Estrogen response element sequence impacts the conformation and transcriptional activity of estrogen receptor α1

https://doi.org/10.1016/S0303-7207(01)00382-3Get rights and content

Abstract

Estrogens play a critical role in mammary gland development, bone homeostasis, reproduction, and the pathogenesis of breast cancer by activating estrogen receptors (ERs) α and β. Ligand-activated ER stimulates the expression of target proteins by interacting with specific DNA sequences: estrogen response elements (EREs). We have demonstrated that the ERE sequence and the nucleotide sequences flanking the ERE impact ERα binding affinity and transcriptional activation. Here, we examined whether the sequence of the ERE modulates ERα conformation by measuring changes in sensitivity to protease digestion. ERα, occupied by estradiol (E2) or 4-hydroxytamoxifen (4-OHT), was incubated with select EREs and digested by chymotrypsin followed by a Western analysis with antibodies to ERα. ERE binding increased the sensitivity of ERα to chymotrypsin digestion. We found both ligand-specific and ERE-specific differences in ERα sensitivity to chymotrypsin digestion. The ERE-mediated increase in ERα sensitivity to chymotrypsin digestion correlates with E2-stimulated transcriptional activity from the same EREs in transiently transfected cells. Transcriptional activity also correlates with the affinity of ERα-ERE binding in vitro. Our results support the hypothesis that the ERE sequence acts as an allosteric effector, altering ER conformation. We speculate that ERE-induced alterations in ERα conformation modulate interaction with co-regulatory proteins.

Introduction

Estrogen receptor (ER) is a member of the steroid/thyroid superfamily of proteins that act as hormone-inducible transcription factors (Mangelsdorf et al., 1995). The products of two ER genes, i.e. ERα and ERβ, mediate the actions of estrogens in target tissues, thus regulating estrogenic effects on reproduction, bone homeostasis, and mammary gland structure and function (Katzenellenbogen and Korach, 1997). Estradiol (E2) and other ligands bind in the ligand-binding domain (LBD) of ERα and induce conformational changes leading to dimerization and high-affinity ERα binding to estrogen response elements (EREs) (Klinge, 2000). Once bound to an ERE, interaction between specific motifs within E2-ERα and coactivator proteins, e.g. SRC-1 (Onate et al., 1995), or the TATA binding complex, e.g. TFIIB (Ing et al., 1992) and TATA-box binding protein (TBP) (Sadovsky et al., 1995), result in increased gene transcription (reviewed in McKenna et al., 1999, Klinge, 2000).

Occupation of ERα by selective estrogen receptor modulators (SERMs), e.g. tamoxifen (TAM), 4-hydroxy-TAM (4-OHT), or raloxifene (RAL), changes the interaction of ERα with coactivators. Whereas E2-occupied ERα interacts with specific coactivators, e.g. SRC-1, occupation of ERα by TAM or 4-OHT prevents interaction with these proteins (reviewed in McKenna et al., 1999, Klinge, 2000). Although antagonist-liganded ERα interacts with corepressors NCoR and SMRT (Smith et al., 1997, Zhang et al., 1998), there is no evidence that these corepressors are required for the antagonist activity of TAM. TAM-occupied ERα also interacts with the ER-selective coregulator REA that potentiates the antagonist activity of TAM and suppresses E2-induced transcriptional activity of ERα and ERβ (Montano et al., 1999). Similarly, the nuclear matrix protein HET/SEF-B shows a preferential interaction with TAM-occupied ERα and inhibits both E2- and TAM- stimulated transcription in HepG2 cells in which TAM is an agonist (Oesterreich et al., 2000). Although crystal structure studies show a different conformation of the LBD occupied by 4-OHT versus E2 or the synthetic estrogen diethylstilbestrol (DES) (Brzozowski et al., 1997, Shiau et al., 1998), specific details of the mechanisms of antagonist activity of 4-OHT remain to be elucidated.

In addition to ligand effects, Lefstin and Yamamoto proposed that response elements recognized by nuclear transcription factors, including members of the steroid/nuclear receptor superfamily, contain information that is interpreted by bound regulator factors (Lefstin and Yamamoto, 1998). Based on Lefstin and Yamamoto's model, we postulate that DNA acts as an allosteric ligand whose binding alters ERs' affinity for other ligands, such as coactivators or corepressors, and predict that the DBD of the ERα harbors activities in addition to DNA recognition. Indeed, the ERα DBD constitutes surfaces for both intramolecular and intermolecular protein–protein contacts (Chen et al., 1999). Thus, ERα-ERE binding should produce changes in ERα conformation that alter ERα interaction with other proteins.

To assess alterations in ERα conformation in the presence of various ERE sequences, we analyzed the sensitivity of ERα to α-chymotrypsin digestion. An earlier study showed that chymotrypsin or trypsin digestion of MCF-7 cell extracts resulted in different sizes of ERα fragments in cells treated with E2 versus tamoxifen (TAM) (Hutchens et al., 1987). This result was an early indication that the conformation of E2-occupied ERα is different from TAM-occupied ERα, a finding confirmed by X-ray crystal structure studies of purified ERα LBD occupied by 4-OHT versus E2 or DES (Brzozowski et al., 1997, Shiau et al., 1998). A more recent report showed that specific amino acid (aa) mutations that constitutively activate ERα result in a proteolytic digestion pattern similar to that of the E2-liganded wild-type (wt) ERα (Lazennec et al., 1997). These reports indicate that alterations in ERα conformation correlate with ERα agonist activity, whether induced by E2 binding or by alterations in aa sequence. Because ER binding to an ERE activates gene expression from that element, it is important to investigate if there is a similar correlation between ERE-driven transcription and altered ER conformation.

Two reports conflict on the effect of DNA binding on ERα conformation as measured by sensitivity of ERα to trypsin digestion (Fritsch et al., 1992, Wood et al., 1998). One report showed that rat uterine ERα conformation is unaffected by DNA binding (Fritsch et al., 1992). The other report showed that yeast-expressed, recombinant human (rh) ERα is more sensitive to trypsin or chymotrypsin digestion when bound to the Xenopus vitellogenin A2 ERE than when ERα was incubated with the ERE from the human pS2 gene, containing a single nucleotide change in the 3’ half-site (Wood et al., 1998). The latter is important because, although many investigations of ERα activity utilize the perfectly palindromic Xenopus vitellogenin A2 ERE, the promoters of most natural estrogen responsive genes contain one or more imperfect EREs and/or ERE half-sites (Anolik et al., 1995). No one has examined how the ligand affects ERα conformation in the presence of perfect versus imperfect EREs.

In the present study, we assessed changes in the conformation of ERα in the presence or absence of a consensus ERE, naturally occurring non-palindromic EREs, or indirect and direct repeats (IR or DR, respectively) of the ERE half-site. We report that ERE binding enhances the sensitivity of ERα to proteolysis by chymotrypsin. In contrast, incubation of ERα with DNA sequences that do not bind ERα does not alter ERα sensitivity to proteolysis. We detected differences in the sensitivity of ERα to chymotrypsin digestion in the presence of different EREs and some changes in the sizes of proteolytic fragments with E2 or 4-OHT. Although the affinity of ERα for the different EREs does not correlate with transcriptional activity in transiently transfected cells, the sensitivity to chymotrypsin digestion correlates with reporter gene expression. These results are consistent with a role for the sequence of the ERE as an allosteric regulator of ERα activity.

Section snippets

Preparation of ERE-containing OLIGOMERs

The sequences of EREs used are included in Table 1. The EREs, EREc38, PR-1148, pS2, and Fos-1211, were cloned into pGEM-7Zf(+) or pGL3-promoter (pro) luciferase reporter vector (Promega, Madison, WI) as described (Klinge et al., 1997a, Klinge et al., 1997b, Klinge et al., 1997c). ERE oligomers were prepared by restriction digestion of the pGL3-pro-luciferase plasmid DNA with HinfI and XhoI or by digestion with EcoRI and BamH1 from pGEM-7Zf(+) and electroelution (Klinge et al., 1996). The

ERE binding increases ERα susceptibility to chymotrypsin digestion

To investigate whether ERE binding induces conformational changes in ERα, rh ERα, occupied by either E2 or 4-OHT, was incubated with limited concentrations of chymotrypsin. Fig. 1 shows four representative ERα chymotrypsin digestion experiments. Western blot analysis was performed with Ab10, a C-terminal specific monoclonal antibody to ERα. Without chymotrypsin treatment, a prominent band was detected at approximately 66 kDa (Fig. 1A–D). This corresponds in size to intact ERα. Identical results

Discussion

ERα and ERβ are members of the nuclear receptor superfamily that mediate the complex signaling processes by which estrogens regulate physiological functions, including mammary gland differentiation, reproduction, cholesterol metabolism, and bone homeostasis. Binding of E2 within the hydrophobic core of the LBD activates the receptor and facilitates high-affinity ER binding to EREs. Although most natural EREs are not perfect palindromes, the majority of transcriptional activation studies have

Acknowledgements

We thank Rosemary L. Sims and Kelly E. Risinger, for their assistance in some of the experiments reported here, and Timothy L. Ramsey for assistance in calculating the MW of chymotryptic ERα fragments. We thank Dr Barbara J. Clark for her suggestions on this manuscript.

References (51)

  • S.J. Hubbard

    The structural aspects of limited proteolysis of native proteins

    Biochim. Biophys. Acta

    (1998)
  • M. Ikeda et al.

    Different DNA elements can modulate the conformation of thyroid hormone receptor heterodimer and its transcriptional activity

    J. Biol. Chem.

    (1996)
  • N.H. Ing et al.

    Members of the steroid hormone receptor superfamily interact with TFIIB (S300-II)

    J. Biol. Chem.

    (1992)
  • P.S. Jones et al.

    Activation of transcription by estrogen receptor alpha and beta is cell type- and promoter-dependent

    J. Biol. Chem.

    (1999)
  • C.M. Klinge et al.

    Antiestrogen-liganded estrogen receptor interaction with estrogen responsive element DNA in vitro

    J. Steroid Biochem. Mol. Biol.

    (1992)
  • C.M. Klinge et al.

    Dissociation of 4-hydroxytamoxifen, but not estradiol or tamoxifen aziridine, from the estrogen receptor when the receptor binds estrogen response element DNA

    J. Steroid Biochem. Molec. Biol.

    (1996)
  • C.M. Klinge et al.

    Hsp70 is not required for high affinity binding of purified calf uterine estrogen receptor to estrogen response element DNA in vitro

    J. Steroid Biochem. Mol. Biol.

    (1997)
  • C.M. Klinge et al.

    COUP-TF interacts with estrogen receptor, binds to estrogen response elements and half-sites, and modulates estrogen-induced gene expression

    J. Biol. Chem.

    (1997)
  • C.M. Klinge et al.

    Comparison of tamoxifen ligands on estrogen receptor interaction with estrogen response elements

    Mol. Cell. Endocrinol.

    (1998)
  • C.M. Klinge et al.

    The aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT) heterodimer interacts with naturally occurring estrogen response elements

    Mol. Cell. Endocrinol.

    (1999)
  • C.M. Klinge

    Estrogen receptor interaction with co-activators and co-repressors

    Steroids

    (2000)
  • D.J. Mangelsdorf et al.

    The nuclear receptor superfamily: the second decade

    Cell

    (1995)
  • E.J. Pavlik et al.

    Hydroxylapatite “batch” assay for estrogen receptor: Increased sensitivity over present receptor assays

    J. Steroid Biochem.

    (1976)
  • A.K. Shiau et al.

    The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen

    Cell

    (1998)
  • T.L. Towers et al.

    Granulocyte-macrophage colony-stimulating factor gene transcription is directly repressed by the vitamin D3 receptor. Implications for allosteric influences on nuclear receptor structure and function by a DNA element

    J. Biol. Chem.

    (1998)
  • Cited by (80)

    • Allosteric pathways in nuclear receptors — Potential targets for drug design

      2018, Pharmacology and Therapeutics
      Citation Excerpt :

      It is speculated that this LBD-targeted inverse agonist also functions through allostery to induce conformational changes within the DBD that decreases the CAR-CARE binding affinity. In other studies, when the two ERα ligands E2 and 4-hydroxytamoxifen, 4-OHT are used in concert with different EREs there are notable differences in the sensitivity of each ER(+ ligand)-ERE molecular complex to digestion by the protease chymotrypsin (Klinge, Jernigan, Smith, Tyulmenkov, & Kulakosky, 2001). Thus, different ERE-ligand combinations appear to induce distinct conformations in ERα.

    • How glucocorticoid receptors modulate the activity of other transcription factors: A scope beyond tethering

      2013, Molecular and Cellular Endocrinology
      Citation Excerpt :

      However, direct evidence is lacking so far. Although the mechanism of allostery readily explains that DNA binding affinity correlates only poorly to transcriptional activity, as observed by several groups (Geserick et al., 2005; Klinge et al., 2001; Meijsing et al., 2009), the disconcordance between energetics and activity has recently been challenged using a different analysis approach (Bain et al., 2012). Bain and coworkers claim that despite the complexity of GR function, DNA binding energetics remain a primary determinant of sequence-specific transcriptional activity and they consider other regulatory mechanisms as minor contributors.

    • DHEA metabolites activate estrogen receptors alpha and beta

      2013, Steroids
      Citation Excerpt :

      There are many reasons that could account for the different responses of ERα detected with 16α-OH-DHEA, 7-oxo-DHEA, or ADIONE treatment in HEK-293 cells between the ERE-luciferase assay and induction of endogenous TFF1 expression. First, the ERE-luciferase reporter is an artificial construct with three tandem perfect, consensus EREs, which binds ERs with high affinity [31,73,74], compared to the endogenous TFF1 which is regulated by imperfect ERE and an AP-1 element [75]. Second, endogenous genes have chromatin structure and epigenetic modifications that have profound effects on transcription.

    View all citing articles on Scopus
    1

    Supported by NIH R01 DK 53220 and a University of Louisville School of Medicine Research Grant to C.M.K.

    View full text