3 and ERAES also possess a corepressor binding function. In vitro binding studies Show that wt ERor, ERAE3, and ERAES share a capacity to bind the corepressor SMRT (Fig. 16). For wt ERa and ERAE3, binding occurs with or without the addition of E2 and in the presence of tamoxifen. The strongest SMRT association is observed in vehicle treated ERAE3 and ERAES samples. 68 Fig. 7. Immunoblot analysis of wt ERor and ERor mRNA splicing variants. Cos7 cells were cultured in phenol red-free DMEM supplemented with 10% calf serum and transiently transfected with 10 pg of pCMV4-derived expression vectors for the indicated ERor isoform. After 48 hours in culture, transfected cells were resuspended in protease inhibitor—supplemented binding buffer and whole-cell extracts were prepared by sonication. Samples containing 20 pg of soluble protein were analyzed by SDS-PAGE and electrophoretically transferred to nitrocellulose filters. Immunoblots were probed with the ERor-specific monoclonal antibody, Mab-l7, obtained from a hybridoma culture supemate that was diluted with an equal volume of PBS. Immunoreactive protein was visualized by chemiluminescence using a horseradish peroxidase-conjugated secondary antibody. The figure is representative of three independent transfection experiments. 69 Fig. 8. Gel mobility shift analysis of ERor mRNA splicing variant DNA-Binding Activity. Cos7 cells were cultured in phenol red-free DMEM supplemented with 10% calf serum and transiently transfected with 10 pg of the pCMV4 expression plasmids containing the indicated ERor cDNAs. Whole-cell extracts were prepared by sonication in protease inhibitor-supplemented binding buffer. For each assay, 8 pl aliquots containing 30 pg of soluble protein were incubated with 6 frnol (40,000 cpm) of a 32P-labeled consensus ERE oligonucleotide. DNA-protein complexes were resolved from fiee DNA probe on a nondenaturing 5% polyacrylamide gel. Confirmation that the indicated band represents an authentic ERG/DNA complex is provided by the ability of an ERor-specific monoclonal antibody (Mab-17) to supershift this complex (compare lane 4 with lane 3). The figure is representative of three independent transfection experiments. Position of the antibody-supershifted complex is indicated by an arrow. 70 Ema; V” .mmrwv mmlm name V eoeoegeeoaeooeve Eu. d M w h m m N. m mdcfidndwnh‘mdmdfla 9090a» %o®¢¢:¢eeo% 71 1250-1 1000- 750- fmollmg proteln 01 O O N 01 O O pCMV4 wt ER ERAE2 ERAE3 ERAE4 ERAES ERAEB ERAE7 Fig. 9. Ligand-binding capacity of ERor and ERor mRNA splicing variants. Ligand binding capacity was assessed by measuring the specific association of 10 nM 3H-1713-estradiol with wt ERor and its splicing variants transiently expressed in Cos7 cells. Cells were transiently transfected with 10 pg of the indicated pCMV4- derived receptor expression vectors and cultured for 48 hours in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum. Cells were resuspended in extraction buffer containing protease inhibitors and sonicated. Aliquots of whole- cell extracts containing 200 pg of soluble protein were incubated overnight at 4°C with 3H-labeled l7B-estradiol in the presence or absence of a ZOO-fold molar excess of unlabeled E2. Free ligand was separated from bound ligand by treatment with dextran-coated charcoal. The figure represents one of three independent experiments. 72 pCMV4 wt ER or ERAE3 Fig. 10. Binding of a fluorescent estrogen analog (nitrile THC) by wt ERor and ERAE3. Cos 7 cells transiently transfected with 10 pg of the wt EROt and ERAE3 expression vectors were plated on glass coverslips and cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum. After 24 hours, cells were stained in culture with 10’7 M nitrile THC. Cells were mounted and photographed using epifluorescence microscopy. Magnification, 400x. 73 Specific Estradiol Binding (% Maximal) Eetredlol Concentratlon (nM) Fig. l 1. Scatchard analysis of E2 binding by wt ERor and ERAE3. Cos7 cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-stripped calf serum and transiently transfected with 10 pg of the indicated pCMV4-derived receptor expression vectors. After 48 hours, cells were resuspended in extraction buffer and sonicated. Aliquots of whole-cell extracts containing 200 pg of soluble protein were incubated overnight at 4°C with various concentrations (0.1 nM - 10 nM) of 3'H-labeled 17B-estradiol in the presence or absence of a ZOO-fold molar excess of unlabeled E2. Free ligand was separated from bound ligand by treatment with dextran-coated charcoal. The measured dissociation constants were 0.66 nM for wt ERor and 0.79 nM for ERAE3. The figure represents one of three independent experiments. 74 Fig. 12. Localization of ERor and ERor splicing variants by confocal microscopy. Cos7 cells transfected with 10 pg of the indicated pCMV4-derived expression vector were plated on glass cover slips and cultured in phenol red-free DMEM supplemented with 5% charcoal stripped calf serum. After 24 hours from transfection, receptor isoforms were detected by indirect immunofluorescence staining using an ERa-specific monoclonal antibody (Mab-l 7) and a rhodamine- conjugated secondary antibody. Irnmunoreactivity was observed with confocal microscopy. The upper left panel shows a representative field from control cells transfected with empty expression vector (pCMV4) displaying minimal nonspecific background. Bar, 10 pm. 75 250T r- 3‘ 200" E Fvehiclej ‘5 ._ IE < 150 - l- < a 0 100-- _, g . . '8 8. 50-- to pCMV4 ERAE2 ERAE3 ERAE4 ERAES ERAEG ERAE7 wt ER Fig. 13. Transcriptional activity of wt ERor and receptor variants on an ERE. HeLa cells were plated at 2 x 10‘6 cells per 100 mm dish and cultured in phenol red- free DMEM supplemented with 5% charcoal-treated calf serum. Cells were cotransfected with 16 pg of reporter (pERE-TK-CAT) and 1 pg of the specified ERor expression vector and treated as indicated with vehicle (0.001% ethanol) or 10 nM 17B-estradiol (E). CAT assays were normalized for equal amounts of protein. Values are expressed relative to vehicle-treated empty expression vector, pCMV4. The figure represents results from one of at least three transfection experiments. 77 Fig. 14. Effect of ERAE3 and ERAES on wt ERa transcription activity. HeLa cells were plated (2 x 10'6 cells per 100 mm dish) and cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum. Cells were cotransfected with 16 pg of pERE-TK-CAT reporter gene, 1 pg of wt ERor expression vector, and increasing amounts of expression vectors for ERAE3 or ERAES (from O to 20 pg). The ratios of wt ERor to variant expression plasmid used in each transfection are indicated. The total amount of DNA in each transfection was held constant with the addition of empty expression vector, pCMV4. CAT assays were normalized for equal amounts of protein. For each of the panels, values are expressed relative to the maximal CAT expression induced by 17B-estradiol treatment in the absence of coexpressed ERAE3 or ERAES. 78 % Maximal CAT Activity 100‘ 40- 50.. 410-- 100? 80:- 60-!- 20:. ERAE3 uvehlcle leetradlel 1:25 1:10 wt ERor:Variant 79 1 :20 Fig. 15. In vitro binding of receptor variants with GST-fused wt ERa and SRC-le fragments. A, In vitro translated 35S-methionine-labeled wt ERa, ERAE3 and ERAES were incubated with bacterial expressed GST or GST fusion protein expressing the ERor LBD (GST-LBD) bound to glutathione-Sepharose beads in NETN buffer supplemented with protease inhibitors. Binding was studied in the absence or presence of 17B-est1adiol as indicated. Bound proteins were separated on a discontinuous 10% polyacrylarnide SDS-PAGE gel and visualized by autoradiography. The top panel represents 10% of the radiolabeled input. The figure is representative of one of three independent experiments. B, In vitro translated 35S-methionine-labeled wt ERa, ERAE3 and ERAES were incubated with GST fusion proteins expressing SRC-le fragments which included amino acids 570- 780 (lanes 1 and 2), 781-988 (lanes 3 and 4), 989-1240 (lanes 5 and 6) and 1241- 1399 (lanes 7 and 8). 80 1/10 Input: ,3? g 6' 47117,}; ‘ t m"'-“.'.'. rr.‘.'v Ma;.‘.rrrvut.’.n- v. - . . .7 GST-LBD Pull-down: ERAE3 eases thRa 58m”? 5‘. * t ' l». ‘o . e . '.t “'1' ' .. , ,' v ‘ qr. . , .‘ ‘ P ’ ‘ - e . .,.»~v, . I,r.r . , . . __' . -1... _, . .. ‘-,. . , '.‘.v .3. _,.-. , _~ .« ._. y .. . ‘ 4 B GST-SRC1 Pull-down: (570-780) (781-988) (989-1 240) (1241-1399) Estredlol - g- - + - + - + 95"‘Ai/‘P .' 1,1. 1 .. m? “ » i. thRor “ ' .- m V‘, 3rd: ”e ‘4. ,_- ,-.:‘..-.'-.‘.a. ,-.~_‘x..'¥ ' ..'..’ _ f".-. . Luv-’3“ l . xv r. ,. . «an m ‘ ‘ ‘ *' ' “ “ “W New. ? ERAES ~. . ._ _ . » - a. \ mut- an». "Lafissfs‘vrniniwk‘u - ' - 81 1/10 input N “ LIB—Tan x_E_Ta_m GST SMRT ERAE3 ERAES v E Tam v E Tam s s, GST SMRT a, Fig. 16. In vitro binding of receptor variants with GST-fused SMRT. In vitro translated 3SS-methionine-labeled wt ERor, ERAE3 and ERAES were incubated with bacterial expressed GST or a GST fusion protein expressing SMRT (GST-SMRT) bound to glutathione-Sepharose beads in NETN buffer supplemented with protease inhibitors. Binding was studied in the absence (V) or presence of 17B-estradiol (E) or tamoxifen (Tam) as indicated. Bound proteins were separated on a discontinuous 10% polyacrylamide SDS-PAGE gel and visualized by autoradiography. The figure represents one of three independent experiments. 82 3. Discussion Our efforts to functionally characterize exon-skipped ERor mRNA splicing variants have identified two receptor isoforms that possess the ability to modulate ERor activity on an ERE. Although their protein structure is significantly altered, the ERAE3 and ERAES splicing variants retain many of the activities attributed to the full-length receptor. Loss of exon 3 results in a receptor protein with an internal deletion that lacks a major portion of the DBD and therefore prevents ERAE3 from binding to a consensus ERE, as confirmed by gel mobility shift analysis. However, ERAE3 retains the LBD and NLS, thereby allowing it to bind hormone with an affinity similar to wt ERor and translocate to the nucleus. The deletion of exon 5 causes a frame-shift mutation and results in a C- tenninally truncated form of the receptor. Loss of the LBD predictably renders ERAES unable to bind E2. Nonetheless, ERAES still retains the NLS, and immunofluorescence analysis shows nuclear staining in Cos7 cells transfected with this variant. The receptor isoforms resulting from deletion of exons 2, 5, 6, and 7 do not bind ligand. Like ERAES, ERAE2, ERAE6, and ERAE7 are C-terminally truncated receptor isoforms that are missing all (ERAE2) or most of the LBD (ERAE6 and ERAE7) and many of the residues critical for ligand binding. The loss of exon 4 causes an infrarne deletion within the receptor that leaves the C-terminus and much of the LBD intact; however, this variant does not bind ligand. Without exon 4 this variant is missing 112 central amino acids that contain 83 the NLS and encompass helices 1, 3 and 4 which comprise the proximal portion of the LBD (Fig. 17). Furthermore, helix 3 makes a number of close contacts with ring A of 17b-estradiol, including Glu353 which forms a direct hydrogen bond with the phenolic hydroxyl group. Exon 4 therefore appears to be crucial for formation of a functional estrogen-binding pocket. While it is unknown what impact the loss of exon 4-encoded helices has on the conformation of the remaining portion of the LBD (helices 5 through 12), it is likely that the overall structure of the LBD remnant is severely distorted. The same probably holds true for ERAES, ERAE6, and ERAE7 truncation mutants. 84 Fig. 17. A depiction of the secondary structure of the ERor ligand binding domain. The ERor ligand binding domain comprises helices 1 through 12. Labeled arrows specify exon junctions. Ligand contacts are indicated in helices 3, 6, 8, and 11. Sites for receptor dirnerization and coactivator contacts are also shown (black and hatched bars, respectively). 85 88: 5 252.0: J1 :1... . « a +5.... a 11.x u a a . :1... m - 10.... m i h--. 96: «B .5620: a ..... v.33. III... a 19.x m , zo=x q s _ a: 1e=x m N 88: c: o: I 12.x .3 NH | :2... 2 In..." an . d .625 3:808 98: NE .5520: I 2335.0: 2.1231 a 33.222 02.8."? 86 Rather than serving to stimulate transcription on a consensus ERE, results from transient transfecion experiments in HeLa cells that combine either ERAE3 or ERAES with wt ERor and an ERE-driven reporter gene indicate that these isoforms actually function to inhibit transcriptional activation by wt ERa. The dominant negative character of ERAE3 and ERAES suggests that, like wt ERor, these variants are able to interact with at least one component of the ERE-directed transcription complex in a manner that disrupts positive gene regulation by wt ERor. Based on gel mobility shift assay analysis, it is unlikely that transcriptional interference by these variants involves binding to an ERE to the exclusion of wt Ear. Our DNA binding analysis indicates that ERAES can bind only weakly to DNA, and only when the formation of this complex is stabilized by the addition of a bivalent antibody. The role of the antibody in this case is presumably to substitute for the missing dimerization interface and to tether the receptor subunits together in a form more able to interact with DNA. DNA binding by ERAE7 similarly requires the addition of antibody, but this binding is even less efficient than binding by ERAES. The relevance of ERE binding by ERAE7 is further reduced with the consideration that ERAE7 does not translocate to the nucleus. Interestingly, a correlation exists among the ERa variants between their ability to translocate to the nucleus and their transcriptional inhibitory effect on wt ERor activity in mammalian cells. As of yet, no clear function has been established for the ERAE7 variant in mammalian cells, despite an earlier report that ERAE7 is a dominant inhibitor of wt ERa function in 87 yeast (Fuqua etal., .1992). This is noteworthy since a number of quantitative studies indicate that ERAE7 generally represents the most abundant of the ERor splicing variants in breast tumors (summarized in Murphy et al, 1997). It has previously been reported that ERAE3 can prevent wt ERor fiom binding to DNA (Miksicek et al., 1993). That ERAE3 inhibits both DNA complex formation and transactivation by wt ERa suggests that the potential targets of interaction by ERAE3 may include contact with the receptor or competitive interaction with coactivators or other receptor-associated factors. ERor function may be disrupted when ERAE3, which lacks the DBD but retains the hormone-inducible dirnerization domain, forms mixed dimers with wt ERor that are inefficient at binding stably to DNA. We are able to show that, in the presence of E2, ERAE3 (but not ERAES) can form a stable complex with the LBD of ERor fused to GST attached to glutathione-Sepharose beads. This is consistent with a model for direct inhibition of the DNA binding activity of the full-length receptor by ERAE3. Experiments using fiagments from SRC-le fused to GST indicate that both ERAE3 and ERAES can bind a nuclear receptor coactivator. Similar to the pattern of wt ERor interaction with SRC-le, in vitro translated ERAE3 is able to associate in an Ez-dependent manner with two regions of the steroid coactivator SRC-le (amino acids 570-780 and 989-1240). This agrees with previous reports that describe three conserved nuclear receptor-binding motifs (LXXLL) within the 570-780 amino acid region and a distinct site for AF] interaction within the 989-1240 amino acid 88 fiagment (Kalkhoven et al, 1998; Webb et al., 1998). The major site for SRC-l interaction with ERa corresponds to the AF 2 domain (Feng et al., 1998), a region that is retained only in the ERAE3 variant. Isoforrns of SRC-l are potent enhancers of agonist-bound ERor and are required for its full transcriptional activity in viva (Ofiate et al., 1995; Xu et al., 1998). Transfection experiments in Ez-treated HeLa cell cultures demonstrate that coexpression of mutants containing the C-terminus of ERor can attenuate ERor-dependent gene expression and that this decreased activity can be overcome with simultaneous over expression of the SRC-l-related coactivator TIF2 (V oegel et al., 1996). These results suggest that coactivators are limiting factors for which the receptors are competing and that ERAE3, like wt ERor, is a target for SRC-l binding. In a surprising result fi'om cotransfection studies using engineered mutants of ERor, maximal expression of an ERE-containing reporter gene could be observed when SRC-l was transfected simultaneously with separate N- and C-tenninal fragments of ERor, containing the AF l/DBD and the LBD/AF 2 regions, respectively (McInery et al., 1996). These results suggest that separate AF 1- and AF 2-containing ERa polypeptides can interact in a transcriptionally productive manner, provided they are brought together by SRC-l. Furthermore, they provide an initial indication that SRC-l interacts separately and perhaps directly with both the AF 1 and AF 2 domains. More support for this notion is provided by our observations that ERAES binds the SRC-le amino acid fragment 989-1240 in solution. This result suggests 89 the possibility that the inhibitory function of ERAES, which itself is relatively inefficient at binding DNA or activating transcription through an ERE, most likely results from competition with wt ERor for interaction with SRC-l. ERAE3, ERAES and wt ERor are also found to interact with the corepressor SMRT. In agreement with previously published results, we find that wt ERor binds the SMRT corepressor in a ligand-independent manner (Smith et al., 1997). Results fiom in vitro binding studies Show a similar degree of wt ERor and SMRT binding in the absence or presence of E2 or tamoxifen. Although ERAE3 also binds SMRT in the presence of tamoxifen and E2, increased binding was consistently observed in vehicle treated samples. These results indicate that the interaction between ERAE3 and corepressor may be enhanced in cells when ligand is unavailable. Thus far there is little reported evidence to suggest what consequence corepressor binding by wt ERor or ERor splicing variants may have on modulating transcription. However, it is conceivable that the activity of some promoters may be affected when ERAE3 or ERAES bind corepressor and thus make it less available to associate with other nuclear receptors. The predicted effect would be a positive influence on corepressor-sensitive promoters that are not directly activated by ERAE3 or ERAES. For example, ERAE3 and ERAES are likely to interfere with basal repression of promoters by TR or RAR, which is effected by receptor association with corepressors in the absence of ligand (Chen et al., 1995; Horlein et al., 1995). Functional analysis of ERor exon-skipped splicing variants shows that, while 90 several of the isoforms are functionally incapacitated by their deletions, two of the variants clearly retain activities assigned to the full-length receptor. The ERAE3 and ERAES variants represent stable receptor isoforms that, like wt ERor, localize efficiently to the nucleus where they may interact with proteins known to associate with the transcription apparatus. However, when acting through a consensus ERE, these variants completely lack (ERAE3) or show only weak (ERAES) transcriptional stimulatory activity, consistent with their poor DNA binding ability. On the contrary, both variants serve to inhibit the ability of coexpressed wt ERor to promote transcription of ERE-containing genes. At the same time, the ability of ERAE3 (and presumably also ERAES) to interact productively with nuclear receptor coactivators or corepressors gives these ERor splicing variants the potential to stimulate or otherwise modulate gene expression. 91 IV. ERor Splicing Variants Lacking Exons 3 or 5 Display Promoter- Specific Transcriptional Effects Through Nononsensus ERES 1. Introduction A wide variety of variant ERor mRNAs are recognized in the literature (Dowsett et al., 1997; Fuqua et al., 1993; Murphy et al., 1997; van Dijk etal., 1997). Those most commonly observed harbor a precise deletion of one of the internal exons that contribute to the structure of the full-length ERor protein. Despite their discovery nearly a decade ago (Fuqua et al., 1991; Wang and Miksicek, 1991), relatively little is known about the functional role of ERa splicing variants. The most prominent activity described for two of the splicing variants (ERAE3 and ERAES) is to interfere with the transcriptional activity of wt ERor on ERE-containing genes, where they behave as dominant-negative mutants (see Fig. 14). The inhibitory activities of ERAE3 and ERAES are likely to result in part from their ability to sequester nuclear receptor coactivators or other limiting transcription factors into a non-productive complex. In addition, ERAE3 may block binding of wt ERor to DNA by forming functionally inactive, mixed dimers (see Fig. 15A, Wang and Miksicek, 1991). The in-frame deletion of exon III harbored by ERAE3 causes partial loss of its DNA-binding domain DBD, but leaves the LBD intact. Predictably, ERAE3 does not bind an ERE; yet its ability to bind and potentially respond to hormones is 92 maintained. Conversely, the LBD is severely truncated in ERAES resulting in the complete loss of hormone binding. Although ERAES retains the DBD, its DNA- binding activity is impaired as a consequence of the loss of helix 11 within the LBD, which contains critical contact sites for subunit dirnerization (Brzozowski et al., 1997 ; Kumar and Chambon, 1988). While the ERAES splicing variant is reported to display modest transcriptional activity in certain cell- and promoter-specific contexts (Chaidarun etal., 1998; Fuqua et al., 1991; ), its relatively poor binding to the palindromic ERE suggests that it is unlikely to be a significant effector of transcription through a consensus ERE. Similarly, the remaining exon-skipped ERor splicing variants (ERAE2, ERAE3, ERAE4, ERAE6, and ERAE7) appear to completely lack transcriptional stimulatory activity when tested in transient transfection assays with an ERE-containing reporter plasmid (Fig. 13). In the studies detailed below, ERAE3 and ERAES splicing variants receive significant attention for the reason that these receptor isoforms retain some of the functional characteristics of wt ERor. Most notably, despite their mutations, they localize to the nucleus, associate with the steroid receptor coactivator SRC-le, and ultimately are observed to modulate gene transcription (see Figs. 12, 14, 15, and below). In recent years, a novel pathway for regulation of transcription by estrogen receptors (both ERor and ERB) has been described that involves cooperation of the receptors with the AP-l transcription factors, c-jun and c-fos (Gaub et al., 1990; Uht et al., 1997). This has been broadened to include additional genes for which estrogen regulation was mapped to binding sites for the Spl (Duan et al., 1998) and 93 ATF2 transcription factors (Sabbah et al., 1999), which bind to GC-rich and CRE (cyclic AMP response element)-like motifs, respectively. An important feature of these nonclassical pathways for estrogen action is that functional domains within the receptor that are crucial for transcriptional activation through a consensus ERE are in some cases dispensable for ERor activity on AP-l, Spl , or CRE-regulated promoters. Mutational analysis revealed that the DBD was not always required for ERor-dependent expression of these genes (Duan et al., 1998; Gaub et al., 1990; Sabbah et al., 1999). Clearly, the mechanisms of ERor transcriptional activity and DNA targeting are complicated by these reports. This led us to consider the possibility that the nuclear ERor splicing variants (specifically ERAE3 and ERAES) may function as transcriptional regulators through noncanonical hormone response elements as opposed to consensus ERES. The following results describe four promoters lacking an ERE that are efficiently stimulated by either ERAE3 or ERAES. In some instances the activity is shared by wt ERor, while in other cases gene activation is limited to the ERAE3 or the ERAES variants alone. The promoters tested in these studies attracted our interest because they are reported to be ERor regulated and/or contain an AP-l motif. They include the chicken ovalbumin, human collagenase and human IGF-l promoters, and a synthetic promoter consisting of three consensus AP-l elements. 94 2. Results In agreement with previously published reports (Wang and Miksicek, 1991; Rea and Parker, 1996), Fig. 13 shows that ERor splicing variants do not effectively promote gene expression from a palindromic ERE . However, this understanding does not preclude the possibility that transcriptional effects of ERa variants may be directed at promoters with nonclassical hormone response elements. To further investigate the role of ERor splicing variants in the regulation of gene expression, we used a cell culture transfection system to assay for receptor activity on a variety of reporter gene constructs that may be targets for regulation by ERor even though they lack a consensus ERE. For preliminary studies, HeLa cells were transfected with each of these reporters together with wt ERor or variant ERor expression plasmids. HeLa cells express minimal endogenous ERor and yet possess a transcriptional milieu that enables reporter plasmids to respond efficiently to transfected receptor. Paired cultures were treated with 10 nM E2 in combination with 20 nM PMA, or with vehicle alone. This was done to ensure that an activating ligand was present, if necessary, and that AP-l factors were maximally stimulated in the event that promoter activation required stimulation of PKC. 95 2.1 Like wt ERa, ERAE3 activates the ovalbumin gene promoter Although there is no consensus ERE in the ovalbumin promoter, it has been shown that this gene is regulated by wt ERor. Mapping of the ERa regulatory element places it at a critical AP-l site near the transcription start site (Schweers et al., 1990; Tora et al., 1988). We performed cotransfection experiments in HeLa cells using vectors expressing wt ERor or the exon-skipped variants ERAE2 through ERAE7 and a CAT reporter gene construct, vaalb-CAT driven by a fragment of the ovalbumin promoter (-1342 to +7 relative to transcription start site) described to encompass much of the regulatory sequence of this gene (Schweers et al., 1990; Tora et al., 1988). Results from these experiments indicate that both wt ERor and ERAE3 support inducible gene expression from the ovalbumin promoter (Fig. 18). The remaining single exon-skipped variants are transcriptionally inactive on this reporter construct (Fig. 18, inset). For wt ERa, this corroborates previously published reports (Gaub et al., 1990; Tora et al., 1988). Maximal activity was measured in cultures treated with both phorbol 12-myristate, 13-acetate (PMA, a phorbol ester) and E2, where a 16-fold induction was observed. Like wt ERor, ERAE3 reproducibly induced this reporter, despite its lack of an intact DBD. While the induction by ERAE3 (averaging 9-fold) is less than that supported by wt ERa, the activity of ERAE3 equaled and occasionally exceeded that of the intact receptor in several individual experiments, confirming that this variant can be a potent inducer of transcription (Fig. 18). In both cases cotreatrnent with PMA and E2 is 96 highly synergistic as E2 treatment alone has no significant effect and PMA treatment alone supports only modest induction for wt ERor (2.5-fold relative to vehicle control). Tamoxifen treatment of wt ERor- or ERAE3 -transfected cultures, either alone or together with phorbol ester, also had no significant effect on vaalb-CAT expression. This contrasts with the stimulatory activity of tamoxifen observed on other AP-l containing estrogen-responsive reporter genes (Webb et al., 1995). In control cells transfected with an empty CMV expression vector, treatment with PMA yielded negligible reporter gene activity. This suggests that, in the absence of ERor, activation of endogenous AP-l alone is not an effective inducer of transcription from the ovalbumin promoter in these cells. To confirm that wt ERor and ERAE3 cooperate with AP-l factors to regulate transactivation of the ovalbumin promoter, we measured vaalb-CAT expression in HeLa cells cotransfected with both a receptor isoform and a c-jun expression vector. Transcriptional activity of wt ERor and ERAE3 supported by PMA and E2 cotreatrnent was enhanced by c-jun over expression. While the presence of endogenous AP-l tended to obscure the synergism between c-jun and wt ERor in this system, the combined effects of these transcription factors were slightly more than additive. Enhanced activation was also observed when c-jun was coexpressed with ERAE3 (Fig. 19). Exogenous c-jun alone elicited only a modest response to PMA and E2 treatment. These observations, combined with the dual requirement for activation of both AP-l and ERa, strongly suggest that these factors are acting cooperatively on the ovalbumin 97 promoter. Further consideration of the mechanism of activation by wt ERor and ERAE3 excludes a requirement for direct binding of receptor to the ovalbumin promoter. ERAE3 is devoid of DNA-binding activity, arguing that activation of ovalbumin by these receptors must be mediated indirectly through specific protein- protein interactions. Figure 20 demonstrates that ERor interacts with c-jun and c-fos in solution and suggests that the receptor may be recruited to the ovalbumin promoter by interacting with AP-l factors. Binding assays with GST-fused ERor and 35S-labeled c-jun and c-fos (translated individually or in combination in vitro) demonstrate that, independent of E2 treatment, ERor binds c-jun and c-fos under all treatment conditions tested. Although ERa variants were not specifically included in these binding studies, Webb and coworkers have reported an interaction between c-jun and the isolated N-terminus of ERot, a region conserved in both ERAE3 and ERAE5 (Webb et al., 1995). We therefore infer that ERAE3 and ERAE5, like their intact counterpart, are also able to interact with AP-l homo- and heterodimers. 2.2 Both ERAE3 and ERAE5 activate the Coll(-73)Luc reporter The promoter of the human collagenase gene has received considerable attention as a target for nonclassical regulation by both ERor and ERB. Collagenase is a member of a family of matrix metalloproteases, which are regulated similarly and demonstrate enhanced expression in response to injury and during inflammatory reactions (Angel et al., 1986; Herrlich et al., 1986; Whitharn et al., 1986). 98 Collagenase gene expression is elevated in certain tumor cells and may contribute to advancement of metastasis (Liotta 1986). In cultured fibroblasts and skin cells treated with carcinogens and tumor promoting agents, induction of collagenase is mediated by phorbol ester treatment (Angel et al., 1986; Angel et al., 1985). A short region of the collagenase promoter (-73 to +63 relative to the transcription start site) which harbors an AP-l element is reported to direct estrogen- or tamoxifen- regulated gene expression of a luciferase reporter gene in a variety of cell lines (Paech et al., 1997; Uht et al., 1997 ; Webb et al., 1995). In contrast to published reports, we find that wt ERor behaves no differently on the pColl(-73)Luc reporter than the empty pCMV4 control vector (Fig. 21). In both cases, a relatively modest (approximately 6-fold) increase is observed that depends on activation of endogenous AP-l by PMA treatment. This same promoter is also activated by ERAE3 and to a lesser extent by ERAE5 in response to combined treatment with estrogen and PMA (Fig. 21). Like wt ERor, the remaining splicing variants (ERAE2, ERAE4, ERAE6, and ERAE7) were without significant effect. Substantiating the necessity for PMA treatment and the role of AP-l in receptor regulation of the collagenase promoter, coexpression of c-jun is observed to enhance ERAE3- and ERAES- induced pColl(-73)Luc reporter gene expression (Fig. 22). Additional analysis indicates that maximal activation of pColl(-73)Luc by ERAE3 requires cotreatrnent with both E2 and PMA (Fig. 23). Phorbol ester alone (or in combination with ER ligands) supports a modest induction of the collagenase reporter that is independent of coexpressed receptor. Regulation pColl(-73)Luc by 99 ERAE5 similarly required activation of endogenous AP-l by PMA, but was independent of E2 treatment, consistent with the inability of ERAE5 to bind ligand. The partial agonist tamoxifen was without effect on induction of this reporter by ERAE3, either alone or in the presence of PMA. Treatment with E2 or tamoxifen alone or together with PMA had no effect on the activity of pColl(-73)Luc cotransfected with wt ERor (or pCMV 4) compared to PMA alone (Fig. 23, inset). The human collagenase promoter therefore demonstrates a difference in behavior compared to the vaalb-CAT reporter which was not induced by ERAE5, but was activated by both ERAE3 and wt ERor in cells cotreated with E2 and PMA. Due to the stimulatory behavior of ERAE3 and ERAE5 on the ovalbumin and collagenase promoters, both of which contain AP-l sites in their native promoter contexts, it was of interest to examine the effects of the various ERor splicing variants on a reporter plasmid containing an AP-l site in the context of a promoter not otherwise regulated by ERor or estrogens. For this purpose, we used p(AP-1)3- TK-CAT, which contains three consensus AP-l sites in tandem upstream of a minimal thymidine kinase promoter from Herpes simplex virus. Somewhat unexpectedly, ERAE5 (but not wt ERa, ERAE3, or any of the remaining splicing variants) was able to induce this promoter approximately 7-fold in HeLa cells (Fig. 24) although this difference failed to reach statistical significance (P S 0.1). Like the pColl(-73)Luc reporter described above, ERAE5 activity on the p(AP-1)3TK- CAT reporter required PMA treatment and did not depend on the presence of E2 100 (Fig. 24, inset). It is clear that by some reports ERa is an estrogen-dependent inducer of transcription through a consensus AP-l element (Gaub et al., 1990; Webb et al., 1999). These results demonstrate that the activity of ERor on AP-l containing promoters is a complicated and conditional effect. The data presented here agree well with a previous report demonstrating that a C-terminally truncated receptor lacking the LBD is more active than E2-liganded wt ERor on an analogous AP-lfl" K reporter construct (Uht et al., 1997). Also, it is evident that the context of an AP-l site within the promoter is critical for conferring its ability to respond, and the degree of that response, to wt ERor or any of the ERor splicing variants. 2.3 ERAE5 activates the human IGF-l promoter Insulin-like grth factor I (IGF-l) is expressed in and effects cellular growth in many tissues. In general, growth hormone acts as the major regulatory factor for the IGF-l gene; however, several other factors (including thyroid hormone, epidermal grth factor, parathyroid hormone and estrogen) function to modulate IGF -1 expression (Ernst et al., 1989; McCarthy et al., 1989; Murphy et al., 1990; Stewart et al., 1996; Wolf et al., 1989). Induction of IGF -1 is stimulated by E2 in the uterus of ovariectomized / hypophysectomized rats (Murphy et al., 1990; Murphy et al., 1988) and in cultured rat osteoblast cells (Ernst et al., 1989). In transient cotransfection studies a defined region of the chicken IGF-l promoter directed reporter gene induction by liganded ER (Umayahara et al., 1994). Like the ovalbumin and collagenase promoters mentioned above, the IGF-l promoter lacks a 101 consensus ERE sequence (Kim et al., 1991; Hall et al., 1992). Further complicating this picture are independent data showing that in primary fetal rat osteoblasts E2 can block the increase in IGF-l synthesis caused by parathyroid hormone (PTH) or prostaglandin E2 (PGE2) (McCarthy et al., 1997). It has been shown that this hormonal regulation of lGF-l expression by PTH and PGE2 is mediated by a binding site for CCAAT/enhancer-binding protein-delta (C/EBP-fi) located between positions +202 and +209 within exon 1 of this gene (Umayahara et al., 1999). For the chicken promoter, a positive ERor regulatory element was mapped to an AP-l motif essential for both E2 and phorbol ester-stimulated gene transcription (Umayahara etal., 1994). Although there is a great deal of homology among IGF -1 promoters of various species, this AP-l site is not conserved in mammals (Hall et al., 1992; Kim et al., 1991; Umayahara et al., 1994). The human IGF-l promoter is thus different from the promoter examples described above in that it lacks any consensus binding sites for either AP-l or Spl that might represent likely targets for regulation by ERor. However, Nagaoka and colleagues report that PKC activation by PMA treatment increases the rate of IGF-I transcription in the human macrophage-like cell line U937 (Nagaoka et al., 1990). This suggests that AP-l factors or another PKC-responsive transcription factor positively regulate the human [GP -1 gene. Furthermore, they may be functioning at a nonconsensus AP-l or ATF2 binding site. When cotransfected into HeLa cells, treatment with PMA induced expression of a luciferase reporter plasmid containing 1.63 kb of sequence upstream 102 from the human IGF-l transcription start site (pIGF1-1630-Luc) (Fig. 25). This same reporter was unresponsive to wt ERor transfection. IGF-l promoter activity is strongly and constitutively induced by ERAE5, greater than 30-fold compared to cells transfected with control plasmid. PMA treatment of ERAE5 transfected cells has no additional stimulatory effect on ERAE5 induction of this reporter gene. In fact, PMA treatment slightly decreases ERAE5 activity (Fig 25). This argues against the involvement of PKC or PKC-activated transcription factors such as AP-l in the mechanism of ERAE5 induction of human IGF-l expression. In an attempt to determine where within the IGF-l promoter the ERAE5 regulation is directed, reporter gene transfection assays were performed with a series of 5'-deletion mutants of the human IGF-l promoter. ERAE5 induced expression from reporter gene constructs containing -1630, -926, -592, and -235 bp (the number reflects nucleotides relative to the transcription start site) of the human IGF -1 promoter (Fig. 26). However, induction of the shortest length of promoter tested (the -235 bp fragment) was significantly less compared to the others. Unexpectedly, ERAE5 was most active on the -592 bp fragment. These results indicate that repressor elements may reside in the human IGF -1 promoter between -l630 and -592 and that a highly responsive ERAE5 enhancer element is located between nucleotides -592 and -235. It should be noted, however, that overall activity was progressively lost with each truncation of the IGF -1 promoter (Fig. 26, inset). Although its induction by ERAE5 was less than that observed for other promoter lengths, the -1630 bp promoter 103 supported maximal reporter gene expression . Efforts to transfer ERAE5- responsiveness onto a heterologous promoter were unsuccessful, suggesting that activation of IGF-1 expression by ERAE5 may require complex interactions between transcription factor binding sites throughout the IGF-l promoter. 104 Fig. 18. Activation of the ovalbumin gene promoter by wt ERor and ERAE3. HeLa cells were plated (2 x 10" cells per 100 mm dish) and cultured in phenol red- free DMEM supplemented with 5% charcoal-treated calf serum. Cells were cotransfected with 16 pg of reporter (vaalb-CAT) and 1 pg of the specified ERor expression vector. Cultures were treated as indicated: vehicle control (0.001% ethanol and 0.002% DMSO); 20 nM phorbol 12-myristate, 13-acetate (PMA); 10 nM WIS-estradiol (E); E + PMA; and 100 nM tamoxifen (T); or T + PMA. CAT assays were normalized for equal amounts of protein. Values are expressed relative to vehicle-treated empty expression vector, pCMV4. Error bars represent the SEM of three independent transfection experiments (*, P < 0.001). Inset, a transient transfection reporter gene assay was used to test the activity of each of the single exon-skipped variants on vaalb-CAT. 105 Relative CAT Activity 20- 15- 10- D 0 d O Ivehicle '7 ’ DE+PMA 1 Relative CAT Activity 0 J a poll“ [M32 IME: [M24 ENE! 3M3. INLET M In .vehicle IPMA EJE IE+PMA UT T-«i-PMA P < 0.001 106 4o-- — 30 .. lvehicle P<0-05 DE-i-PMA I I. 2... I l 10-- * I 0._£.~_£4.j+‘— L| Relative CAT Activity ERAE3 "’ " + + - " thRa _ _ — _ + 1" cJun - + " + " + Fig. 19. Effect of c-jun over expression on wt ERor and ERAE3 activation of the ovalbumin promoter. HeLa cells were plated (2 x 10* cells per 100 mm dish) and cultured in phenol red-fiee DMEM supplemented with 5% charcoal-treated calf serum. Cells were cotransfected with 16 pg of reporter (pERE-TK-CAT), 1 pg of the specified ERor expression vector and 2 pg of c-jun expression vector where indicated. Cells were treated with vehicle or 10 nM l7B-estradiol and 20 nM phorbol lZ-myristate, 13-acetate (E + PMA). CAT assays were normalized for equal amounts of protein. Values are expressed relative to vehicle-treated empty expression vector, pCMV4. Error bars represent the SEM of four independent experiments (*, P < 0.05; ns, not significant). 107 1110 input: GST-Elia Pull-down: .- + II + ambl W “a? Al I'? . , ,1 .3 "are; ‘ s . “.qemnv- n—vb- edun . '. .-‘s<-;..s;.ss:‘s. “Zia . Q1; c-fos W'- e a .. I 3 4 firms-hits .I" ' 2:1: I' . '. c40n -i- c-foe .— . . ‘4'7I“.“".‘§_‘F. ., 1 2 GST GST-ER Fig. 20. Binding of c-jun and c-fos with GST-fused wt ERa. Right panel, in vitro translated 35S-methionine-labeled c-jun and c-fos were incubated independently and in combination (c-jun + c-fos) with bacterial expressed GST (lanes 1 and 2) or GST-ERor (a GST fusion protein expressing wt ERor, lanes 3 and 4). GST proteins were bound to glutathione-Sepharose beads in NETN buffer supplemented with protease inhibitors. Binding was studied in the absence or presence of 17B-estradiol as indicated. Bound proteins were separated on a discontinuous 10% polyacrylarrride SDS-PAGE gel and visualized by autoradiography. Left panel shows 10% of the radiolabeled input. The figure represents results from one of three independent experiments. 108 P5 0.1 80 T T g. 60... ~- .2 ‘6 < Psoas i ' a 40... l , '8 I vehicle 3 r: E-i-PMA m Pseos .2 l E g 20-- pCMV4 ERAE2 ERAE3 ERAE4 ERAES ERAEB ERAE7 wt ERG Fig. 21. Activity of ERa splicing variants on a cotransfected human collagenase reporter plasmid. HeLa cells were plated at 1 x 10‘6 cells per 60 mm dish and transiently transfected with 10 pg of the pColl(-73)Luc reporter gene and 0.5 pg of receptor expression vector. Cells were cultured in phenol red-lice DMEM supplemented with 5% charcoal-heated calf serum and treated with vehicle or 10 nM WIS-estradiol and 20 nM phorbol 12-myristate, l3-acetate (E + PMA). Luciferase activities were normalized for protein and results are eXpressed relative to vehicle-treated empty expression vector (pCMV4) controls. Error bars represent the SEM of three independent transfection experiments. 109 r P 5' 0'01 lIIDCMWI—P-é—Oi-I (poiiivnlféil.| chm 300 [ f I 3. P g 0.1 E 250 .. l'_ 'l E .T. g 200 1- 9 £3 nvehicle 8 15° " o E+PMA ._l '22, 100 «- E Q) CE 50 .. o . I pCMV4 c-jun ERAE3 ERAEa ERAE5 ERAE5 thFia thRa + c-jun + c-jun + c-jun Fig. 22. Effect of c-jun coexpression on ERAE3 and ERAE5 transactivation of the pColl(-73)luc reporter gene. HeLa cells were plated (l x 10'6 cells per 60 mm dish) and transiently transfected with 10 pg of reporter gene, 0.5 pg of receptor expression vector, and 2 pg c-jun expression vector where indicated. Cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum and treated with vehicle or 10 nM 17B-estradiol and 20 nM phorbol 12- myristate, 13-acetate (E + PMA). Luciferase activities were normalized for protein and results are expressed relative to vehicle-treated empty expression vector (pCMV4) controls. Error bars represent the SEM of three independent transfection experiments. 110 Fig. 23. Induction of the pColl(-73)Luc reporter gene by ERAE3 requires 1713- estradiol and PMA cotreatment. HeLa cells were plated at 1 x 10‘6 cells per 60 mm dish and transiently transfected with 10 pg of reporter gene and 0.5 pg of receptor expression vector. Cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum and treated as indicated: vehicle control; 20 nM phorbol 12-myristate, 13-acetate (PMA); 10 nM 17B-estradiol (E); E + PMA; and 100 nM tamoxifen (T); or T + PMA. CAT assays were normalized for equal amounts of protein. Values are expressed relative to vehicle-treated empty expression vector, pCMV4. Luciferase activities were normalized for protein and results are expressed relative to vehicle-treated empty expression vector (pCMV4) controls. Error bars represent the SEM of three independent experiments. The indicated significance C“, P 50.1; **, P _< 0. 05) is relative to corresponding vehicle control. Inset, a representative transient transfection assay was used to determine the transcriptional activity of wt ERa on the pColl(-73)Luc reporter gene in the presence of tamoxifen. lll MEI-'1 pCMV4 25 V 2.3.3 823.2... 2:23. 100 1- T+PMA uE-i-PMA IT Ivehlcle DE BPMA 0.- .1 u .r 0 0 0 0 8 6 4 2 >=>=o< 03.2.03 5...»..me .x. Fig. 24. Transcriptional activity of ERor splicing variants on the p(APl)3TK- CAT reporter plasmid. HeLa cells were plated at 1 x 10“5 cells per 60 mm dish and transiently transfected with 0.5 pg of pCMV4 vectors expressing the indicated receptor isoforms along with 10 pg of the p(AP1)3TK-CAT reporter gene. Cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum and treated with vehicle or 10 nM 17B-estradiol and 20 nM phorbol 12- myristate, 13-acetate (E + PMA). CAT assays were normalized for equal amounts of protein. Values are expressed relative to vehicle-treated empty expression vector, pCMV4. Error bars represent the SEM of three independent experiments. Inset, a transient transfection assay was used to determine ligand requirements for ERAE5 activation of the p(AP1)3TK-CAT reporter gene. 113 Relative CAT Activity 33513:: I vehlcle 525152 IT+PMA I vehicle a E+PMA flflfl pCMV4 ERAE2 ERAE3 ERAE4 ERAE5 ERAE6 ERAE7 wt ERa 114 P5031 iooTI ' P3035 \l 01 l a vehicle I E+PMA %Maxlmum Luciferase Activity N U! 01 O pCMV4 ERAE2 ERAE3 EHAE4 ERAES ERAEG ERAE7 wt ERa Fig. 25. Transcriptional activity of wt ERa and wt ERG. splicing variants on the human lGF-l promoter. HeLa cells were plated at l x 10"5 cells per 60 mm dish and transiently transfected with 0.5 pg of pCMV4 vectors expressing the indicated receptor isoforms along with 10 pg of the IGF -1 promoter driven reporter gene pIGF1-1630-Luc. Cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum and treated with vehicle or 10 nM WIS-estradiol and 20 nM phorbol 12-myristate, 13-acetate (E + PMA). Luciferase activities were normalized for protein and results are expressed relative to vehicle-treated empty expression vector (pCMV 4) controls. Error bars represent the SEM of three independent experiments. 115 Fig. 26. ERAE5 induces the expression of a reporter gene driven by a series of truncated IGF—l promoters. HeLa cells were plated at 1 x 10‘6 cells per 60 mm dish and transiently transfected with 0.5 pg of the pCMV4 derived ERAE5 expression vector and 10 pg of a luciferase reporter gene construct driven by various lengths of the IGF-l promoter. Cells were cultured in phenol red-free DMEM supplemented with 5% charcoal-treated calf serum. The length of [GP -1 promoter used in each luciferase reporter construct is specified by the number of base pairs upstream from the IGF-l transcription start site: -1630, -926, -592 and -235 bp. Luciferase activities were normalized for protein and results are expressed relative to empty expression vector (pCMV4) controls for each reporter construct. Error bars represent the SEM of at least three independent experiments. The inset shows absolute promoter activity in control cells cotransfected with the empty pCMV4 expression vector compared to the enhanced activity supported by ERAE5 cotransfection from one representative experiment. 116 ERAE5 [3592 I235 I1630 I926 pCMV4 mgmzcwgwomwzn o W. M. R — I _: ,, m - n p p . p . . u u u .1 . 4 0 6 2 8 4 2 2 1 1 c2520:- Eon. ll7 3. Discussion The traditional view of estrogen action depicts ERa as the central player in target gene selection by virtue of its ability to bind to DNA containing a close match to the ERE consensus sequence (Tremblay et al., 1997). It has been necessary to modify this view to accommodate the newly discovered ERB, which binds to the same palindromic ERE sequence (Chien et al., 1994). Recent evidence also indicates that many estrogen-regulated genes may contain noncanonical hormone response elements organized around AP-l or SP1 sites. Among these targets are genes such as IGF-l, cyclin D1, c-fos, c-myc, RARa, collagenase, and cathepsin D that play important roles in controlling cell grth and tissue remodeling (Duan et al., 1998; Gaub et al., 1990; Krishnan et al., 1994; Pilihowska et al., 1997; Sabbah et al., 1999; Sun et al., 1998). It appears that the structural requirements for ERa to regulate many noncanonical hormone response elements are less stringent than transcriptional stimulation through a consensus ERE (Duan et al., 1998; Gaub et al., 1990; Sabbah et al., 1999). For this reason, we have examined the behavior of ERa splicing variants on several promoter constructs containing noncanonical regulatory elements. In agreement with previously published reports, our studies indicate that wt ERa stimulates ovalbumin promoter activity. The actvity of wt ERa and engineered ERa mutants on this promoter is mapped to a critical AP-l motif which when mutated causes the loss of ERG responsiveness (Gaub et al., 1990; Tora et al., 1988). Maximal activation of the ovalbumin promoter by wt ERG requires estradiol ”8 and phorbol ester co-treatment. This induction by wt ERa is enhanced with c-jun cotransfection. The dual requirement for estradiol and a phorbol ester treatment and the enhanced activity observed with c-jun over expression concurs with promoter mutational studies to suggest that ERa regulation of the ovalbumin promoter is directed by an AP-l motif identified in the upstream region of the promoter (Gaub et al., 1990; Tora et al., 1988). Furthermore, these results also indicate that this regulation involves a cooperative interaction between EROL and AP-l factors. A significant estradiol- and phorbol ester-dependent induction of ovalbumin activity is also supported by ERAE3, despite its lack of an intact DNA-binding domain. While it is evident that the ERAE3 splicing variant is not as active as wt ERa on this promoter, the capacity for ERAE3 to induce the ovalbumin promoter implies that direct DNA binding by the receptor is not required for this regulation. Indeed, induction of this promoter by wt ERa was clearly shown not to require the direct binding of the full-length receptor to the DNA (Paech, et al., 1997). In contrast to the situation with vaalb-CAT, the activity of wt ER): on the remaining three noncanonical response element-containing promoters examined here was either non-existent or insignificant compared to that of the ERAE3 or ERAE5 splicing variants. Specifically, both ERAE3 and ERAE5 effectively enhanced expression of the pColl(-73)Luc reporter, and ERAE5 alone strongly activated pIGF-Luc and p(AP-l)3TK-CAT. The behavior of these reporters in response to the various receptor isoforms was not readily predicted from their 119 known biochemical properties and significant differences are evident for each of the reporters tested. Similar to ERa-directed regulation of the ovalbumin promoter, activity of ERa isoforms on the collagenase promoter can be mapped to an AP-l motif. Induction of pColl(-73)Luc reporter construct, which contains a short region of collagenase promoter including the AP-l motif, by ERAE3 required both estradiol and phorbol ester treatment and ERAE3 induction was enhanced with c-jun over expression. Considering the facts that ERAE3 lacks the DBD, that ERAE3 cooperates with c-jun to maximally activate this promoter, and that c-jun binds the ERa N-terminus (Webb et al., 1995), these results strongly suggest that ERAE3 is recruited to the AP-l motif through protein-protein interactions to activate the collagenase promoter. Whether transcriptional stimulation of reporters containing . noncanonical hormone response elements (such as vaalb-CAT or pColl(-73)Luc) involves a direct interaction between ERa or its isoforms and the AP-l components jun and fos, or an indirect interaction mediated through a bridging factor such as CBP/p300 or SRC-l needs to be further clarified. Additionally, the identities of the downstream targets for activation by phorbol ester, while presumed to involve the PKC pathway, remain to be identified. Potential targets to consider include AP-l, nuclear receptor co-regulators, or the ERa isoforms themselves. Activation of the collagenase promoter construct by ERAE5 in HeLa cells required PMA treatment and was enhanced by c-jun over expression suggesting that 120 ERAE5 cooperates with AP-l factors to regulate this promoter. Induction of the p(AP-1)3TK-CAT reporter gene by ERAE5 also required phorbol ester treatment. In contrast, ERAE5 induction of the IGF-l promoter was not enhanced by phorbol ester treatment. In fact, ERAE5 activity on the IGF-l promoter was slightly attenuated with PMA treatment. This suggests that activation of AP-l , presumably by stimulation of PKC and phosphorylation of AP-l factors (e.g., c-jun and c-fos), plays different roles in the stimulation of these various reporter constructs. These data also suggest that the presence of a functional AF 2 domain is detrimental for activation of some (but not all) noncanonical hormone response elements by ERa since ERAE5, but not wt ERa or ERAE3 stimulated the p(AP-1)3TK-CAT and pIGF(-1630)Luc reporter plasmids. ERAE5 activity on the pColl(-73)Luc, p(AP- 1)3TK—CAT, and pIGF(-1630)Luc reporters showed no estrogen responsiveness, consistent with the inability of this variant to bind ligand. In contrast to the reporter plasmids described above, neither wt EROL, ERAE3, nor ERAE5 stimulated reporter gene expression from a variety of control promoters representing the HSV thymidine kinase or c-Met genes, or the early region of Simian Virus 40. From our studies, we know that wt ERa, ERAE3 and ERAE5 do not transactivate the same promoters. Compared to the activities observed for ERAE3, ERAE5 activity is complicated by results that show that efficient induction of the collagenase and AP-l reporters by this splicing variant requires treatment with phorbol ester, while stimulation of the IGF -1 promoter does not. The disparities 121 observed for the behavior of these promoters in response to the receptor isoforms indicate that the context of the AP-l motif (or other noncanonical ERES) can exert a significant influence on how these promoters respond to ERon and its variants. Although our cotransfection studies serve to identify promoters activated by ERa mRNA splicing variants without directly addressing how they do so, published studies suggest a variety of potential mechanisms that could account for our observations. According to the first of these models, regulation of some noncanonical promoters by wt ERa and ERoc splicing variants may involve a direct protein-protein interaction with other upstream factors such as c-jun and c-fos that in turn act through their cognate DNA-response elements (summarized in Fig. 27). As described above, this appears to be the case for ERAE3 regulation of the collagenase and ovalbumin promoters. Stimulation of transcription by ERAE3 appears to require dual activation by both E; and phorbol ester. Evidence suggests that ERAE3-directed transcription of the collagenase and ovalbumin promoters involves both receptor interaction with AP-l factors and ligand-induced recruitment of transcriptional co-regulators. The role of ERAE3 and ERAE5 in this activation may be to recruit coactivator proteins through their intrinsic activation domains. Two activation functions have been described in wt ERa, AF] is localized to the amino- terminus and is expressed in both ERAE3 and ERAE5. A second estrogen-inducible activation function (AF2) resides within the carboxy-terminal ligand-binding domain and is therefore present in ERAE3, but not in ERAE5. Studies performed 122 using wt ERa demonstrate that both of these activation fimctions correspond to binding sites for various co-regulatory proteins including CBP/p300 or p160 coactivators such as SRC-l (Endoh et al., 1999; Feng et al,. 1998; Kamei et al., 1996; Kobayashi et al., 2000; Metivier et al., 2001; Metivier et al., 2000; Tremblay et al., 1999; Watanabe et al., 2001; Webb et al., 1998). Indeed, we have previously demonstrated that both ERAE3 and ERAE5, like wt ERa, can interact with the nuclear receptor coactivator SRC-l. For ERAE3, this interaction involves at least two SRC-l interaction sites and is dramatically stimulated by ligand. Only the constitutive amino-terminal site (AF 1) is present in ERAE5 (supporting ligand- independent interaction with SRC-l ). Cotransfection and binding studies suggest that ERAE3 and ERAE5 may cooperate with AP-l factors to activate transcription of noncanonical promoters containing an AP-l motif. A study showing increased induction of the pColl(- 73)Luc reporter by an ERa-DBD mutant (which resembles ERAE3) when it is expressed as a VP16 chimera provides evidence that ERAE3 is translocated to an AP-l motif to stimulate transcription (Webb et al., 1995). Yet there is no direct evidence that ERAE5 is physically recruited to the promoters that it regulates. Moreover, the IGF-l promoter does not contain an AP-l motif. Clearly other mechanisms of activation must be considered. If a receptor isoform is not located to the site of transcription initiation perhaps it effects transcription by modulating the activity or expression of other transcription factors (see Fig. 28). Previous reports have indicated that ERa is capable of inducing the mitogen-activated protein kinase 123 (MAPK) cytoplasmic signalling cascade by a non-genomic mechanism that stimulates the expression and activation of transcription factors such as c-fos that ultimately influence cell growth (Migliaccio et al., 1996; van der Burg et al., 1991). Alternatively, receptor isoforms that are not necessarily chromatin-associated may still regulate transcription indirectly by binding to a repressor protein that if it were readily available might otherwise act to silence the induced genes (see Fig. 29). Results from in vitro binding studies show that wt ERa, ERAE3 and ERAE5 can all bind the corepressor SMRT. For the promoters we tested, however, titration of SMRT is unlikely to be the mechanism for transcriptional activation since SMRT interaction with ERa appears to be constitutive and does not adequately account for the promoter specificity of the various ER isoforms. Although SMRT does not appear to function in this role, there may be other unidentified inhibitory factors that may. While our transfection studies do not directly address the impact that ERAE3 and ERAE5 have on the transcription of endogenous genes in normal tissues or in tumors, these results suggest that they nonetheless have the potential to make distinctive and possibly unique contributions to patterns of gene regulation. As an extracellular matrix protease, collagenase (along with other metalloproteases) is likely to play an important role in the tissue remodeling and stromal invasion that occurs in metastatic disease. The IGF-l gene is similarly expressed in a variety of tissues, including the liver, bone, uterus and breast where it contributes to the normal growth and differentiation of these structures (Murphy et al., 1990; Murphy 124 et al., 1988; Pilihowska et al., 1997; Stewart et al., 1996). In the breast, some uncertainty remains regarding the cell type of origin of IGF-I and with respect to its precise role in breast tumor growth. It is likely that the mammary epithelium is exposed to IGF-l produced locally by the breast stroma as well as from systemic sources such as the liver. In addition, some breast tumors produce IGF-I and other growth factors of their own (Pilihowska et al., 1997). Expression of IGF-I appears to be deregulated in some tumors, contributing an autocrine component to the tumor cell growth. The ability of ERAE5 to stimulate transcription of the IGF-l gene is therefore of potential clinical significance. Interestingly, we observe that ERAE5, but not wt ERa promotes expression of a cotransfected IGF-l reporter plasmid, predicting that IGF-l production by breast tumors that overexpress ERAE5 will be insensitive to tamoxifen treatment. Figures 27 through 29 summarize three models depicting what we believe are the most likely mechanisms for transcriptional regulation by the ERa splicing variants ERAE3 and ERAE5. It should be emphasized that these mechanisms may coexist in estrogen receptor expressing tissues, since they are not mutually exclusive. 125 Fig. 27. Transactivation Model 1: Coactivator recruitment. A model depicting a mechanism of transactivation whereby promoter specificity and kinase dependence are governed by the selection of coactivators that each ER isoform recruit. J and F refer to jun and fos, respectively, while TF is meant to refer to an undefined transcription factor. 126 >1-.. gone. in 032220.. 133232.: 23.5.5: 3.0.1.2130... a 3.9.5.2932. 00328" 31:53.0: 02:on 127 Fig. 28. Transactivation Model 2: Kinase Activation. Model two demonstrates that promoter specificity and phorbol ester dependence may be governed by the identity of protein kinases with which each ER isoform can interact. J and F refer to jun and fos, respectively, while TF is meant to refer to an undefined transcription factor. PK signifies protein kinases such as JNK, MAPK, c-src, and casein kinase II (CKII). 128 UOIWMIOV 999"” 334i lapOW Fig. 29. Transactivation Model 3: Corepressor Neutralization. The third model suggests a mechanism for transcriptional regulation whereby promoter specificity and kinase dependence are governed by the spectrum of corepressors which associate with each ER isoform. Abbreviations are as defined in Figs. 27 and 28. I30 2.002 aw” 00308000.. 2053:5203 V. Summary and Conclusions The ERa gene gives rise to a markedly heterogeneous population of RNA transcripts that includes exon-skipped, as well as correctly spliced ERa mRN As. (Castles et al., 1995; Gotteland et al., 1995; Murphy et al., 1997). For this dissertation project, the biochemical properties of ERa variants lacking a single internal coding exon were characterized and compared to wt ERa function. These studies have shown that, while all of the variants can be expressed to give rise to stable proteins, ERAE2, ERAE4, ERAE6, and ERAE7 represent functionally impaired receptor isoforms that fail to localize to the nucleus and have no demonstrable effects (either stimulatory or inhibitory) on gene expression. Whether these variants fulfill some as of yet undefined role within the cytoplasmic compartment is currently unknown. It should be stressed, however, that none of these cytoplasmic ERa variants can bind ligand and therefore cannot be involved in mediating any of the biological effects of estrogens. The ERAE3 and ERAE5 variants, in contrast, represent nuclear isoforms that retain at least some aspects of receptor function. For ERAE3, this includes the ability to bind ligand, to form homologous or mixed dimers with itself or wt ERa, respectively, and to interact with at least some of the same co-regulatory proteins (specifically, SRC-le and SMRT) that modulate the transactivating fiinction of nuclear receptor family members. Although it is unable to bind ligand or dimerize, the ERAE5 variant may I32 bind weakly to DNA, but more importantly retains the ability to interact with the nuclear receptor coactivator SRC-le and the nuclear corepressor SMRT. These properties enable both ERAE3 and ERAE5 to potentially exert complex effects on gene expression. In stark contrast with wt ERa, ERAE3 and ERAE5 do not effectively promote gene transcription from an ERE. The observation that ERAE3 and ERAE5 inhibit wt ERor activity on a palindromic ERE provided an early indication that these variants have the capacity to modulate gene transcription. More compelling evidence of their transcriptional activity that is seen in transfection experiments involving reporter gene constructs containing nonconsensus regulatory elements. Our observations provide the first clear evidence that two ERor splicing variants, namely ERAE3 and ERAE5, like wt ERa can exert significant positive effects on gene transcription on their own and raise the intriguing possibility that each one of these receptor isoforms may target a unique, but overlapping subset of genes for transcriptional regulation. A novel pathway for ER function has been described that involves cooperation of the ER with AP-l (i.e., jun and fos family members) and perhaps with other transcription factors as well. An important distinction of this nonclassical pathway for estrogen action is that functional domains within ERa that are crucial for action through palindromic estrogen response elements (most notably the DBD) appear to be dispensable for activation of transcription by ERa through AP-l elements. We have confirmed that two ERa splicing variants (ERAE3 and ERAE5) 133 can stimulate transcription of several promoters that contain AP-l or other transcription factor binding sites, rather than classical ERa binding motifs. These include promoters for the human collagenase and IGF-1 genes. AP-l and its isoforms represent a family of nearly ubiquitous transcription factors whose activity is crucial in mediating the effects of serum and growth factors on cellular proliferation (Boyle et al., 1991; Westwick et al., 1994). Some of the target genes involved in the AP-l-directed pathway for estrogen action include significant players in the transformation process (e.g., growth factors and matrix proteases). Only rarely do human breast tumors display a simple pattern of variant receptor expression or contain more variant than wt ERa mRN A. Even so, it is conceivable that the modest increases in expression of selected ERa splicing variants relative to wt ERor that are observed in some tumors and cell lines might tend to direct ERa transcriptional activity away from ERE-containing genes towards genes involved in cell grth and transformation (driven by AP-l response elements). Several studies using quantitative PCR analysis have concluded that certain variants (notably ERAE5 and ERAE7) may be elevated relative to wt ERa in breast tumors generally, or in subsets of tumors with defined metastatic potential or steroid receptor status (Daffada et al., 1995; Leygue et al., 1996). One such study has suggested that elevated expression of ERAE5 may be indicative of the ER-lPR+ tumor phenotype and may account for some cases of tamoxifen resistance (Daffada et al., 1995). It has also been proposed that changes in the expression of ERAE3 I34 relative to wt ERoc may interfere with ERor function and modulate the growth stimulatory effects of estrogens. A comparison of ERAE3 mRN A expression in normal and tumorigenic breast specimens suggests that ERAE3 expression is reduced in breast tumors and studies show that stable over expression of ERAE3 can slow estrogen-dependent MCF-7 cell grth (Erenburg et al., 1997). Interplay between ERa splicing variants and ERB (and it’s variants) probably also exists. Defining ER function is complicated when we consider that ERa, ERB and each of their variants are all often coexpressed in cells. Estrogen signaling is a phenomenon that involves the combined contribution of many factors including coregulatory proteins, enhancer binding sites and all of the various ER isoforms. ERa, ERB and variants interact with and probably compete for binding with corepressors and coactivators in vivo, thereby limiting the availability of specific cofactors in certain cells to the detriment or benefit of gene transcription. Similarly, ERa, ERB and the ER variants, through protein-protein interactions, may compete for positioning on enhancer elements. The coexpression of ERB and ERa splicing variants may also affect in vivo dimer formation. Since ERa heterodimerizes with both ERAE3 and ERB, it is likely that receptor homology allows for ERB and ERAE3 dimer formation as well. Such a dimer may direct promoter activities differently than ERB homodimers or ERa/ERB heterodimers. For example, a particular dimer may bind a specific promoter for which another dimer has little or no affinity; and these dimer isoforms may compete to modulate the activity of a 135 single promoter. Efforts described herein to establish a transcriptional role for ERa mRNA splicing variants have identified three genes with promoters that are regulated by one or both of the isoforms, ERAE3 and ERAE5. Expression of a reporter gene driven by the chicken ovalbumin promoter is induced by ERAE5 in cells treated with PMA, and by ERAE3 and wt ERa in response to E2 and PMA cotreatment. Estrogen regulation of the ovalbumin promoter has been mapped to a critical AP-l enhancer motif (Gaub et al., 1990). Over expression of c-jun enhanced wt EROL, ERAE3 and ERAE5 activities on the ovalbumin promoter. The human collagenase promoter also harbors an AP-1 element described to direct E2 regulated gene expression (Uht et al., 1997). A short region of the collagenase promoter (~73 to +63 relative to the transcription start site) containing an AP-l motif is activated by ERAE5 and Ez-liganded ERAE3 in the presence of PMA. In contrast to the report from Uht and colleagues, in our hands wt ERa demonstrates no activity on this promoter. Further analysis indicates that ERAE3 activation of the collagenase promoter requires PMA and E2 cotreatment. These results provide additional evidence that, like wt ERa, ERAE3 and ERAE5 can activate gene expression. Interestingly, these results also indicate that although estrogen regulation for both the collagenase and the ovalbumin promoter has been mapped to an AP-l motif, these promoters are not activated with the equal effectiveness by the same receptors. This distinction suggests that flanking sequences surrounding the AP-l motif may 136 confer a difference on the mechanism of regulation, as well as the structural requirements for ERa-induced activity. The differences seen in regulation of various AP-l-containing promoters by structurally distinct ERa isoforms is reflected in the ability of other NRs to transactivate AP- 1 -directed promoters. In cotransfection studies reported by Barnberger et al. (1996), unliganded PR was also observed to act through an AP-l motif to stimulate reporter gene expression (Bamberger et al., 1996). In contrast, GR inhibits basal activity of a reporter gene construct driven by a region of collagenase promoter containing the AP-l response element (Webb et al., 1995). Another promoter included in our studies is the human IGF-l promoter. Reporter gene expression fi‘om the human IGF -1 promoter is modestly induced by PMA treatment. This promoter does not respond to wt ERa or ERAE3 and is constitutively induced by ERAE5. It is unclear where ERAE5 regulation is directed in the sequence of the IGF-l promoter, since it lacks an ERE or consensus AP-l element (Kim et al., 1991). However, 5’ deletion analysis suggests that an ERAE5 regulatory element may be localized in the region between nucleotides -532 and - 235. Efforts to transfer ERAE5 responsiveness onto a heterologous promoter were unsuccessful, showing that this effect requires support from a core promoter element. With the characterization of the ERB isoform and the initial discovery of ERa mRN A splicing variants, the results described above call into question the appealing, but simplistic notion that all of the biologically important regulatory 137 effects of ERa are mediated by a single 595 amino acid form of this receptor that was originally described by Chambon and colleagues in 1986 (Green et al., 1986). Rather, there is growing evidence that ERa splicing variants (most notably ERAE3 and ERAE5) may have a significant impact on the ability of the breast and other tissues to respond to estrogens and other regulatory signals. This regulation appears to occur through complex genomic and possibly non-genomic pathways that were not fully envisioned when Jensen and colleagues first proposed their two-step mechanism for estrogen action. I38 VI. Materials and Methods 1. Expression Vectors Plasmids for ERa mRNA splicing variant cDNAs were generated as derivatives of pCMV4 (Andersson et al., 1989) and pcDNA3.1 (Invitrogen, San Diego, CA), which support high levels of receptor expression in HeLa and C087 cell lines (Miksicek et al., 1995). Plasmids expressing ERAE4, ERAE5, and ERAE6 were generated using synthetic oligonucleotides to construct the variant splice junctions within an otherwise wt ERa cDNA expression plasmid. The remaining plasmids were constructed with the use of flanking restriction sites to shuttle cloned cDNAs (Wang and Miksicek, 1991) into the appropriate expression vectors. Mouse c-jun and c-fos cDNA, cloned into the pCMV2 expression vector, were provided by L. McCabe (Michigan State University, East Lansing, Mich.) 2. Cell Culture, Transfection, and CAT Assays C037 and HeLa cells were grown in phenol red-free DMEM supplemented with 10% calf-serum, SmM HEPES (pH 7.4), 2 mM glutamine, penicillin (50 U/ml), and strptomycin (50 ug/ml). Cells were transfected by the CaPO4 method, as previously described (Jordan et al., 1996). HeLa cells (1 x 106 cells per 60-mm dish) were transfected with 0.5 ug of the indicated ERa expression plasmid, 2 pg of 139 the c-jun expression plasmid and 10 pg of reporter plasmid. The reporter plasmids used for these experiments included pERE-TK-CAT (Klock et al., 1987), vaalb- CAT (Schweers et al., 1990), p(AP-1)3TK-CAT kindly provided by L. McCabe (Dept. of Physiology, MSU), pColl(-73)Luc received from P.J. Kushner (Webb et al., 1995), and pIGF-Luc constructs (-926, -592, -235) obtained from P. Rotwein (Kim et al., 1991). Calf thymus DNA (10 pg) was added as carrier. After overnight incubation with DNA, culture medium was replaced with 5% charcoal-treated serum-supplemented DMEM. Cell cultures were treated with the indicated hormones (20 nM PMA, 10 nM 17B-estradiol, 100 nM tamoxifen) or vehicle (ethanol + DMSO, 1% each). After 24-hour incubation, cells were harvested and CAT assays were performed as previously described (Gorman et al., 1982) using 100 pg protein. Quantitation of CAT activities was performed by phosphorimage analysis of thin layer chromatographs (ImageQuaNT, Molecular Dynamics, Inc., Sunnyvale, CA). For experiments involving biochemical or cytochemical analysis of ERa variants, Cos7 cells were similarly transfected with 10 pg of the indicated expression plasmid and 10 pg of calf thymus carrier DNA. After overnight exposure to DNA, cells were cultured for 48 hours in 10% calf serum-supplemented DMEM. All experiments involving extracts from transfected cells were normalized with respect to protein, as measured using the method of Lowry et al. (Lowry et al., 1951). Two-way AN OVA and comparison with Student’s t test were used to assess statistical differences between groups. The level of statistical significance is indicated in each figure. 140 3. E2 Binding Analysis Ligand-binding assays were performed as previously described (Neff et al., 1994). Whole-cell extracts were prepared from transfected Cos7 cells that were resuspended and sonicated in extraction buffer (20mm HEPES, pH 7.4, 20% glycerol, 0.4 M KCL, ImM MgClz) supplemented immediately before use with protease inhibitors (0.05 mg/ml each of chymostatin, trypsin inhibitor, antipain, leupeptin, aprotinin, and pepstatin). Aliquots containing 200 pg of protein were incubated overnight at 4° C with various concentrations (0.1 nM - 10 nM) of 3H- labeled E; (N EN Life Science Products, Boston, Mass.) in the presence or absence of a ZOO-fold molar excess of unlabeled E2. Free ligand was separated fiom bound ligand by treatment with dextran-coated charcoal. For determination of equilibrium binding constants, these data were plotted according to the method of Scatchard (Scatchard, 1949). 4. DNA Binding Assays DNA binding assays were performed as previously described (N eff et al., 1994). Aliquots containing 30 pg of protein from extracts prepared as above from transfected Cos7 cells were preincubated for 15 minutes at room temperature in 10 pl binding buffer [10 mM HEPES (pH 7.4), 1 mM dithiothreitol, and 20% glycerol] 141 containing 1 pg poly (dI-dC), with or without 1 pl of added human ER—specific monoclonal antibody (Mab-17), generated as described (N eff et al., 1994). Approximately 6 frnol (40,000 cpm) ofa 32P-labeled double-stranded ERE oligonucleotide (Wang and Miksicek, 1991) were added to the samples and incubated for 30 minutes at room temperature, followed by an additional S-minute incubation at 4° C. Samples were then loaded on a preelecrophoresed nondenaturing 5% polyacrylarnide gel that was run in 0.5 x Tris-Borate-EDTA at 275 V for 2 hours. The gel was dried and exposed for autoradiography. 5. Immunoblot Analysis Discontinuous 12% SDS-PAGE was carried out as previously described (Laemmli, 1970). After electrophoresis of 30 pg of whole-cell protein from extracts of transfected Cos7 cells, proteins were elecrophoretically transferred to nitrocellulose filters with 3 Trans Blot apparatus (Bio-Rad Labotatories, Inc. Richmond, CA) using the procedure previously detailed (Erickson et al., 1982). Immunblots were probed with the ER-specific monoclonal antibody, Mab—17, obtained from a hybridoma culture supemate that was diluted with an equal volume of PBS (Neff et al., 1994). Irnmunoreactive protein was visualized by enhanced chemiluminescence using a horseradish peroxidase-conjugated goat antimouse IgG, following manufacturer’s instructions (Amersham Pharmacia Biotech, Arlington Heights, IL) 142 6. In Vitro Protein-Protein Interaction Assays Variant and wt ERa receptor protein was translated in the presence of 358- methionine using TNT Coupled Reticulocyte System (Promega Corp., Madison, WI). GST-fusion proteins were expressed in the pGEX system (Pharmacia Biotech, Uppsala, Sweden) (Cavailles et al., 1994; Kalkhoven et al., 1998). The GST-SMRT fusion vector was provided by R. M. Evans . The GST-AF 2 and GST-SRC-le fusion vectors were provided by and M. G. Parker. Overnight cultures of transformed bacteria were diluted 1:20 and cultured for 2 hours before protein expression was induced with the addition of isopropyl B-D-thiogalactoside (IPTG, 0.2 mM final concentration). Bacteria were collected by centrifugation 2 hours following IPTG induction, and pellets were resuspended in 400 pl of extraction buffer supplemented with protease inhibitors. Cells were sonicated briefly, and the resulting lysates were centrifuged for 20 minutes at 20,000 rpm, 4° C. Protein concentrations were determined (Lowry et al., 1951) and extracts were diluted to 2 pg/ pl in extraction buffer and stored at -70° C until binding assays were performed. Before use in protein interaction assays, 25 pl of glutathione-Sepharose 4B beads (Pharmacia Biotech) were washed three times in 100 pl NETN [0.5% Nonidet P-40, 1 mM EDTA, 20 mM Tris (pH 8.0), 100 mM NaCl] and suspended in 100 pl NETN, 0.5% powdered milk. Washed beads were incubated with 40 pg of GST- fusion protein for 2 hours, rotating at room temperature. Beads complexed with GST-fusion proteins were washed three times with 100 pl NETN, 4° C. For protein 143 binding assays, 5 pl of in vitro translated protein was added to washed complexed beads resuspended in 100 pl NETN supplemented with protease inhibitors (as above) with and without 2.5 pM E2. After a 2-hour incubation during which the samples were rotated at room temperature, the beads were pelleted and washed four times with 100 pl NETN, 4° C. Bound proteins were separated on a discontinuous 10% polyacrylamide SDS-PAGE gel (Laemmli, 1970). The gels were dried and exposed for autoradiography. 7. Immunohistochemical and Cytochemical Analysis Indirect immunofluorescence analysis was performed as previously described (Neff et al., 1994) using Cos7 cells that were plated and transfected on glass cover slips. On the second day after transfection, cells were washed three times wih Tris-buffered saline (TBS), fixed for 3 minutes in cold 95% methanol, rehydrated by three washes with TBS, and incubated 30 minutes at 37° C with primary antibody (Mab-l 7 hybridoma supemate used at a l :10 dilution in TBS). Bound antibody was detected by staining with a rhodamine-conj ugated affinity- purified goat anti-mouse IgG (Roche Molecular Biochemicals, Indianapolis, IN) diluted 1:2000 in TBS, and incubating for 30 minutes at 37° C in the presence of 0.02 pg/ml of 4’, 6-diamidine-2-phenylindole dihydrochloride. Confocal images were recorded using the Odyssey system (Noran Instruments, Middleton, WI) on an 144 Optiphot 2 Nikon (Melville, NY) microscope. Fluorescent ligand staining of transfected Cos7 cells was performed as described (Miksicek et al., 1995) on live, whole-cell mounts treated in DMEM with 10—7 M nitrile THC. For these studies, cells were visualized using a Nikon UFX microscope equipped with a 100 watt mercury lamp for fluorescence excitation, and a 40 x 0.7 numerical aperture Plan objective. 145 VII. List of References Andersson S, Davis DN, Dahlback H, Jomvall H, Russell DW (1989) Cloning, structure, and expression of the mitochondrial cytochrome P450 sterol 26- hydroxylase, a bile acid biosynthetic enzyme. J Biol Chem, 264, 8222-8229. Angel P, Poting A, Mallick U, Rahmsdorf HJ Schorpp M, Herrlich P (1986) Induction of metallothionein and other mRNA species by carcinogens and tumor promoters in primary human skin fibroblasts. Mol Cell Biol, 6, 1760- 1766 Angel P, Rahmsdorf HJ, Pfiting A, Lficke-Huhle C, Herrlich P (1985) 12-0- tetradecanoylphorbol-l 3-acetate (TPA)-induced gene sequences in human primary diploid fibroblasts and their expression in SV40-transformed fibroblasts. J Cell Biochem, 29, 351-360 Arnold SF, Melamed M, Vororjeikina DP, Notides AC, Sasson S (1997) Estradiol- binding mechanism and binding capacity of the human estrogen receptor is regulated by tyrosine phosphorylation. Mol Endocrinol, 11, 48-53. Arnold SF, Vorojeikina DP, Notides AC (1995) Phosphorylation of tyrosine 537 on the human estrogen receptor is required for binding to an estrogen response element. J Biol Chem, 270, 30205-30212. Arnold SF, Oboum JD, Jaffe HJ, Notides AC (1995) Phosphorylation of the human estrogen receptor by mitogen-activated protein kinase and casein kinase H: consequence on DNA binding. J Steroid Biochem Mol Biol, 55, 163-172. Aronica SM, Kraus WL, Katzenellenbogen BS (1994) Estrogen action via the cAMP signaling pathway: stimulation of adenylate cyclase and CAMP- regulated gene transcription. Proc Natl Acad Sci USA, 91, 8517-8521. Bamberger AM, Bamberger CM, Gellersen B, Schulte HM (1996) Modulation of AP-l activity by the human progesterone receptor in endometrial adenocarcinoma cells. Proc Natl Acad Sci USA, 93, 6169-6174. Baniahmad A, Kohne AC, Renkawitz R (1992) A transferable silencing domain is present in the thyroid hormone receptor, in the v-erbA oncogene product and in the retinoic acid receptor. Cell, 11, 1015-1023. 146 Barettino D, Vivanco Ruiz MM, Stunnenberg HG 1994 Characterization of the ligand- dependent transactivation domain of thyroid hormone receptor. EMBO J, 13, 3039-3049. Bartsch J, Truss M, Bode J, Beato M (1996) Moderate increase in histone acetylation activates the mouse mammary tumor virus promoter and remodels its nucleosome structure. Proc Natl Acad Sci USA, 93, 10741- 10746. Beato M, Sanchez-Pacheco A (1996) Interaction of steroid hormone receptors with the transcription initiation complex. Endocr Rev, 17, 587-609. Benecke A, Chambon P, Gronemeyer H (2000) Synergy between estrogen receptor alpha activation functions AF 1 and AF 2 mediated by transcription intermediary factor TIF2. EMBO Rep, 1, 151-157. Berry M, Metzger D, Chambon P (1990) Role of the two activating domains of the oestrogen receptor in the cell-type and promoter-context dependent agonistic activity of the antioestrogen 4-hydroxytamoxifen. EMBO J, 9, 2811-2818. Blenis J (1993) Signal transduction via the MAP kinases: proceed at your own risk. Proc Natl Acad Sci USA, 90, 5889-5892. Bokemeyer D, Sorokin A, Dunn MJ (1996) Multiple intracellular MAP kinse signaling cascades. Kidney Int, 49, 1187-1198. Bowler J, Lilley TJ, Pittam JD, Wakeling AE (1989) Novel steroidal pure antiestrogens. Steroids, 54, 71-99. Boyle WJ, Smeal T, Defize LHK, Angel P, Woodgett JR, Karin M, Hunter T (1991) Activation of protein kinase C decreases phosphorylation of cJun at sites that negatively regulate its DNA-binding activity. Cell, 64, 573-584. Bresnick EH, Rories C, Hager GL (1992) Evidence that nucleosomes on the mouse mammary tumor virus promoter adopt specific translational positions. Nucleic Acids Res, 20, 865-870. Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T, Engstrom O, Ohman L, Greene GL, Gustafsson J-A, Carlquist M (1997) Molecular basis of agonism and antagonism in the oestrogen receptor. Nature, 389, 753-758. 147 Bunone G, Briand PA, Miksicek RJ, Picard D (1996) Activation of the unliganded estrogen receptor by EGF involves the MAP kinase pathway and direct phosphorylation. EMBO J, 15, 2174-2183. Burke LJ, Baniahmad A (2000) Co-repressors 2000. FASEB J, 14, 1876-1888. Castles CG, F uqua SAW, Klotz DM, Hill SM (1993) Expression of a constitutively active extrogen receptor variant in the estrogen receptor-negative BT-20 human breast cancer cell line. Cancer Res, 53, 5934-5939. Castles CG, Klotz DM, Fuqua SAW, Hill SM (1995) Coexpression of wild-type and variant oestrogen receptor mRNAs in a panel of human breast cancer cell lines. Br J Cancer, 71, 974-980. Cavailles V, Dauvois S, Danielian PS, Parker MG (1994) Interaction of proteins with transcriptionally active estrogen receptors. Proc Natl Acad Sci USA, 91, 10009-10013. Cavailles V, Dauvois S, L’Horset J, Lopez G, Hoare S, Kushner PJ, Parker MG (1995) Nuclear factor RIPl40 modulates transcriptional activation by the estrogen receptor. EMBO J, 14, 3741-3751. Chaidarun SS, Alexander J M (1998) A tumor-specific truncated estrogen receptor splice variant enhances estrogen-stimulated gene expression. Mol Endocrinol, 12, 1355-1366. Chen D, Ma H, Hong H, Koh SS, Huang S-M, Schurter BT, Aswad DW, Stallcup MR (1999) Regulation of transcription by a protein methyltransferase. Science, 284, 2174-2177. Chen H, Lin RJ, Schlitz L, Chakravarti D, Nash A, Nagy L, Privalsky ML, Nakatani Y, Evans RM (1997) Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multirneric activation complex with P/CAF a CBP/p300. Cell, 90, 569-580. Chen JD, Evans RM (1995) A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature, 377, 454-457. Chen JD, Umesono K, Evans RM (1996) SMRT isoforms mediate repression and anti-repression of nuclear receptor heterodimers. Proc Natl Acad Sci USA, 93, 7567-7571. 148 Chen WY, Townes TM (2000) Molecular mechanism for silencing virally transduced genes involves histone deacetylation and chromatin condensation. Proc Natl Acad Sci USA, 97, 377-382. Cheskis BJ, Karathanasis S, Lyttle CR (1997) Estrogen receptor ligands modulate its interaction with DNA. J Biol Chem, 272, 11384-11391. Chien CH, Wang FF, Hamilton TC (1994) Transcriptional activation of c-myc proto-oncogene by estrogen in human ovarian cancer cells. Mol cell Endocrinol, 99,1 1-19. Cho H, Katzenellenbogen BS (1993) Synergistic activation of estrogen receptor- mediated transcription by estradiol and protein kinase activators. Mol Endocrinol, 7, 441-452. Chu S and Fuller PJ (1997) Identification of a splice variant of the rat estrogen receptor 0 gene. Mol Cell Endocrinol, 132, 195-199. Coleman KM, Smith CL (2001) Intracellular signaling pathways: nongenomic actions of estrogens and ligand-independent activation of estrogen receptors. Front Bioscience, 6, 1379-1391. Collingwood TN, Butler A, Tone Y, Clifton-Bligh RJ Parker MG, Chatterjee VK (1997) Thyroid horrnone-mediated enhancement of heterodirner formation between thyroid hormone receptor beta and retinoid X receptor. J Biol Chem, 272, 13060-13065. Compston JE (2001) Sex steroids and bone. Physiol Rev, 81, 419-447. Couse JF, Curtis SW, Washburn TF, Lindzey J, Golding TS, Lubahn DB, Smithies O, Korach KS (1995) Analysis of transcription and estrogen insensitivity in the female mouse afier targeted disruption of the estrogen receptor gene. Mol Endocrinol, 9, 1441-1454. Couse JF, Hewitt SC, Korach KS (2000) Receptor null mice reveal contrasting roles for estrogen receptor a and b in reproductive tissues. J Steroid Biochem Mol Biol, 74, 287-296. Curtis Hewitt S, Couse J F, Korach KS (2000) Estrogen receptor transcription and transactivation: estrogen receptor knockout mice: what their phenotypes reveal about mechanisms of estrogen action. Breast Cancer Res, 2, 345-352. 149 Daffada AAI, Johnston SRD, Smith IE, Detre S, King N, Dowsett M (1995) Exon 5 deletion variant estrogen receptor messenger RNA expression in relation to tamoxifen resistance and progesterone receptor/p32 status in human breast cancer. Caner Res, 55, 288-293. Danielian PS, White U, Lees JA, Parker MH (1992) Identification of a conserved region required for hormone dependent transcriptional acivation by steroid hormone receptors. EMBO J, 11, 1025-1030. Darimont BD, Wagner RL, Apriletti J W, Stallcup MR, Kushner PJ, Baxter JD, Fletterick RJ, Yamarnoto KR (1998) Structure and specificity of nuclear receptor-coactivator interactions. Gene Dev, 12, 3343-3356. Dechering K, Boersma C, Mosselman S (2000) Estrogen receptors alpha and beta: two receptors of a kind? Current Med. Chem, 7, 561-576. Denner LA, Schrader WT, O’Malley BW, Weigel NL (1990) Hormonal regulation and identification of chicken progesterone receptor phosphorylation sites. J Biol Chem, 265, 16548-16555. Denton RR, Koszewski NJ, Notides AC (1992) Estrogen receptor phosphorylation. Hormonal dependence and consequence on DNA binding. J Biol Chem, 267, 7263-7268. Decensi A, Fontana V, Bruno S, Gustavino C, Gatteschi B, Costa A (1996) Effect of tamoxifen on endometrial proliferation. J Clin Oncol, 14, 434-40 Dhandaparri KM, Brann DW (2002) Protective effects of estrogen and selective estrogen receptor modulators in the brain. Biol Repro, 67, 1379-1385. Ding XF, Anderson CM, Ma H, Hong H, Uht RM, Kushner PJ, Stallcup MR (1998) Nuclear receptor-binding sites of coactivators glucocorticoid receptor interacting protein 1 (SRC-l ): multiple motifs with different binding specificities. Mol Endocrinol, 12, 302-313. DiRenzo J, Shang Y, Phelan M, Sif S, Myers M, Kingston R, Brown M (2000) BRG-l is recruited to estrogen-responsive promoters and cooperates with factors involved in histone acetylation. Mol Cell Biol, 20, 7541-7549. Dowsett M, Daffada A, Chan CM, Johnston SR (1997) Oestrogen receptor mutants and variants in breast cancer. Eur J Cancer, 33, 1177-1183. 150 Draper MW, Flowers DE, Huster WJ, Neild JA, Harper KD, Arnaud C (1996) A controlled trial of raloxifene (LY139481) HCL: impact on bone turnover and lipid profile in healthy postmenopausal women. J Bone Miner Res, 11, 835- 842. Duan R, Porter W, Safe S (1998) Estrogen-induced c-fos protooncogene expression in MCF-7 human breast cancer cells: role of estrogen receptor Spl complex formation. Endocrinol, 139, 1981-1990. Durand BM, Saunders M, Gaudon C, Roy B, Losson R, Chambon P (1994) Activation function 2 (AF-2) of retinoic acid receptor and 9-cis retinoic acid receptor: presence of a conserved autonomous constitutive activating domain and influence of the nature of the response element on AF -2 activity. EMBO J, 13, 5370-5382. Ekena K, Weis KE, Katzenellenbogen JA, Katzenellenbogen BS (1997) Different residues of the human estrogen receptor are involved in te recognition of structurally diverse estrogens and antiestrogens. J Biol Chem, 272, 5069-5075. Endoh H, Maruyama K, Masuhiro Y, Kobayashi Y, Goto M, Tai H, Yanagisawa J, Metzger K, Hashimoto S, Kato S (1999) Purification and identification of p68 RNA helicase acting as a transcriptional coactivator specific for the activation function 1 of human estrogen receptor alpha. Mol Cell Biol, 19, 5363-5372. Enmark E, Pelto-Huikko M, Grandien K, Lagercrantz S, Lagercrantz J, Fried G, Nordenskjéld M, Gustafsson J -A (1997) Human estrogen receptor B-gene structure, chromosomal localization, and expression pattern. J Clinical Endocrinol, 82, 4258-4265. Ernst M, Rodan GA (1991) Estradiol regulation of insulin-like growth factor-1 gene expression in osteoblastic cells: evidence for transcriptional control. Mol Endocrinol, 5, 1081-1089. Ernst M, Heath JK, Rodan GA (1989) Estradiol effects on proliferation, messenger ribonucleic acid for collagen and insulin-like growth factor-1, and parathyroid hormone-stimulated adenylate cyclase activity in osteoblastic cells from calvariae and long bones. Endocrinology, 125, 825-833 151 Erenburg I, Schachter B, Mira y Lopez R, Ossowski I (1997) Loss of an estrogen receptor isoform (ERaAE3) in breast cancer and the consequences of its reexpression: interference with estrogen-stimulated properties of malignant transformation. Mol Endocrinol, 11, 2004-2015. Erickson PF, Minier LN, Lasher RS (1982) Quantitative electorphoretic transfer of polypeptides from SDS polyacrylamide gels to nitrocellulose sheets: a method for their re-use in irnmunoautoradiographic detection of antigens. J Immunol Methods, 51, 241-249. Ernst M, Rodan GA (1991) Estradiol regulatioin of insulin-like growth factor-1 gene expression in osteoblastic cells: evidence for transcriptional control. Mol Endocrinol, 5, 1081-1089. Feng W, Ribiero RCJ, Wagner RL, Nguyen H, Apriletti J W, Fletterick RJ, Baxter JD, Kushner PJ, West BL (1998) Hormone-dependent coactivator binding to a hydrophobic clefi on nuclear receptors. Science, 280, 1747-1749. Fiorelli G, Sori F, Frediani U, Franceschelli F, Tanini A, Tosti-Guerra, Benvenuti S, Gennari L, Becherini L, Brandi ML (1996) Membrane binding sites and non- genomic effects of estrogen in cultured human pre-osteoclastic cells. J Steroid Biochem Mol Biol, 59, 233-240. Fletcher TM, Xiao N, Mautino G, Baumann CT, Wolford R, Warren BS, Hager GL (2002) ATP-dependent mobilization of the glucocorticoid receptor durring chromatin remodeling. Mol Cell Biol, 22, 3255-3263. F ondell JD, Guerrnan M, Malik S, Roeder RG (1999) Thyroid hormone receptor- associated proteins and general positive cofactors mediate thyroid hormone receptor function in the absence of the TATA box-binding protein-associated factors of TFHD. Proc Natl Acad Sci, 96, 1959-1964. Fragoso G, John S, Toberts MS, Hager GL (1995) Nucleosome positioning on the MMTV LTR results fiom the frequency-biased occupancy of multiple frames. Genes Dev, 9, 1933-1947. Freedman LP (1999) Increasing the complexity of coactivation in nuclear receptor signaling. Cell, 97, 5-8. Friend KE, Resnick EM, Ang LW, Shupnik MA (1997) Specific modulation of estrogen receptor mRN A isoforms in rat pituitary throughout the estrous cycle and in reponse to steroid hormones. Mol Cell Endocrinol, 13, 147-155. 152 Fr6din M, Gammeltofi S (1999) Role and regulation of 90kDa ribosomal S6 kinase (RSK) in signal transduction. Mol Cell Endocrinol, 151, 65-77. Fuqua SAW, Charnness GC, McGuire WL (1993) Estrogen receptor mutations in breast cancer. J Cell Biochem, 51, 135-139. Fuqua SAW, Fitzgerald SD, Allred DC, Elledge RM, Nawaz Z, McDonnel DP, O’Malley BW, Green GL, McGuire WL (1992) Inhibition of estrogen receptor action by a naturally occurring variant in human breast tumors. Cancer Res, 52, 483-486. Fuqua SAW, Fitzgerald SD, Charnness GC, Tandon AK, McDonnell DP, Nawaz Z, O’Malley BW, McGuire WL (1991) Variant human breast tumor estrogen receptor with constititive transcriptional activity. Cancer Res, 51, 105-109. F uqua SAW, Wolf DM (1995) Molecular aspects of estrogen receptor variants in breast cancer. Breast Cancer Res Treat, 35, 233-241. Furrnan E, Rushkin E, Margalit R, Bendel P, Degani H (1992) Tamoxifen induced changes in MCF7 human breast cancer: in vitro and in vivo studies using nuclear magnetic resonance spectroscopy and imaging. J Steroid Biochem Mol Biol, 43, 189-195. Gaub MP, Bellard M, Scheuer I, Chambon P, Sassone-Corsi P (1990) Activation of the ovalbumin gene by the estrogen receptor involves the Fos-Jun complex. Cell, 63, 1267-1276. Glass CK, Rosenfeld MG (2000) The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev, 14, 121-141. Gluzrnan Y (1981) SV40-transformed simian cells support the replication of early SV40 mutants. Cell, 23, 175-182. Gotteland M, Desauty G, Delarue J C, Liu L, May E (1995) Human estrogen receptor messenger RNA in both normal and tumor breast tissues. Mol Cell Endocrinol, 112, 1-13. Gorman CM, Moffat LF, Howard BH (1982) Recombinant genomes which express chloramphenicol acetyltransferase in mammalian cells. Mol Cell Biol, 2, 1044-105 1 . 153 Gosden JR, Middleton PG, Rout D (1986) Localisation of the human oestrogen receptor gene to chromosome 6q24-q27 by insitu hybridisation. Cytogenet Cell Genet, 43, 218-220. Graham JD, Bain DL, Richer JK, Jackson TA, Tung L, Horowitz KB (2000) Nuclear receptor conformation, coregulators, and tamoxifen-resistant breast cancer. Steroids, 65, 579-584. Green S, Walter P, Kumar V, Krust A, Bomet JM, Argos P, Chambon P (1986) Human oestrogen receptor cDNA: sequence, expression and homology to v- erbA. Nature, 320,134-139. Grumbach MM (2000) Estrogen, bone, grth and sex: a sea change in conventional wisdom. J Pediatr Endocrinol Metab, 13, Suppl 6, 1439-1455. Guo Z, Krucken J, Benten WP, Wunderlich F (2002) Estradiol-induced nongenomic calcium signaling regulates genotropic signaling in macrophages. J Biol Chem, 277, 7044-7050. Gustafsson J -A, Warner M (2000) Estrogen receptor [3 in the breast: role in estrogen responsiveness and development of breast cancer. J Steroid Biochem Mol Biol, 74, 245-248. Hall LJ, Kajimoto Y, Bichell D, Kim S-W, James PL, Counts D, Nixon LJ, Tobin G, Rotwein P (1992) Functional analysis of the rat insulin-like growth factor I gene and identification of an IGF—I gene promoter. DNA Cell Biol, 11, 301 -3 13. Hall RF, Lee CSL, Alexander IE, Shine J, ClarkCL, Sutherland R (Steroid hormone receptor gene expression in human breast cancer cells: inverse relationship between oestrogen and glucocoticoid receptor messenger RNA levels. Int J Cancer, 46, 1081-1087. Haslam SZ (1987) Role of sex steroid hormones in normal mammary gland function. Neville MC, Daniel CW (eds.) The Mammary Gland. Plenum, New York, 499-533. Hayashi Y, Ohmori S, Ito T, Seo H (1997) A splicing variant of steroid receptor coactivator-1 (SRC-le): the major isoform of SRC-l to mediate thyroid hormone action. Biochem Biophys Res Corn, 236, 83-87. Heery Dm, Kalkhoven E, Hoare S, Parder MG (1997) A signature motif in coactivators mediates binding to nuclear receptors. Nature, 387, 733-736. 154 Heinzel T, Lavinsky RM, Mullen T-M, Stiderstrtim M, Laherty CD, Torchia J, Yang W-M, Brard G, Ngo SD, Davie JR, Seto E, Eisenman RN, Rose DW, Glass CK, Rosenfeld MG (1997) A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression. Nature, 387, 43-48. Herrlich P, Angel P, Rahmsdorf HJ, Mallick U, Pfiting A, Hieber L, Lficke-Huhle C, Schorpp M (1986) The mammalian genetic stress response. Adv Enzyme Reg, 25, 485-504. Hess RA, Bunick D, Lee KH, Bahr J, Taylor JA, Korach KS, Lubahn DB (1997) A role for oestrogens in the male reproductive system. Nature, 390, 447-448. Hisamoto K, Ohmichi M, Kurachi H, Hayakawa J, Kanda Y, Nishio Y, Adachi K, Tasaka K, Miyoshi E, Fujiwara N, Taniguchi N, Murata Y (2001) Estrogen induces the Akt-dependent activation of endothelial nitric-oxide synthase in vascular endothelial cells. J Biol Chem, 276, 3459-3467. Horlein AJ, Naar AM, Heinzel , Torchia J, Gloss B, Kurokawa R, Ryan A, Kamel Y, Soderstrém M, Glass CK, Rosenfeld MG (1995) Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor. Nature, 377, 397-404. Horowitz KB, Jackson TA, Bain DL, Richer JK, Takimoto GS, Tung L (1996) Nuclear receptor coactivators and corepressors. Mol Endocrinol, 10, 1167-1177. Howard KJ, Holley SJ, Yamarnoto KR, Distelhorst CW (1990) Mapping the hsp90 binding region of the glucocorticoid receptor. J Biol Chem, 265, 11928-11935. Hu C, Hyder SM, Needleman DS, Baker VV (1996) Expression of estrogen receptor variants in normal and neoplastic human uterus. Mol Cell Endocrinol, 118, 173-179. Huston L (1999) Raloxifene reduces breast cancer risk. Lancet, 353, 43. Ignar—Trowbridge DM, Pimentel M, Parker MG, McLachlan JA, Korach KS (1996) Peptide growth factor cross-talk with the extrogen receptor requires the NB domain and occurs independently of protein kinase C or estradiol. Endocrinol, 137, 1735-1744. 155 Improta-Brears T, Whorton AR, Codazzi F, York JD, Meyer T, McDonnell DP (1999) Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proc Natl Acad Sci USA, 96, 4686-4691. Ing NH, Beekman HM, Tsai SY, Tsai MJ, O’Malley BW (1992) Members of the steroid homone recepttor superfamily interact with TFHB (S300-II). J Biol Chem, 267, 17617-17623. Iwao K, Miyoshi Y, Egawa C, Ikeda N, Noguchi S (2000) Quantitative analysis of estrogen receptor-B mRNA and its variants in human breast cancers. Int J Cancer, 88, 733-736. Jackson TA, Richer JK, Bain DL, Takimoto GS, Tung L, Horowitz KB (1997) The partial agonist activity of antagonist-occupied steroid receptors is controlled by a novel hinge domain-binding coactivator L7/SPA and the corepressors N-CoR or SMRT. Mol Endocrinol, 11, 693-705. Jacq X, Brou C, Lutz Y, Davidson 1, Chambon P, Tom L (1994) Human Tafn30 is present in a distinct TFHD complex and is required for transcriptional activation by the estrogen receptor. Cell, 79, 107-117. J anknecht R, Hunter T (1996) A growing coactivator network. Nature, 383, 22-23. Jensen EV, Suzuki T, Kwashima T, Stumpf WE, Jungblut PW, DeSombre ER (1968) A 2 step mechanism for interaction of estradiol with rat uterus. Proc Natl Acad Sci USA, 59, 632-638. J uneja M, Jose R, Kekre AN, Viswanathan F, Seshadri (2002) Tarnoxifen-induced endometrial changes in postmenopausal women with breast carcinoma. Int J Gynaecol Obstet, 76, 279-84. Joel PB, Smith J, Sturgill TW, Fisher TL, Blennis J, Lanningan DA (1998) p90rsk1 regulates estrogen receptor-mediated transcription through phosphorylation of Set-167. Mol Cell Biol, 18, 1978-1984. Jordan M, Schallhom A, Wurm FM (1996) Transfecting mammalian cells: optimization of critical parameters affecting calcium-phosphate precipitate formation. Nucleic Acids Res, 24, 596-601. Kalkhoven E, Valentine JE, Heery DM, Parker MG (1998) Isoforms of steroid receptor co-activator 1 differ in their ability to potentiate transcription by the oestrogen receptor. EMBO J, 17, 232-243. 156 Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, Lin S-C, Heyrnan RA, Rose DW, Glass CK, Rosenfeld MG (1996) A CBP integrator complex mediatees transcriptional activation and AP-l inhibition by nuclear receptors. Cell, 85, 403-414. Kato S, Endoh H, Masuhiro Y, Kitarnoto T, Uchiyama S, Sasaki H, Masushige S, Gotoh Y, Nishida E, Kawashima H, Metzger D, Chambon P (1995) Activation of the estrogen receptor through phosphorylation by mitogen- activated protein kinase. Science, 270, 1491-1494. Katzenellenbogen J A, O’Malley BW, Katzenellenbogen BS (1996) Tripartite steroid hormone receptor pharmacology: interaction with multiple effector sites as a basis for the cell- and promoter-specific action of these hormones. Mol Endocrinol, l0, 1 19-13 1 . Katzenellenbogen BS, Choi I, Delage-Mourroux R, Ediger TR, Martini PGV, Montano M, Sun J, Weis K, Katzenellenbogen JA (2000) Molecular mechanisms of estrogen action: selective ligands and receptor pharmacology. J Steroid Biochem Mol Biol, 74, 279-285. Kaufrnann M (1997) A review of endocrine Options for the treatment of advanced breast cancer. Oncology, 54 suppl, 2-5. Khavari PA, Peterson CL, Tamkun JW, Mendel DB, Crabtree GR (1993) BRGI contains a conserved domain of the SWIZ/SNFZ family necessary for normal mitotic growth and transcription. Nature, 366, 170-174. Kim SW, Lajara R, Rotwein P (1991) Structure and function of a human insulin-like growth factor-1 gene promoter. Mol Endocrinol, 5, 1964-1972. Kimmins S and MacRae TH (1999) Maturation of steroid receptors: an example of functional cooperation among molecular chaperones and their associated proteins. Cell Stress Chaperones, 5, 76-86. King MC, Wieand S, Hale K, Lee M, Walsh T, Owens K, Tait J, Ford L, Dunn BK, Costantino J, Wickerharn L, Wolmark N, Fisher B (2001) Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: national surgical adjuvant breast and bowel project (N SABP- Pl) Breast Cancer Prevention Trial. JAMA, 286, 2251-2256. King WJ, Greene GL (1984) Monoclonal antibodies localize oestrogen receptor in the nuclei of target cells. Nature, 307, 745-747. 157 Kingston RE, Narlikar GJ (1999) ATP-dependent remodeling and acetylation as regulators of chromatin fluidity. Genes Dev, 13, 2339-2352. Klein-Hitpass L, Ryffel GU, Heitlinger E, Cato AC (1988) A 13 bp palindrome is a functional estrogen responsive element and interacts specifically with estrogen receptor. Nucleic Acids Res, 16, 647-663. Klock G, Strahle U, Schfitz G (1987) Oestrogen and glucocorticoid responsive elements are closely related but distinct. Nature, 329, 734-736. Klotz DM, Castles CG, Fuqua SAW, Spriggs LL, Hill SM (1995) Differential expression of wild-type and variant ERmRNAs by stocks of MCF-7 breast cancer cells may account for differences in estrogen responsiveness. Biochem Biophys Res Corn, 210, 609-615. Kobayashi Y, Kitamoto T, Masuhiro Y, Watanabe M, Kase T, Metzger D, Yanagisawa J, Kato S (2000) p3 00 mediates functional synergism between AF-l and AF —2 of estrogen receptor alpha and beta by interacting directly with the N-terrninal A/B domains. J Biol Chem, 275, 15645-15651. Koehorst SGA, Cox JJ, Donker GH, Lopes da Silva S, Burbach JPH, Thijssen JHH, Blankenstein MA (1994) Functional analysis of an alternatively spliced estrogen receptor lacking exon 4 isolated from MCF-7 breast cancer cells and meningioma tissue. Mol Cell Endocrinol, 101, 237-245. Koh SS, Chen D, Lee YH, Stallcup MR (2001) Synergistic enhancement of nuclear receptor function by p160 coactivators and two coactivators with protein methylatransferase activities. J Biol Chem, 276, 1089-1098. Korach KS (1994) Insights from the study of animals lacking functional estrogen receptor. Science, 266, 1524-1527. Kraus WL, McInerney EM, Katzenellenbogen BS (1995) Ligand-dependent, transcriptionally productive association of the amino- and caboxyl-terminal regions of a steroid hormone nuclear receptor. Proc Natl Acad Sci USA, 92, 12314-12318. Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson J -A, Smithies O (1998) Generation and reproductive phenotypes of mice lacking estrogen receptor [3. Proc Natl Acad Sci USA, 95, 15677-15682. 158 Krishnan V, Wang X, Safe S (1994) Estrogen receptor-Spl complexes mediate estrogen-induced cathepsin D gene expression in MCF-7 human breast cancer cells. J Biol Chem, 269, 15912-15917. Kumar V, Chambon P (1988) The estrogen receptor binds tightly to its responsive element as a ligand-induced homodirner. Cell, 55, 145-156. Kumar V, Green S, Stack G, Berry M, J in JR, Chambon P (1987) Functional domains of the human estrogen receptor. Cell, 51, 941-951. Kumar V, Guren S, Staub A, Chambon P (1986) Localization of the oestradiol- binding and putative DNA binding domain of the human oestrogen receptor. EMBO J, 5, 2231-2236. Kuiper GGJ M, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson J—A (1996) Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA, 93, 5926-5930. Kuroski de Bold ML (1999) Estrogen natriuretic peptides and the renin-angiotensin system. Cardiovasc Res, 41, 524-531. Laemmli UK (1970) Cleavage of structural proteins during assembly of the head of bacteriophage T4. Nature, 227, 680-685. Lamb J, Ladha MH, McMahon C, Sutherland RL, Ewen ME (2000) Regulation of the functional interaction between cyclin D1 and the estrogen receptor. Mol Cell Biol, 20, 8667-8675. Lamy PJ, Pujol P, Thezenas S, Kramar A, Rouanet P, Guilleux F, Grenier J (2002) Progesterone receptor quantification as a strong prognostic determinant in postmenopausal breast cancer women under tamoxifen therapy. Breast Cancer Res Treat, 76, 65-71 Laurent BC, Treich I, Carlson M (1993) The yeast SNF2/SW12 protein has DNA- stimulated ATPase activity required for transcriptional activation. Genes Dev, 7, 583-591. Lavinsky RM, Jepsen K, Heinzel T, Torchia J, Mullen TM, Schiff R, Del-Rio AL, Ricote M, Ngo S, Gemsch J, Hilsenbeck SG, Osborne DK, Glass Ck, Rosenfeld MG (1998) Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR complexes. Proc Natl Acad Sci USA, 95, 2920-2925. 159 Lawson J S, Field AS, Champion S, Tran D, Ishikura H, Trichopoulos D (1999) Low oestrogen receptor alpha expression in normal breast tissue underlies low breast cancer incidence in Japan. Lancet, 354, 1787-1788. Lazennec G, Bresson D, Lucas A, Chauveau C, Vignon F (2001) BR beta inhibits proliferation and invasion of breast cancer cells. Endocrinology, 142, 4120-4130. Leygue ER, Watson PH, Murphy LC (1996) Estrogen receptor variants in normal human mammary tissue. J Natl Cancer Inst, 88, 284-290. Li H, Leo C, Schroen DJ, Chen JD (1997) Characterization of receptor interaction and transcriptional repression by the corepressor SMRT. Mol Endocrinol, 11, 2025-2037. Liotta LA 1986 Tumor invasion and metastases — role of the extracellular matrix: rhoads memorial award lecture. Cancer Res, 46,1-7. L0pez GN, Webb P, Shinsako JH, Baxter JD, Greene GL, Kushner PJ (1999) Titration by estrogen receptor activation function-2 of targets that are downstream from coactivators. Mol Endocrinol, 13, 897-909. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein measurement with the folin phenol reagent. J Biol Chem, 193, 265-275. Lubahn DB, Moyer J S, Golding TS, Couse JF, Korach KS, Smithies O (1993) Alteration of reproductive function but nor prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci USA, 90, 1 1162-1 1166. Ma H, Hong H, Huang SM, Irvine RA, Webb P, Kushner PJ, Coetzee GA, Stallcup MR (1999) Multiple signal input and output domains of the 160-kilodalton nuclear receptor coactivator proteins. Mol Cell Biol, 19, 6164-6173. Mak HY, Hoare S, Henttu PMA, Parker MG (1999) Molecular determinants of the estrogen receptor-coactivator interface. Mol Cell Biol, 19, 3895-3903. Matsuda K, Ochiai I, Nishi M, Kawata M (2002) Colocalization and ligand- dependent discrete distribution of the estrogen receptor (ER) alpha and ER beta. Mol Endocrinol, 10, 2215-2230. 160 McCarthy TL, Centrella M, Canalis E (1989) Parathyroid hormone enhances the transcript and polypeptide levels of insulin-like growth factor I in osteoblast- enriched cultures from fetal rat bone. Endocrinology, 124, 1247-1253 McCarthy TL, J i C, Shu H, Casinghino S, Crothers K, Rotwein P, Centrella M (1997) l7B-estradiol potently suppresses CAMP-induced insulin-like growth factor-I gene activation in primary rat osteoblast cultures. J Biol Chem, 272, 18132-18139 McDonnell DP, Clemm DL, Hermann T, Goldman ME, Pike J W (1995) Analysis of estrogen receptor function in vitro reveals three distinct classes of antiestrogens. Mol Endocrinol, 9, 659-669. Mclnemey EM, Tsai MJ, O’Malley BW, Katzenellenbogen BS (1996) Analysis of estrogen receptor transcriptional enhancement by a nuclear hormone receptor coactivator. Proc Natl Acad Sci USA, 93, 10069-10073. McKenna NJ, Lanz RB, O’Malley BW (1999) Nuclear receptor co-regulators: cellular and molecular biology. Endocrinol Rev, 20, 321-344. McNally JG, Muller WG, Walker D, Wolford R, Hager GL (2000) The glucocorticoid receptor: rapid exchange with regulatory sites in living cells. Science, 287, 1262-1268. Menasce LP, White GRM, Harrison CJ, Boyle JM (1993) Localization of the estrogen receptor locus (ESR) to chromosome 6q25.1 by FISH and a simple post-fish banding technique. Genomics, 17, 263-265. Metivier R, Penot G, F louriot G, Pakdel F (2001) Synergism between ER-alpha transactivation function 1 (AF -1) and AF -2 mediated by steroid receptor coactivator protein-1: requirement for the AF -1 alpha-helical core and for a direct interaction between the N- and C-terminal domains. Mol Endocrinol, 15, 1953-1970. Metivier R, Petit F G, Valotaire Y, Padkel F (2000) Function of N-terminal transactivation domain of the estrogen receptor requires a potential alpha- helical structure and is negatively regulated by the A domain. Mol Endocrinol, 14, 1849-1871. Metzger K, Ali S, Bonnert JM, Chambon P (1995) Characterization of the amino- terrninal transcriptional activation function of the human estrogen receptor in animal and yeast cells. J Biol Chem, 270, 9535-9542. 161 Meyer ME, Gronmeyer H, Turcotte B, Bocquel MT, Tasset D, Chambon P (1989) Steroid hormone receptors compete for factors that mediate their enhancer function. Cell, 57, 433-442. Migliaccio A, di Domenico M, Castoria G, de Falco A, Bontempo P, Nola E, Auricchio F (1996) Tyrosine kinase/p21ras/MAP-kinase pathway activation by estradiol-receptor complex in MCF-7 cells. EMBO J, 15, 1292-1300. Miksicek RJ, Carlson KE, Hwang K-J, Katzenellenbogen JA (1995) Studies using fluorescent tetrahydrochrysene estrogens for in situ visualization of the estrogen receptor in living cells. Mol Endocrinol, 9, 592-604. Miksicek RJ, Lei Y, Wang Y (1993) Exon skipping gives rise to alternatively spliced forms of the estrogen receptor in breast tumor cells. Breast Cancer Res Treat, 26, 163-174. Montano MM, Jaiswal AK, Katzenellenbogen BS (1998) Transcriptional regulation of the human quinone reductase gene by antiestrogen-liganded estrogen receptor-alpha and estrogen receptor-beta. J Biol Chem, 273, 25443-25449. Mosselman S, Polman J, Dijkema R (1996) ERB: identification and characterization of a novel human estrogen receptor. F EBS Lett, 392, 49-53. Muchardt C, Yaniv M (1993) A human homologue of Saccharomyces cerevisiae SNF2/ SW12 and Drosophila brrn genes potentiates transcriptional activation by the glucocorticoid receptor. EMBO J, 12, 4279-4290. Murphy LJ, Friesen HG (1988) Differential effects of estrogen and growth hormone on uterine and hepatic insulin-like growth factor I gene expression in the ovariectomized hypophysectomized rat. Endocrinology, 122, 325-332 Murphy LJ, Ghahary A (1990) Uterine insulin-like growth factor-1: regulation of expression and its role in estrogen-induced uterine proliferation. Endocr Rev, 3, 443-453. Murphy LC, Leygue E, Dotzlaw H, Douglas D, Coutts A, Watson PH (1997) Oestrogen receptor variants and mutations in human breast cancer. Ann Med, 29, 221-234. Naar AM, Beaurang PA, Zhou S, Abraham S, Solomon W, Tijan R (1999) Composite co-activator ARC mediates chromatin directed transcriptional activation. Nature, 398, 828-832 162 Nagaoka I, Trapnell BC, Crystal RG (1990) Regulation of insulin-like growth factor I gene expression in the human macrophage-like .cell line U937. J Clinical Invest, 85, 448-455 Nagy L, Kao H-Y, Chakravarti D, Lin RJ, Hassig CA, Ayer DE, Schreiber SL, Evans RM (1997) Nuclear receptor repression mediated by a complex containing SMRT, mSin3A, and histone deacetylase. Cell, 89, 373-380. Nagy L, Kao H-Y, Love JD, Li C, Banayo E, Gooch JT, Krishna V, Chatterjee K, Evans RM, Schwabe J WR (1999) Mechanism of corepressor binding and release from nuclear hormone receptors. Genes Dev, 13, 3209-3216. Neff S, Sadowski C, Miksicek RJ (1994) Mutational analysis of cysteine residues within the hormone-binding domain of the human estrogen receptor identifies mutants that are defective in both DNA-binding and subcellular distribution. Mol Endocrinol, 8, 1215-1223. Newton CJ, Buric R, Trapp T, Brockmeier S, Pagotto U, Stalla GK (1994) The unliganded estrogen receptor (ER) transduces growth factor signals. J Steroid Biochem Mol Biol, 48, 481-486. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kuokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV (1998) Ligand binding and coactivator assembly of the peroxisome proliferator-activated receptor- garnma. Nature, 395, 137-143. Nuttall ME, Stroup GB, Fisher PW, Nadeau DP, Gowen M, Suva LJ (2000) Distinct mechanisms of action of selective estrogen receptor modulators in breast and osteoblastic cells. Am J Physiol Cell Physiol, 279, 1550-1557. Ogawa S, Inoue S, Watanabe T, Hiroi H, Orimo A, Hosoi T, Ouchi Y, Murarnatsu M (1998) The complete primary structure of human estrogen receptor B (hERB) and its heterodirnerization with ERa in vivo and in vitro. Biochem Biophys Res Corn, 243, 122-126. Ogryzko VV, Schiltz RL, Russanova V, Howard BH, Nakatani Y (1996) The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell, 87, 953-959. Ohlsson H, Lykkesfeldt AE, Madsen MW, Briand P (1998) The estrogen receptor variant lacking exon 5 has dominant negative activity in the human breast epithelial cell line HMT-352281. Cancer Res, 58, 4264-4268. 163 Ofiate SA, Boonyaratanakomkit V, Spencer TE, Tsai SY, Tsai M-Y, Edwards DP, O’Malley BW (1998) The steroid receptor coactivator-l contains multiple receptor interacting and activation domains that cooperatively enhance the activation function 1 (AF 1) and AF 2 domains of steroid receptors. J Biol Chem, 273,121010-12108. Ofiate SA, Tsai SY, Tsai M-J, O’Malley BW (1995) Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science, 270, 1354-1357. Owen -Hughes T, Utley RT, Cote J, Peterson CL, Workman JL (1996) Persistent site-specific remodeling of a nucleosome array by transient action of the SWI/SNF complex. Science, 273, 513-516. Paech K, Webb P, Kuiper GGJM, Nilsson S, Gustafsson J-A, Kushner PJ, Scarrlan TS (1997) Differential ligand activation of estrogen receptors ERa and ERB at APl sites. Science, 277, 1508-1510. Pasqualini C, Guivarc’h D, Barnier J-V, Guibert B, Vincent J -D, Vernier P (2001) Differential subcellular distribution and transcriptional activity of 2E3, 2E4, and 2E3-4 isoforms of the rat estrogen receptor-0t. Mol Endocrinol, 15, 894-908. Pearson G, Robinson F, Gibson TB, Xu B-E, Karandikar M, Berman K, Cobb MH (2001) Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocrine Rev, 22, 153-183. Perissi V, Staszewski LM, Mclnemey EM, Kurokawa R, Krones A, Rose DW, Lambert MH, Milburn MV, Glass CK, Rosenfeld MG (1999) Molecular determinants of nuclear receptor-corepressor interaction. Genes Dev, 13, 3198-3208. Pilihowska M, Kimua N, Fujiwara H, Nagura H (1997) Immunohistochemical study of TGF-alpha, TGF-beta, EGF R, and IGF -1 expression in human breast carcinoma. Mod Pathol, 10, 9699-9675 Ponglikitrnongkol M, Green S, Chambon P (1988) Genomic organization of the human estrogen receptor gene. EMBO J, 7, 3358-3388. Poola I, Abraham J, Baldwin K (2002) Identification of ten exon deleted ER beta mRNAs in human ovary, breast, uterus and bone tissues: alternate splicing pattern of estrogen receptor beta mRN A is distinct from that of estrogen receptor alpha. FEBS Lett, 516, 133-138. 164 Porter W, Saville B, Hoivik D, Safe S (1997) Functional synergy between the transcription factor Spl and the estrogen receptor. Mol Endocrinol, 11, 1569-1580. Price RH, Butler CA, Webb P, Uht R, Kushner P, Handa RJ (2001) A splice variant of estrogen receptor [3 missing exon 3 displays altered subnuclear localization and capacity for transcriptional activation. Endocrinol, 142, 203 9-2049. Rachez C, Lemon BD, Suldan Z, Bromleigh V, Gamble M, Naar AM, Erdjurnent- Bromage H, Tempst P, Freedman LP (1999) Ligand-dependent transcription activation by nuclear receptors requires the DRIP complex. Nature, 398, 824-828. Rameh LE and Cantley LC (1999) The role of phosphinositide 3-kinase lipid products in cell function. J Biol Chem, 274, 8347-8350. Razandi M, Pedram A, Greene GL, Levin ER (1999) Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERa and ERB expressed in chinese hamster ovary cells. Mol Endocrinol, 13, 307-319. Rea D, Parker MG (1996) Effects of an exon 5 variant of the estrogen receptor in MCF-7 breast cancer cells. Cancer Res, 56, 1556-1563. Robyr D, Wolffe AP, Walter W (2000) Nuclear hormone receptor co-regulators in action: diversity for shared tasks. Mol Endocrinol, 14, 329—347. Sabbah M, Courilleau D, Mester J, Redeuilh G (1999) Estrogen induction of the cyclin D1 promoter: involvement of a CAMP response-like element. Proc Natl Acad Sci USA, 96, 11217-11222 Sadovsky Y, Webb P, Lopez G, Baxter JD, Fitzpatrick PM, Giang-Ginsberg E, Cavailles V, Parker G, Kushner PJ (1995) Transcriptional activators differ in their responses to over expression of TATA-box-binding protein. Mol Cell Biol, 15, 1554-1563. Sanders M, McKnight GS (1988) Positive and negative regulatory elements control the steroid-responsive ovalbumin promoter. Biochem, 27, 6550-6557 Scatchard G (1949) The attractions of proteins for small molecules and ions. Ann NY Acad Sci, 51, 660-672. 165 Schwabe J WR, Rhodes D (1991) Beyond zinc fingers: steroid hormone receptors have a novel structural motif for DNA recognition. Trends Biochem Sci, 16, 291-296. Schweers LA, Frank DE, Weigel NL, Sanders MM (1990) The steroid-dependent regulatory element in the ovalbumin gene does not function as a typical steroid-response element. J Biol Chem, 265, 7590-7595. Sewack GF, Ellis TW, Hansen U (2001) Binding of TATA binding protein to a naturally positioned nucleosome is facilitated by histone acetylation. Mol Cell Biol, 21,1404-1415. Sewack GF and Hansen U (1997) Nucleosome positioning and transcrition- associated chromatin alterations on the huan estrogen-responsive pS2 promoter. J Biol Chem, 272, 31118-31129. Shang Y and Brown M (2002) Molecular determinants for the tissue specificity of SERMS. Science, 295, 2465-2468. Sheppard HM, Harries JC, Hussain S, Bevan C, Heery DM (2001) Analysis of the steroid receptor coactivator 1(SRC1)-CREB binding protein interaction interface and its importance for the function of SRCl. Mol Cell Biol, 21, 39-50. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA, Greene GL (1998) The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell, 95, 927-937. Speirs V, Malone C, Walton DS, Kerin MJ, Atkin SL (1999) Increased expression of estrogen receptor beta mRN A in tamoxifen-resistant breast cancer patients. Cancer Res, 59, 5421-5424. Sylvia VL, Boyan BD, Dean DD, Schwartz Z (2000) The membrane effects of 170- estradiol on chondrocyte phenotypic expression are mediated by activation of protein kinase C through phospholipase C and G-proteins. J Steroid Biochem Mol Biol, 73, 211-224. Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK (2000) Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature, 407, 53 8-541. 166 Sluyser M 1995 Mutations in the estrogen receptor gene. Human Mut, 6, 97-103. Smigel K (1998) Breast cancer prevention trial shows major benefit, some risk. J Natl Cancer Inst, 90, 647-648. Smith CL, Nawaz Z, O’Malley BW (1997) Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4- hydroxytarnoxifen. Mol Endocrinol, 11, 657-666. Smith CL, Ofiate SA, Tsai M-J, O’Malley BW (1996) CREB binding protein acts synergistically with steroid receptor coactivator-l to enhance steroid receptor-dependent transcription. Proc Natl Acad. Sci USA, 93, 8884-8888. Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J, Mizzen CA, McKenna NJ, Oflate SA, Tsai SY, Tsai M-J, O’Malley BW (1997) Steroid receptor coactivator-1 is a histone acetyltransferase. Nature, 389, 194-198. Srabile LP, Davis AL, Gubish CT, Hopkins TM, Lukerich JD, Christie N, Finkelstein S, Siegfried JM (2002) Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen. Cancer Res, 62, 2141-2150. Stewart CEH, Rotwein P (1996) Growth, differentiation, and survival: multiple physiological function for insuling-like grth factors. Physiological Rev, 76, 1005-10026 Sun G, Porter W, Safe S (1998) Estrogen-induced retinoic acid receptor alpha 1 gene expression role of estrogen receptor-Spl complex. Mol Endocrinol, 12, 882-890. Takeshita A, Yen PM, Misiti S, Cardona GR, Liu Y, Chin WW (1996) Molecular cloning and properties of a full-length putative thyroid horone receptor coactivator. Endocrinol, 137, 3594-3597. Tanenbaum DM, Wang Y, Williams SP, Sigler PB (1998) Crystallographic comparison of the estrogen and progesterone receptor’s ligand binding domains. Proc Natl Acad Sci USA, 95, 5998-6003. Tof’t D, Gorski J (1966) A receptor molecule for estrogens: isolation from the rat uterus and preliminary characterization. Proc Natl Acad Sci USA, 53, 1574-1581. 167 Tora L, Gaub MP, Mader S, Dierich A, Bellard M, Chambon P (1988) Cell-specific activity of a GGTCA half-palindromic oestrogen-responsive element in the chicken ovalbumin gene promoter. EMBO J, 7, 3771-3778. Tora L, White J, Brou C, Tasset D, Webster N, Scheer E, Chambon P (1989) The human estrogen receptor has two independent nonacidic transcriptional activation functions. Cell, 59, 477-487. Toran-Allerand CD, Singh M, Setalo Jr. G (1999) Novel mechanisms of estrogen action in the brain: new players in an old story. Front Neuroendocrin, 20, 97-121. Traish AM, Al-Fadhli S, Klinge C, Kounine M, Quick TC (1995) Identification of structurally altered estrogen receptors in human breast cancer by site-directed monoclonal antibodies. Steroids, 60, 467-474. Tremblay A, Tramblay GB, Labrie F, Giguere V (1999) Ligand-independent recruitment of SRC-l to estrogen receptor beta through phosphorylation of activation function AF -1. Mol Cell, 3, 513-519. Tremblay GB, Tremblay A, Copeland NG, Gilbert DJ, Jenkins NA, Labrie F, Giguere V (1997) Cloning, chromosomal localization, and firnctional analysis of the murine estrogen receptor beta. Mol Endocrinol, 11, 353-365. Truss M, Beato M (1993) Steroid hormone receptors: interaction with deoxyribonucleic acid and transcription factors. Endocrine Rev, 14, 459-479 Tsai M-J, O’Malley BW (1994) Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem, 63, 451-486. Uht RM, Anderson CM, Webb P, Kushner PJ (1997) Transcriptional activities of estrogen and gluccocorticoid receptors are functionally integrated at the AP-l element. Endocrinology, 138, 2900-2908. Umayahara , Kawamori R, Watada H, Irnano E, Iwarna N, Morishima T, Yarnasaki Y, Kajimoto Y, Karnada T (1994) Estrogen regulation of the insulin-like growth factor 1 gene transcription involves an AP-l enhancer. J Biol Chem, 269, 16433-16442. Umayahara Y, Billiard J, Ji C, Centrella M, McCarthy TL, Rotwein P (1999) CCAAT/enhancer-binding protein 8 is a critical regulator of insulin-like grth factor-I gene transcription in osteoblasts. J Biol Chem, 274, 10609-10617. 168 van der Burg B, de Groot RP, Isbrficker L, Kruijer W, de Laat SW (1991) Oestrogen directly stimulates growth factor signal transduction pathways in human breast cancer cells. J Steroid Biochem Mol Biol, 40, 215-221. van Dijk MA, Floore AN, Kloppenborg KI, van’t Veer LJ (1997) A functional assay in yeast for the human estrogen receptor displays wild-type and variant estrogen receptor messenger RNAs present in breast carcinoma. Cancer Res, 57, 3478-3485. Vuong H, Patterson T, Shapiro P, Kalvaklanu DV, Wu R, Ma WY, Dong Z, Kleeberger SR, Reddy SP (2000) Phorbol ester-induced expression of airway squamous cell differentiation marker, SPRRl B, is regulated by protein kinase Cd/Ras/MEKK17MKK1-dependent/AP-l signal transduction pathway. J Biol Chem, 275, 32250-32259. Voegel JJ, Heine MJ S, Zechel C, Chambon P, Gronemeyer H (1996) TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF -2 nuclear receptors. EMBO J, 15, 3667-3675. Wade CB, Robinson S, Shapiro RA, Dorsa DM (2001) Estrogen Receptor ERor and ERB exhibit unique pharmacologic properties when coupled to activation of the mitogen-activated protein kinase pathway. Endocrinol, 142, 2336-2342. Wakeling AE, Dukes M, Bowler J (1991) A potent specific pure antiestrogen with cinical potential. Cancer Res, 51, 3867-3873. Wang Y, Miksicek RJ (1991) Identification of a dominant negative form of the human estrogen receptor. Mol Endocrinol, 5, 1707-1715. Watanabe M, Yanagisawa J, Kitagawa H, Takeyarna K, Ogawa S, Arao Y, Suzawa M, Kobayashi Y, Yano T, Yoshikawa H Masuhiro Y, Kato S (2001) A subfamily of RNA-binding DEAD-box proteins acts as an estrogen receptor alpha coactivator through the N-terrninal activation domain (AF -1) with an RNA coactivator, SRA. EMBO J, 20, 1341-1352. Welshons WV, Lieberman ME, Gorski J (1984) Nuclear localization of unoccupied oestrogen receptors. Nature, 307, 747-749. Webb P, Anderson CM, Valentine C, Nguyen P, Marimuthu A, West BL, Baxter JD, Kushner PJ (2000) The nuclear receptor corepressor (N -CoR) contains three isoleucine motifs (I/LXXII) that serve as receptor interaction domains (IDs). Mol Endocrinol, 14, 1976-1985. 169 Webb P, Lopez GN, Uht RM, Kushner PJ (1995) Tamoxifen activation of the estrogen receptor/AP-l pathway: potential origin for the cell-specific estrogen-like effects of antiestrogens. Mol Endocrinol, 9, 443-456. Webb P, Nguyen P, Shinsako J, Anderson C, F eng W, Nguyen MP, Chen D, Huang S-M, Subramanian S, McKinemey E, Katzenellenbogen BS, Stallcup MR, Kushner PJ (1998) Estrogen receptor activation fimction 1 works by binding p160 coactivator proteins. Mol Endocrinol, 12, 1605-1618. Westin S, Rosenfeld MG, Glass CK (2000) Nuclear receptor coactivators. Adv Pharrnacol, 47, 89-112. Westwick JK, Weitzel C, Minden A, Karin M, Brenner DA (1994) Tmnor necrosis factor a stimulates AP-l activity through prolonged activation of the c-Jun kinase. J Biol Chem, 269, 2639-26401. Whitham SE, Murphy G, Angel P, Rahmsdorf H-J, Smith BJ, Lyons A, Harris TJR, Reynolds JJ, Hierrlich P, Docherty AJP (1986) Comparison of human stromelysin and collagenase by cloning and sequence analysis. Biochem J, 240, 913-916. Wijayaratne AL, Nagel SC, Paige LA, Christensen DJ, Norris JD, Fowlkes DM, McDonnell DP (1999) Comparative analyses of mechanistic differences among antiestrogens. Endocrinol, 140, 5828-5840. Wiseman LR, Johnson MD, Wakeling AE, Lykkesfeldt AE, May FEB, Westley BR (1993) Type I lGF receptor and acquired tamoxifen resistance in oestrogen- responsive human breast cancer cells. Eur J Cancer, 29A, 2256-2264. Wolf KM, Jordan VC (1994) Characterization of tamoxifen stimulated MCF-7 tumor variants grown in athymic mice. Breast Cancer Res Treat, 31, 117-127. Wolf M, Ingbar SH, Moses AC (1989) Thyroid hormone and growth hormone interact to regulate insulin-like growth factor-I messenger ribonucleic acid and circulating levels in the rat. Endocrinology, 125, 2905-2914 Wolffe AP (1994) Transcription: in tune with the histones. Cell, 77, 13-16. Wong C-W, Privalsky ML (1998) Transcriptional repression by the SMRT-mSin3 corepressor: multiple interactions, multiple mechanisms, and a potential role for TFIIB. Mol Cell Biol, 18, 5500-5510. 170