steroid hor mon

Upload: chazmarilyn

Post on 07-Apr-2018

232 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 Steroid Hor Mon

    1/14

    1

    Endocrine-Related Cancer(1998) 5 1-14

    Endocrine-Related Cancer(1998) 5 1-14 1998 Journal of Endocrinology Ltd Printed in Great Britain1351-0088/98/005-001 $08.00/0

    Introduction

    The ability of oestrogens and androgens to stimulate thegrowth of a number of endocrine cancers is well

    established and several endocrine therapies are widelyused to reduce the availability of the hormones or to blocktheir action. These include non-steroidal hormoneantagonists such as tamoxifen and flutamide, steroidalcompounds which include ICI 182780 and cyproteroneacetate, and both steroidal and non-steroidal aromataseinhibitors. Although we have learned a great deal about themolecular mechanism of steroid hormone action, it is stillunclear how hormones stimulate the proliferation oftumour cells and how hormone antagonists function. Inpart this is a result of the ability of oestradiol andtestosterone to regulate the expression of many proteinsimplicated in the control of cell proliferation making it

    difficult to identify the crucial targets. Some of thesetargets are growth factors and/or their receptors whichsuggests that the mitogenic effects of steroids may bemediated by indirect autocrine or paracrine mechanisms(Clarke et al. 1991, Roberts & Sporn 1992). Alternativelysince steroids regulate the expression of certain cyclins orkinase inhibitors (Musgrove & Sutherland 1994, Altucciet al. 1996) they may control cell cycle progressiondirectly. Recent work suggests that as well as the cyclinD1 gene, cyclin D1 itself may be a crucial target (Zwijsenet al. 1997) but additional proteins could also be importantin different subsets of tumours.

    The identification of target genes for steroid hormones

    is further complicated by the observation that receptorsignalling is cross-coupled with that of other signallingpathways. It used to be thought that signalling by receptorswas relatively straightforward compared with most othersignalling pathways, since the receptor itself is atranscription factor. It is now clear, however, that growthfactors, neurotransmitters and other hormones are able tomodulate the activity of steroid hormone receptors (Poweret al. 1991, Aronica & Katzenellenbogen 1993, Ignar-Trowbridge et al. 1993). This means that alterations in theactivity of receptors and the expression of individual targetgenes involved in cell proliferation is determined not only

    by hormonal signals but also by changes in othersignalling pathways, which undoubtedly take place duringbreast and prostate cancer progression. Receptors are alsocapable of regulating the activity of a number of other

    transcription factors, either directly or indirectly, andthereby modulate the ability of other signalling pathwaysto control gene transcription (Shemshedini et al. 1991,Philips et al. 1993, Stein & Yang 1995, Webb et al. 1995).

    Although the precise role of steroid hormones in cellproliferation is still ill-defined, tremendous progress hasbeen made in elucidating the role of hormones in receptoractivation and the mechanism by which hormoneantagonists block this activity. In particular two recentadvances have been made which provide new insights intosteroid hormone action and the function of nuclearreceptors in general. First, models for the three-dimensional structure of the ligand binding domain of

    several receptors have been determined (Bourguet et al.1995, Renaud et al. 1995, Wagner et al. 1995, Brzozowskiet al. 1997) and secondly, novel proteins have beenidentified (Cavaills et al. 1994, Halachmi et al. 1994)which interact with receptors in a ligand dependentmanner and may play a role in gene transcription. Thesedevelopments are helping to provide a betterunderstanding of the mechanism of action of bothhormones agonists and antagonists and the links betweennuclear receptors and other signalling pathways. In thisreview we will outline these advances, with reference tothe oestrogen receptor (ER), and discuss their relevance toantioestrogen therapy and tamoxifen resistance.

    Steroid hormone receptors are membersof the nuclear receptor family

    Steroid hormone receptors are members of the nuclearreceptor family, a large group currently totallingapproximately 150 different proteins, which function astranscription factors in many different species includingboth invertebrates and vertebrates. In addition to steroidhormone receptors, the nuclear receptor family consists ofreceptors for retinoids, thyroid hormone, fatty acids andprostaglandins and a number of so-called orphan

    Molecular mechanisms of steroid

    hormone actionR White and M G Parker

    Molecular Endocrinology Laboratory, Imperial Cancer Research Fund, 44 Lincolns Inn Fields,London WC2A 3PX, UK

    (Requests for offprints should be addressed to M G Parker)

  • 8/6/2019 Steroid Hor Mon

    2/14

    White and Parker: Molecular mechanisms of steroid hormone action

    2

    Figure 1 (A) Organisation of functional domains in the nuclear receptors. DBD and LBD refer to the

    positions of the DNA binding domain and the ligand binding domain respectively. The relative

    positions of the two transactivation domains AF1 and AF2 are also shown. (B) Schematic diagram

    of steroid hormone receptor binding to DNA. The receptors bind as homodimers to palindromic

    sequences. The specific sequences of simple response elements for the ER and other steroid

    hormone receptors are shown.

  • 8/6/2019 Steroid Hor Mon

    3/14

    Endocrine-Related Cancer(1998) 5 1-14

    3

    receptors, the ligands of which have yet to be identified(Parker 1993, Mangelsdorfet al. 1995). They are highlyrelated in both primary amino acid sequence and the

    organisation of functional domains suggesting that manyaspects of their mechanism of action are conserved. Thus,progress in our understanding of steroid hormone actionhas been facilitated by studies of many nuclear receptorfamily members.

    The sex steroid receptors are not only expressed in sexaccessory tissues but in many other types of cellsincluding liver, bone, pituitary and cardiovascular cells.Androgen receptors are encoded by a single gene whereasthere are two genes for ERs, ER and ER (Kuiper et al.1996, Mosselman et al. 1996). Our current knowledge ofthe distribution of ER (Kuiper et al. 1997) is based onmRNA analysis either by RT-PCR or by in situ

    hybridisation and the relative levels of ER and ERprotein levels have yet to be determined. Analysis of micemade devoid of ER by targeted gene disruption(Bocchinfuso & Korach 1977) indicates that this receptoris essential for uterine growth and mammary glanddevelopment but not for mediating the inhibitory effects ofoestrogens in vascular injury (Jafrati et al. 1997). Thus, itis possible that ER and ER have distinct functions insome tissues but not in others. Undoubtedly the role ofthese individual receptors will be confirmed in the futureby gene knock-out experiments to generate mice lackingeither both receptors or one isoform only and analysing theresulting phenotypes.

    A simplified diagram of the organisation of functionaldomains in nuclear receptors is shown in Fig. 1A. Thereceptors are characterised by a highly conserved DNAbinding domain and a moderately conserved ligandbinding domain which also functions in dimer formationand transcriptional activation (Parker 1993, Mangelsdorfet al. 1995). A second dimerisation domain is locatedwithin the DNA binding domain itself and the receptorscontain two transcriptional activation functions by whichthey are able to regulate the expression of target genes;AF1, which is located in the N-terminal or A/B domain,and AF2 in the ligand binding domain. When activated byhormone binding, the receptors may act directly as

    transcription factors by binding to specific DNAsequences, termed hormone response elements, found inthe vicinity of target genes (Evans 1988, Beato 1989). Thesequences of simple binding sites for the steroid receptorsare shown in Fig. 1B. Composite response elements havealso been identified which bind receptors in addition toother transcription factors such that the binding of oneinfluences, either positively or negatively, the activity ofthe other (Diamond et al. 1990). In addition, it is nowevident that nuclear receptors are also capable ofregulating the transcription of genes that lack hormoneresponse elements by modulating the activity of other

    transcription factors such as AP-1 (Philips et al. 1993,Webb et al. 1995) and NF-B (Stein & Yang 1995). Todate most of the analysis which has been carried out has

    been to determine the mechanism by which receptorsfunction as DNA dependent transcription factors but it islikely that the transcription of many genes is regulatedindirectly by non-classical mechanisms.

    Nuclear receptors function as liganddependent transcription factors

    In the absence of hormone, steroid hormone receptorsexist as inactive oligomeric complexes with a number ofother proteins including chaperon proteins, namely theheat shock proteins Hsp90 and Hsp70 and cyclophilin-40and p23 (Smith & Toft 1993, Pratt & Toft 1997). The role

    of Hsp90 and other chaperons may be to maintain thereceptors folded in an appropriate conformation torespond rapidly to hormonal signals. Whether differenttypes of receptor complexes occur in cells, eachcontaining a subset of chaperons and able to respond todifferent types of signal, or whether receptors are foldedinto a mature complex containing chaperons in anassembly line type of process is as yet unclear (Bohen etal. 1995, Pennisi 1996). Following hormone binding, theoligomeric complex dissociates allowing the receptors tofunction either directly as transcription factors by bindingto DNA in the vicinity of target genes or indirectly bymodulating the activity of other transcription factors.

    Nuclear receptors may be classified into four typesbased on their dimerisation and DNA binding properties.Steroid hormone receptors bind to DNA as homodimerseither to simple response elements comprising shortpalindromic sequences or to composite response elementswith other transcription factors. Oestrogen responseelements consist of inverted repeats of the sequence A/GGGTCA separated by three nucleotides, whereas thereceptors for androgens, glucocorticoids and progestinsbind to inverted repeats of the sequence AGA/GACA, alsoseparated by three nucleotides. In contrast, a second groupof nuclear receptors, including the retinoic acid receptor(RAR), thyroid hormone receptor (TR) and vitamin D

    receptor, bind to DNA as heterodimers in combinationwith the retinoid X receptor (RXR) (Mangelsdorf et al.1995). The binding sites for these receptors consist ofdirect repeats of the sequence A/GGTCA separated by upto five nucleotides with the DNA binding specificity ofdifferent receptors being determined to some extent by thespacing of the repeats. In addition RXR itself and someorphan receptors bind to direct repeats as homodimerswhile a final group of orphan receptors are also able tobind as monomers to single copies of the extended bindingsite sequence TCAAGGTCA. The major part of thedimerisation interface is formed from sequences within

  • 8/6/2019 Steroid Hor Mon

    4/14

    White and Parker: Molecular mechanisms of steroid hormone action

    4

    the ligand binding domain of the receptors while a second

    dimerisation function is located within the DNA bindingdomain itself (Kumar & Chambon 1988, Fawell et al.1990a).

    Transcriptional activation by steroid receptors ismediated by at least two distinct activation functions; one,referred to as AF1, is located in the N-terminal domain anda second, AF2, is in the hormone binding domain (Lees etal. 1989, Tora et al. 1989). In the ER, and probably in otherreceptors as well, the absolute and relative activities ofthese two domains vary depending on the target promoterand the cell type. However, while the two activationdomains have the potential to act independently, they

    appear to interact with each other in the context of the

    intact receptor in a way which has yet to be determined.The N-terminal activation function, AF1, variesconsiderably in both size and primary amino acidsequence in different steroid hormone and nuclearreceptors. A number of studies have shown that thisdomain is a target for phosphorylation by other signallingpathways (Ali et al. 1993, Kato et al. 1995, Bunone et al.1996). The second activation function, AF2, is induced byhormone binding and a short sequence encoding anamphipathic -helix in the C-terminal part of the ligandbinding domain, which is conserved in all trans-criptionally active nuclear receptors, has been shown to be

    Figure 2 Diagrams of models of the structure of the ligand binding domains of nuclear

    receptors to demonstrate the effect of agonist binding. Panel A shows the arrangement of

    sequence elements for three different receptors, human RXR in the absence of ligand and

    human RARand rat TR in the presence of their respective ligands. The numbers 1 to 12

    correspond to separate -helices and s1 to s4 to -strands. The position of amino acids which

    are in close contact with ligand are marked and are found in helices 3, 5, s1 and s3, the loop

    between helices 6 and 7 and helices 11 and 12. Panels B and C show diagrams of the folded

    structure in the absence and presence of ligand and the relative positions of helices 1 to 12

    (H1-H12). This shows the realignment of helices 10 and 11 and the alteration in position of

    helix 12 to form a lid over the ligand binding pocket. This repositioning results in the formation

    of a new surface on the ligand binding domain, indicated by the white circles, which is believed

    to be required for the interaction with coactivator proteins.

  • 8/6/2019 Steroid Hor Mon

    5/14

    Endocrine-Related Cancer(1998) 5 1-14

    5

    essential in receptor function (Danielian et al. 1992,

    Saatcioglu et al. 1993, Barettino et al. 1994, Durand et al.1994). However, it is doubtful whether this short sequencefunctions autonomously since mutation of conservedresidues in other parts of the ligand binding domain, whichdo not affect ligand binding or dimerisation, also generatetranscriptionally defective receptors suggesting that AF2is derived from a number of different elements (ODonnell& Koenig 1990, Henttu et al. 1997).

    Structure of the ligand binding domain ofnuclear receptors

    A major advance in our understanding of how nuclearreceptors function has been the determination of modelsfor the ligand binding domain based on crystal structuresof three different receptors either in the absence orpresence of their cognate ligands. The secondary structureof all three nuclear receptors is shown in Fig. 2A andschematic diagrams of models of the structures obtainedin the absence and presence of ligand in Fig. 2B and Crespectively. The structure of the ligand binding domain ofRXR crystallised in the absence of ligand, consists of 12-helices and two -strands arranged in three layers toform an antiparallel -helical sandwich (Bourguet et al.1995). This receptor was crystallised in the form of adimer with the dimer interface formed mainly from helices9 and 10. Subsequently, crystals of the ligand binding

    domains of RAR(Renaud et al. 1995) and TR (Wagneret al. 1995) generated in the presence of their respectiveligands were shown to have a similar overall structure, but

    with one major exception. The C-terminal helix, helix 12,

    which protrudes beyond the core of the ligand bindingdomain in the unliganded RXR, is folded back across thecore of the domain in both RARand TR. It has thereforebeen proposed that in the case of RAR, ligand bindingresults in a realignment of helices 10 and 11 which re-arrange to form a continuous helix. This creates ashortened helix 12, corresponding to the amphipathic -helix described earlier, which is released from the core ofthe domain to fold back over the top of the ligand bindingpocket where it forms both contacts with the ligand and asalt bridge in helix 4. Helix 12 is similarly aligned in thecase of the TR although a salt bridge is not formed.Protein sequence comparisons between different receptorssuggest that their overall helical structure is conserved(Wurtz et al. 1996) and unpublished results indicate thatthe structures of peroxisome proliferator activatedreceptor (PPAR) and ER are consistent with thisprediction. Thus it appears that a major role for ligandbinding is an alteration in the conformation of the ligandbinding domain to form a novel surface. This may thenallow the binding of coactivators and other regulatoryproteins.

    Recruitment of receptor interactingproteins

    Gene transcription depends on the formation of a pre-

    initiation transcription complex that includes basaltranscription factors and one of the roles of transcriptionalactivators, including steroid hormone receptors, is thought

    Table 1 Receptor interacting proteins.

    Predicted size

    Group Name(s) Protein (kDa) Receptor binding Function(s)

    I CBP/p300 CBP 265 ER, RAR, RXR, TR Histone acetylase

    p300 300 ER, RAR, RXR, TR Bind transcription factors

    p160 SRC-1a 156.7 ER, RAR, RXR, TR, PR

    RIP160 SRC-1e/NCoA-1 152.3

    ERAP160 TIF2 159.6 ER, RAR, RXR, TR, PR Form complexes with CBP/p300GRIP1

    NCoA-2

    pCIP 152 ER, RAR, TR, PR

    II ARA70 ARA70 70 AR Androgen receptor coactivator

    RIP140 RIP140 128 ER, RAR, RXR, TR, PR Multiple nuclear receptor

    binding sites

    TIF1 TIF1 112 ER, RAR, TR, PR Chromatin remodelling?

    SUG1 SUG1 46 RAR, RXR, ER, TR, PR, VDR Associated with proteosome

    TRIP1

    AR, androgen receptor; PR, progesterone receptor; VDR, vitamin D receptor.

  • 8/6/2019 Steroid Hor Mon

    6/14

    White and Parker: Molecular mechanisms of steroid hormone action

    6

    to be the stabilisation of this complex. Although receptorshave been shown to bind directly to basal transcriptionfactors in vitro the significance of these interactions is

    unclear since they are unaffected by mutations in thereceptor that destroy transcriptional activity (Sadovsky etal. 1995). The involvement of additional targets is sug-gested by the observation that AF2 activity is inhibitedwhen receptors are overexpressed suggesting that limitingdownstream target proteins, required for gene trans-cription, are being sequestered (Tasset et al. 1990). Anumber of candidate target proteins have been detected inextracts from mammalian cells which bind to the ligandbinding domain of receptors in a ligand dependent manner(Cavaills et al. 1994, Halachmi et al. 1994). Since theseproteins fail to interact with mutant receptors which aretranscriptionally inactive it has been proposed that they

    may have a role in ligand dependent transcriptionalactivation by AF2.

    Receptor interacting proteins may be classified intotwo groups (Table 1). The first group consists of twofamilies of proteins which appear to stimulate trans-cription and represent bona-fide coactivators. One family,originally identified as proteins of 160 kDa, consists of anumber of related proteins; the steroid receptor coactivatorproteins (SRC-1a and SRC-1e) (Onate et al. 1995, Kameiet al. 1996), transcription intermediary factor TIF2(Voegel et al. 1996) (independently identified as GRIP-1(Hong et al. 1996)) and CREB binding protein (CBP)interacting protein (p/CIP) (Torchia et al. 1997). The

    second family comprises CBP and p300, which wereoriginally shown to function as coactivators for cAMPresponse element binding protein (CREB), the trans-cription factor that mediates responses to protein kinase Astimulation. Subsequently, however, CBP/p300 wereshown to function as coactivators for many othertranscription factors and may play a central role in manysignalling pathways (Janknecht & Hunter 1996, Shikamaet al. 1997).

    The second group contains a number of unrelatedproteins which also interact with the ligand bindingdomain but their role in receptor function is still unclear.This group includes a 140 kDa protein termed RIP 140

    (Cavaills et al. 1995), TIF1 (Le Douarin et al. 1995),ARA70 (Yeh & Chang 1996), which has been shown tostimulate transactivation mediated by the androgenreceptor, and TRIP1/SUG1 (Lee et al. 1995, vom Baur etal. 1996). RIP 140 stimulates receptor mediated trans-cription in yeast (Joyeux et al. 1996) and contains anactivation domain which is functional in mammaliancells (LHorset et al. 1996) but it is still unclear whether itis a bona-fide coactivator. TIF1 has been shown to interactwith proteins associated with heterochromatin and mayplay a role in chromatin remodelling (Le Douarin et al.1996). All these proteins have been shown to contain one

    of more copies of a short sequence motif LXXLL which isnecessary and sufficient to mediate the binding of ligandbound nuclear receptors. This motif has been described as

    a signature sequence for the interaction of proteins withnuclear receptors (Heery et al. 1997, Torchia et al. 1997).

    Other proteins have also been isolated which interactwith other regions of the ligand binding domain. Forexample two related proteins, N-CoR and SMRT, whichfunction as repressors for RXR and TR in the absence ofligand (Horlein et al. 1995, Heinzel et al. 1997, Nagy etal. 1997), have been reported to bind to oestradiol andprogesterone receptors in the presence of antagonists(Jackson et al. 1997, Smith et al. 1997). A detaileddescription of all these proteins, both activators andrepressors, is beyond the scope of this article; they arereviewed in detail elsewhere (Horwitz et al. 1996, Heery

    & Parker 1997) and therefore we will focus on theimportance of coactivators in steroid hormone actiontogether with potential interactions between receptors andother transcriptional regulators.

    Structure and function of coactivators fornuclear receptors

    To date, it appears that the p160 proteins and CBP/p300are the most important of the receptor interacting proteinsin terms of their ability to potentiate the transcriptionalactivity of receptors. The p160 proteins SRC1, TIF2 andp/CIP share significant sequence homology, most notably

    in a central region found to be required for interaction withthe ligand binding domain of nuclear receptors. Theyshow ligand dependent binding to nuclear receptors invitro but fail to bind to transcriptionally inactive receptorsin which mutations have been introduced in the conservedhydrophobic residues in helix 12 (Kamei et al. 1996).Similarly mutations in conserved residues in helix 3 whichalso impair AF2 also prevent binding of these coactivators(Henttu et al. 1997). This suggests that, in the presence ofligand, helices 3 and 12 form a composite surfacerecognised by the p160 proteins, a hypothesis which isconsistent with the models of the holo ligand bindingdomains. Similarly, CBP/p300 have been reported to bind

    to the ligand binding domains of nuclear receptors in aligand dependent manner and increase AF2 activity intransfected cells (Chakravarti et al. 1996, Hanstein et al.1996, Kamei et al. 1996, Yao et al. 1996). This binding isalso dependent on the integrity of helix 12, implying thatthese proteins recognise a similar if not identical bindingsite (Heery et al. 1997). The p160 and CBP/p300 familymembers also interact directly with one another (Hansteinet al. 1996, Kamei et al. 1996, Yaoet al. 1996) but we havelittle information about the stoichiometry of binding ofany of these proteins to receptors. For example, it isunclear whether the proteins bind to receptors

  • 8/6/2019 Steroid Hor Mon

    7/14

    Endocrine-Related Cancer(1998) 5 1-14

    7

    independently or as complexes, whether they bindsequentially or simultaneously, which may pose stericproblems given their relative sizes, and whether there is

    redundancy or receptor selectivity. These questions arebeing tackled by many laboratories at the present time.

    When coactivators are recruited to receptors they mayserve at least two distinct roles. First, they may stabilisethe formation of a pre-initiation transcription complex ofDNA polymerase and basal transcription factors, andsecondly they may remodel chromatin by destabilising thebinding of nucleosomes. Regions required for trans-criptional activation have been found in both p160 andCBP/p300 family members. These have been identified byfusing fragments of these proteins to a DNA bindingdomain and testing their ability to stimulate transcriptionof reporter genes in transfected cells. In SRC1, one

    corresponds to the region involved in CBP binding andmay reflect the recruitment of CBP and/or p300 to thepromoter. In CBP, an activation domain corresponds to aregion that binds to TATA-box binding protein (TBP) andso may directly interact with the basal transcriptionmachinery. Both families of proteins also encode histoneacetyl transferases that acetylate lysine residues onspecific histones which may destabilise nucleosomes topromote gene transcription (Ogryzko et al. 1996). Inaddition, they are also reported to be capable of recruitingP/CAF, another histone acetyl transferase (X-J Yang et al.1996). What emerges from these studies is that co-activators are multidomain proteins that presumably

    function either independently or as complexes and thegoal is now to determine the precise roles and relativeimportance of each of these domains when they arerecruited by different receptors to target genes. A modelshowing some of the functions of coactivator proteins isshown in Fig. 3.

    Role of hormonal agonists andantagonists in coactivator recruitment

    Although crystal structures have yet to be solved for theligand binding domain of any one receptor in the ligandedand unliganded state, the overall similarity of the

    structures for apo-RXR and holo-RAR and the high

    level of sequence conservation in the nuclear receptorfamily, allows a model to be proposed for the mechanismof action of hormone agonists and antagonists. The

    realignment of helix 12 as a lid over the ligand bindingpocket provides a potential molecular explanation for thedisparate activities of different ligands. Ligands whichfunction as agonists would be expected to allow theformation of an interacting surface made up from residuesin helices 3 and 12. In contrast, ligands that fail to allowthe correct realignment of helix 12 are likely to functionas antagonists. Interestingly, although the structure of theligand appears to determine its affinity for binding to thereceptor it does not seem to be as important in determiningAF2 activity, implying that the role of agonists may besimply to release helix 12 from an inactive conformationrather than to promote its correct realignment. This may

    be true in the case of the ER since many environmentaloestrogens that bind relatively poorly still function asagonists (Soto et al. 1991, White et al. 1994, Jobling et al.1995).

    Hormone antagonists may be classified into two basictypes, either mixed agonists/antagonists or pure ant-agonists, exemplified by the antioestrogens tamoxifen andICI 182780 respectively. The binding site in the receptorfor both types of compound overlaps with that of oestra-diol and so they act as competitive inhibitors of oestrogenaction (Dauvois & Parker 1993). In vitro assays andtransient transfection studies suggest that tamoxifenbinding allows the receptor to dimerise and bind to DNAwith high affinity but blocks transcriptional activitymediated by AF2. The agonist activity is thought to bederived primarily from AF1, which is active even whentamoxifen is bound to the receptor (Berry et al. 1990,Danielian et al. 1993, Tzukerman et al. 1994). Thereforetamoxifen has the potential to act as an antagonist whenthe transcriptional activity of the receptor is mediated byAF2 but as an agonist on promoters or in cell types whereAF1 is active. The pure antioestrogens, however, seem toblock the activity of both AF1 and AF2. Evidence isemerging that part of the mechanism of antagonism maybe a result of the recruitment of corepressors to thereceptor (Horwitz et al. 1996, Jackson et al. 1997, Smithet al. 1997). The lack of AF2 activity in the presence of

    Figure 3 Interactions between receptors and

    coactivator proteins. Activated receptors bind p160

    proteins and proteins of the CBP/p300 family in a ligand

    dependent manner. This DNA bound complex may

    then stimulate the expression of hormone responsive

    genes by a number of possible mechanisms including

    the remodelling of chromatin, interaction with other

    transcription factors and the stabilisation of the pre-

    initiation complex which consists of DNA polymerase

    and basal transcription factors.

  • 8/6/2019 Steroid Hor Mon

    8/14

    White and Parker: Molecular mechanisms of steroid hormone action

    8

    either type of antioestrogen is likely to reflect the incorrectrealignment of helix 12. This has recently been confirmedby the determination of models for the structure of theligand binding domain of the ER both in the presence ofoestradiol and in the presence of the antagonist raloxifene(Fig. 4). The major difference in the two structural modelsis an alteration in the positioning of helix 12 which is aconsequence of the need to accommodate the long, bulkyside chain of the antioestrogen in the ligand binding cavity(Brzozowski et al. 1997). The failure to correctly re-position helix 12 and the inability to form an appropriatesurface for the binding of coactivator proteins in thepresence of hormone antagonists may be analysed in vitro.Addition of either tamoxifen or ICI 182780 preventsbinding in assay systems in which coactivator proteins areanalysed for their ability to associate with the ligandbinding domain of the ER (Henttu et al. 1997). Similarlythese antagonists inhibit the stimulatory effects ofcoactivators such as SRC1 on receptor mediated trans-criptional activation in mammalian cells. Finally it may besignificant that many steroid antagonists, e.g. tamoxifen,ICI 182780 and RU486, are of greater size than the naturalligand suggesting that interference with the realignment ofhelix 12 may be a common mechanism of hormoneantagonism.

    In addition to their effects on the realignment of helix12 pure antioestrogens have been found to suppress the

    action of ERs in other ways. They clearly reduce theintracellular concentration of ERs by reducing their halflife (Gibson et al. 1991, Dauvois et al. 1992). In thepresence of ICI 182780, receptors accumulate in thecytoplasm as a consequence of a block in nuclear uptakeof the protein (Dauvois et al. 1993) and it is conceivablethat this aberrant accumulation and/or conformationtriggers degradation. It has also been proposed that ICI182780 blocks receptor dimerisation and, as aconsequence, inhibits DNA binding (Fawell et al. 1990b).These observations, however, were based on in vitroexperiments and have been questioned by a number ofworkers analysing the effects of antioestrogens in intactcells (Reese & Katzenellenbogen 1991, Metzger et al.1995).

    Although it may be possible to assign all anti-oestrogens as either mixed agonists/antagonists or pureantagonists it is unlikely that their mechanism of actionwill be precisely the same as either tamoxifen or ICI182780. Partial agonists are particularly interestingbecause their agonist activity is cell specific. Thus, bothraloxifene and tamoxifen act as antagonists in breastcancer and agonists in bone while their action differs in theendometrium where only raloxifene is an antagonist(Draper et al. 1996, N N Yang et al. 1996). This differencemust reflect distinct conformations induced in the receptorby the two antioestrogens which might, for example,

    Figure 4 A model for the structure of the ligand binding domain in the presence of hormone agonists and

    antagonists. Although antagonists may bind with high affinity the structure of the domain may be unable to

    fold to generate the surface required for the binding of other proteins and realignment of helix 12 with helices

    3 and 5 may be prevented. The helix numbers are based on those descr ibed by Brzozowski et al. (1997).

    These differ from those described by Wurtz et al. (1996) and presented in Fig. 2 due to the presence of an

    additional helix in ER, helix 4.

  • 8/6/2019 Steroid Hor Mon

    9/14

    Endocrine-Related Cancer(1998) 5 1-14

    9

    differentially affect the sensitivity of the N-terminalactivation domain to phosphorylation by other signallingpathways or alter interactions between AF1 and AF2.

    Ligand independent stimulation of ERs

    The transcriptional activity of the ER has been reported tobe modulated by a number of other signalling pathwaysinvolving different protein kinase cascades. The bestcharacterised of these is the ability of epidermal growthfactor (EGF) to modulate the activity of the ER. It wasinitially observed that the effects of oestrogen on themouse uterus can be mimicked by the administration ofEGF to animals and that this is a result of the activation ofthe ER in an oestrogen independent manner (Ignar-Trowbridge et al. 1992). These results have been

    confirmed by analysing the uterotrophic response in ERknock-out mice, where the oestrogen-like effects of EGFare absent, demonstrating that these responses aredependent on the expression of the ER (Curtis et al. 1996).The activation of ERs by EGF has been analysed in cell

    lines (Ignar-Trowbridge et al. 1993, 1996) and shown tobe mediated by the MAP kinase pathway whichphosphorylates a serine residue at position 118 in AF1 (Aliet al. 1993, Kato et al. 1995, Bunone et al. 1996).Phosphorylation of this residue seems to be required forAF1 activity but little is known about its role intranscriptional activation. Since AF1 is functional in thepresence of tamoxifen it is likely to be important in theagonist effects of this antioestrogen. Cell specificvariations in AF1 activity probably reflect differences inthe stimulation of the MAP kinase pathway in response togrowth factors and this may account for the cell-specificdifferences observed for the agonist activity of tamoxifen.Agents that increase cyclic AMP levels and stimulateprotein kinase A activity also increase transactivation bythe ER, an effect which was first demonstrated by treating

    cells with the neurotransmitter dopamine (Power et al.1991). However, it is not known whether this is achievedby phosphorylation of the receptor itself or other proteinsinvolved in transcriptional activation.

    Figure 5 Activation of the oestrogen receptor. The receptor may be activated by ligand binding and in addition by

    stimulation with growth factors acting via MAP kinase (MAPK) to phosphorylate AF1. The receptor may also be a

    target for tyrosine kinase(s) resulting in activation of AF2. Partial antagonists such as tamoxifen block ligand

    dependent activity but not growth factor stimulation of AF1. Pure antagonists prevent ligand dependent activation

    and may inhibit other pathways by completely blocking the action of the receptor protein.

  • 8/6/2019 Steroid Hor Mon

    10/14

    White and Parker: Molecular mechanisms of steroid hormone action

    10

    The ER is also modified by phosphorylation ontyrosine residues, in particular at Y537 in the humanprotein (Arnold et al. 1995), which is located at the N

    terminus of helix 12 in the hormone binding domain.Phosphorylation of this tyrosine residue is not absolutelyrequired for AF2 activity since it can be replaced withphenylalanine without affecting oestrogen dependentactivation (White et al. 1997). However, analysis of othermutations introduced at this position suggests thatphosphorylation may represent an alternative mechanismfor ligand independent activation of the ER for thefollowing reason. Replacement of the tyrosine residuewith charged residues, either aspartic acid or glutamicacid, which might mimic tyrosine phosphorylation,generates constitutively active receptors (Weis et al. 1996,White et al. 1997). We think that this is likely to occur as

    a consequence of the realignment of helix 12 to form theinteracting surface required to recruit coactivators.Surprisingly an alanine replacement or a serine at thisposition has a similar effect. One interpretation of theseresults is that the tyrosine residue takes part inhydrophobic interactions which maintain the receptor inan inactive state and that activation is achieved bydisruption of these interactions allowing the release ofhelix 12, analogous to the conformational change thatoccurs as a result of ligand binding. The importance of thisresidue, and the possibility that phosphorylation mayrepresent a means for activating the receptor, is suggestedfrom the sequence conservation between both ER and

    ER, which have been analysed from a number ofdifferent species all of which have a tyrosine residue inthis position. However, the identification of a growthfactor/kinase pathway which both phosphorylates andactivates the receptor at this position remains elusive. Asummary of the various pathways which may be involvedin the activation of the receptor is shown in Fig. 5.

    Interactions between nuclear receptorsand other transcription factors

    Interactions may also occur within the cell nucleusbetween receptors and other transcription factors, for

    example AP-1. This transcription factor is implicated as acritical target for many signalling pathways that regulatecell differentiation, proliferation and transformation(Angel & Karin 1991). It is important as a target forgrowth factors which stimulate the phosphorylation ofFos/Jun family members and may also play a role ingrowth inhibition by retinoids which have been shown toblock AP-1 activity (Pfahl 1993, Chen et al. 1995).Oestrogens, which stimulate the proliferation of MCF-7breast cancer cells, increase growth factor induced AP-1activity whereas antioestrogens, which inhibit growth ofthese cells, inhibit the activity of AP-1 (Philips et al. 1993,

    Webb et al. 1995). A number of alternative mechanismshave been described to explain how nuclear receptors maymodulate the activity of other transcription factors. These

    include the binding of receptors to DNA at compositeresponse elements in association with other factors, e.g.the control of proliferin gene expression by the gluco-corticoid receptor and AP-1 (Diamond et al. 1990), andthe oestrogen regulation of ovalbumin gene expression(Gaub et al. 1990). The recent discovery of the ability ofreceptors to associate with proteins such as CBP/p300,which themselves have the potential to make multiple con-tacts with other transcription factors, suggests a possiblemechanism for the integration of different signallingpathways at target gene promoters. For example, it hasbeen reported that the ability of receptors to repress AP-1activity results from the sequestering of CBP (Kamei et al.

    1996) although other factors, as yet unidentified, have alsobeen proposed (Saatcioglu et al. 1994, 1997).

    The problem of tamoxifen insensitivity andtamoxifen resistance

    There is no a priori reason why ER positive breasttumours should all be sensitive to tamoxifen treatmentsince their proliferation may be completely independent ofthe activity of the ER. Similarly, tumours may becometamoxifen resistant as a consequence of further geneticchanges that allow them to proliferate in the absence offunctional ER. However, a relatively high proportion of

    tamoxifen resistant tumours are still sensitive to alternateendocrine therapies suggesting that the proliferation of asubset of tumours is receptor dependent.

    The molecular basis for the tamoxifen resistance ofthis type of tumour is unknown but several possibilitieshave been proposed. A point mutation has been identifiedat amino acid 537 in the human ER from a breast tumoursample in which a tyrosine residue, previously shown tobe a site of protein modification by phosphorylation, asdescribed earlier, is altered to asparagine. This generates aconstitutively active protein which appears to beinsensitive to the affects of antioestrogens (Zhang et al.1997). However, mutation of this residue to a number of

    other amino acids, including alanine and serine, results inreceptor proteins which although constitutively active areinhibited by treatment with either tamoxifen or ICI182780 (Weis et al. 1996, White et al. 1997). Receptormutations (Jordan et al. 1995) and variants (Fuqua 1994,Fuqua et al. 1995) have also been proposed to account fortamoxifen insensitivity. In particular, a variant lacking theoestrogen binding domain has been reported to exhibitconstitutive activity and stimulate the proliferation ofbreast cancer cells in vitro in the presence of tamoxifen.We have been unable to confirm this observation when theexpression of this variant was induced in cell lines and we

  • 8/6/2019 Steroid Hor Mon

    11/14

    Endocrine-Related Cancer(1998) 5 1-14

    11

    conclude that changes in other signalling pathways mustalso be necessary (Rea et al. 1996). One candidate is theMAP kinase pathway, which potentiates the activity of the

    N-terminal activation domain, AF1, even in the presenceof tamoxifen. Tamoxifen, like oestrogen, seems topromote the binding of the receptor to stimulate theexpression of target genes and it is conceivable that AF1mediated transcription may therefore be increased in cellswith enhanced MAP kinase activity. These target genes,however, may remain sensitive to alternative endocrinetreatments. For example, ER dependent transcriptionwould remain sensitive to aromatase inhibitors becausethere would be insufficient available oestrogen to activatethe receptor. Similarly, ICI 182780 treatment would notonly reduce ER levels but may inhibit both activationdomains. Thus, the ability of alternative endocrine

    therapies to block tumour growth in tamoxifen resistantpatients might reflect the failure of the receptor to activatea subset of target genes even when the N-terminal domainAF1 is stimulated by other signalling pathways.

    Future prospects

    An understanding of the molecular mechanisms of actionof the ER is not only beginning to provide an explanationfor the function of clinically useful antioestrogens but isalso suggesting novel therapeutic targets. These include,for example, the interactions between coactivators andreceptors which can be disrupted by specific peptides invitro (Heery et al. 1997, Torchia et al. 1997). It maytherefore be possible to prevent these interactions in vivoand block the agonistic effects of oestrogens. The twoareas in which progress has been relatively slow in thepast, namely the identification of the crucial oestrogenregulated target genes involved in cell proliferation andthe mechanism of tamoxifen resistance, remain veryactive areas of research. Hopefully, with a betterunderstanding of both the mammalian cell cycle andalternate intracellular signalling mechanisms, moreprogress will continue to be made in the future.

    References

    Ali S, Metzger D, Bornert J-M & Chambon P 1993 Modulationof transcriptional activation by ligand-dependent phosphory-lation of the human oestrogen receptor A/B region.EMBO

    Journal 12 1153-1160.Altucci L, Addeo R, Cicatiello L, Dauvois S, Parker MG, Truss

    M, Beato M, Sica V, Bresciani F & Weisz A 1996 17-beta-estradiol induces cyclin d-1 gene-transcription, p36(d1)-p34(cdk4) complex activation and p105(rb) phosphorylationduring mitogenic stimulation of g(1)-arrested human breast-cancer. Oncogene 12 2315-2324.

    Angel P & Karin M 1991 The role of Jun, Fos and the AP-1complex in cell-proliferation and transformation.Biochimicaet Biophysica Acta 1072 129-157.

    Arnold SF, Obourn JD, Jaffe H & Notides AC 1995 Phosphory-lation of the human estrogen receptor on tyrosine-537 in vivoand by Src family tyrosine kinases in vitro. Molecular

    Endocrinology 9 24-33.Aronica SM & Katzenellenbogen B 1993 Stimulation of estrogen

    receptor-mediated transcription and alteration in thephosphorylation state of the rat uterine estrogen receptor byestrogen, cyclic adenosine monophosphate, and insulin-likegrowth factor-I.Molecular Endocrinology 7 743-752.

    Barettino D, Ruiz MDMV & Stunnenberg HG 1994Characterization of the ligand-dependent transactivationdomain of thyroid-hormone receptor.EMBO Journal 133039-3049.

    vom Baur E, Zechel C, Heery D, Heine MJS, Garnier JM, VivatV, Le Douarin B, Gronemeyer H, Chambon P & Losson R1996 Differential ligand-dependent interactions between theAF-2 activating domain of nuclear receptors and the putative

    transcriptional intermediary factors mSUG1 and TIF1.EMBOJournal15 110-124.Beato M 1989 Gene regulation by steroid hormones. Cell 56 335-

    344.Berry M, Metzger D & Chambon P 1990 Role of the two

    activating domains of the oestrogen receptor in the cell-typeand promoter-context dependent agonistic activity of the anti-oestrogen 4-hydroxytamoxifen.EMBO Journal 9 2811-2818.

    Bocchinfuso WP & Korach KS 1997 Biological impact of adisrupted estrogen receptor gene on estrogen-related cancer.

    Endocrine-Related Cancer4 387-406.Bohen SP, Kralli A & Yamamoto KR 1995 Hold em and fold

    em: chaperons and signal transduction. Science 268 1303-1304.

    Bourguet W, Ruff M, Chambon P, Gronemeyer H & Moras D1995 Crystal-structure of the ligand-binding domain of thehuman nuclear receptor RXR-alpha.Nature 375 377-382.

    Brzozowski AM, Pike ACW, Dauter Z, Hubbard RE, Bonn T,Engstrom O, Ohman L, Greene GL, Gustafsson J- &Carlquist M 1997 Molecular basis of agonism and antagonismin the oestrogen receptor.Nature 389 753-758.

    Bunone G, Briand P-A, Miksicek RJ & Picard D 1996 Activationof the unliganded estrogen receptor by EGF involves the MAPkinase pathway and direct phosphorylation.EMBO Journal15 2174-2183.

    Cavaills V, Dauvois S, Danielian PS & Parker MG 1994 Inter-action of proteins with transcriptionally active estrogenreceptors. Proceedings of the National Academy of Sciences

    of the USA91

    10009-10013.Cavaills V, Dauvois S, LHorset F, Lopez G, Hoare S, KushnerPJ & Parker MG 1995 Nuclear factor RIP140 modulatestranscriptional activation by the estrogen receptor.EMBO

    Journal14 3741-3751.Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T, Schulman

    IG, Juguilon H, Montminy M & Evans RM 1996 Role of CBP/P300 in nuclear receptor signalling.Nature 383 99-103.

    Chen JY, Penco S, Ostrowski J, Balaguer P, Pons M, Starrett JE,Reczek P, Chambon P & Gronemeyer H 1995 Rar-specificagonist/antagonists which dissociate transactivation and AP-1transrepression inhibit anchorage-independent cell-proliferation.EMBO Journal14 1187-1197.

  • 8/6/2019 Steroid Hor Mon

    12/14

    White and Parker: Molecular mechanisms of steroid hormone action

    12

    Clarke R, Dickson RB & Lippman ME 1991 The role of steroidhormones and growth factors in the control of normal andmalignant breast. InNuclear Hormone Receptors, pp 297-

    319. Ed. M Parker. London: Academic Press.Curtis SW, Washburn T, Sewall C, Diaugustine R, Lindzey J,

    Couse JF & Korach KS 1996 Physiological coupling ofgrowth-factor and steroid-receptor signaling pathways -estrogen-receptor knockout mice lack estrogen-like responseto epidermal growth-factor. Proceedings of the National

    Academy of Sciences of the USA93 12626-12630.Danielian PS, White R, Lees JA & Parker MG 1992

    Identification of a conserved region required for hormonedependent transcriptional activation by steroid hormonereceptors.EMBO Journal 11 1025-1033.

    Danielian PS, White R, Hoare SA, Fawell SE & Parker MG 1993Identification of residues in the estrogen receptor whichconfer differential sensitivity to estrogen and hydroxy-

    tamoxifen.Molecular Endocrinology7

    232-240.Dauvois S & Parker MG 1993 Mechanism of action of hormoneantagonists. In Steroid Hormone Action, pp 161-185. Ed. MParker. Oxford: IRL Press.

    Dauvois S, Danielian PS, White R & Parker MG 1992 Anti-estrogen ICI 164,384 reduces cellular estrogen receptorcontent by increasing its turnover.Proceedings of the National

    Academy of Sciences of the USA 89 4037-4041.Dauvois S, White R & Parker MG 1993 The antiestrogen ICI

    182780 disrupts estrogen receptor nucleocytoplasmicshuttling.Journal of Cell Science 106 1377-1388.

    Diamond MI, Miner JN, Yoshinaga SK & Yamamoto KR 1990Transcription factor interactions: selectors of positive ornegative regulation from a single DNA element. Science 2491266-1272.

    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 serum-lipid profile inhealthy postmenopausal women.Journal of Bone and Mineral

    Research11 835-842.Durand B, Saunder M, Gaudon C, Roy B, Losson R & Chambon

    P 1994 Activation function 2 (AF-2) of retinoic acid receptorand 9-cis retinoic acid receptor: presence of a conservedautonomous constitutive activating domain and influence ofthe nature of the response element on AF-2 activity.EMBO

    Journal 13 5370-5382.Evans RM 1988 The steroid and thyroid hormone receptor

    superfamily. Science 240 889-895.Fawell SE, Lees JA, White R & Parker MG 1990a

    Characterization and colocalization of steroid binding anddimerization activities in the mouse estrogen receptor. Cell 60953-962.

    Fawell SE, White R, Hoare S, Sydenham M, Page M & ParkerMG 1990b Inhibition of estrogen receptor-DNA binding bythe pure antiestrogen ICI 164,384 appears to be mediated byimpaired receptor dimerization. Proceedings of the National

    Academy of Sciences of the USA 87 6883-6887.Fuqua SAW 1994 Estrogen receptor mutagenesis and hormone

    resistance. Cancer74 1026-1029.Fuqua SAW, Wiltschke C, Castles C, Wolf D & Allred DC 1995

    A role for estrogen-receptor variants in endocrine resistance.Endocrine Related Cancer2 19-25.

    Gaub M-P, Bellard M, Scheuer I, Chambon P & Sassone-Corsi P1990 Activation of the ovalbumin gene by the estrogenreceptor involves the Fos-Jun complex. Cell 63 1267-1276.

    Gibson MK, Nemmers LA, Beckman Jr WC, Davis VL, CurtisSW & Korach KS 1991 The mechanism of ICI 164,384antiestrogenicity involves rapid loss of estrogen receptor inuterine tissue.Endocrinology 129 2000-2010.

    Halachmi S, Marden E, Martin G, MacKay H, Abbondanza C &Brown M 1994 Estrogen receptor-associated proteins -possible mediators of hormone-induced transcription. Science264 1455-1458.

    Hanstein B, Eckner R, DiRenzo J, Halachmi S, Liu H, Searcy B,Kurokawa R & Brown M 1996 p300 is a component of anestrogen receptor coactivator complex. Proceedings of the

    National Academy of Sciences of the USA 93 11540-11545.Heery DM & Parker MG 1997 Ligand induced transcription by

    nuclear receptors.Retinoids 13 26-30.

    Heery DM, Kalkhoven E, Hoare S & Parker MG 1997 Asignature motif in transcriptional co-activators mediatesbinding to nuclear receptors.Nature 387 733-736.

    Heinzel T, Lavinsky RM, Mullen T-M, Soderstrom M, LahertyCD, Torchia J, Yang W-M, Brard G, Ngo SD, Davie JR, SetoE, Eisenman RN, Rose DW, Glass CK & Rosenfeld MG 1997A complex containing N-Cor, mSin3 and histone deacetylasemediates transcriptional repression.Nature 387 43-48.

    Henttu PMA, Kalkhoven E & Parker MG 1997 AF-2 activity andrecruitment of steroid receptor coactivator 1 to the estrogenreceptor depend on a lysine residue conserved in nuclearreceptors.Molecular and Cellular Biology17 1832-1839.

    Hong H, Kohli K, Trivedi A, Johnson DL & Stallcup MR 1996Grip1, a novel mouse protein that serves as a transcriptionalcoactivator in yeast for the hormone-binding domains of

    steroid-receptors. Proceedings of the National Academy ofSciences of the USA 93 4948-4952.

    Horlein AJ, Naar AM, Heinzel T, Torchia J, Gloss B, KurokawaR, Ryan A, Kamei Y, Soderstrom M, Glass CK & RosenfeldMG 1995 Ligand-independent repression by the thyroidhormone receptor mediated by a nuclear receptor co-repressor.Nature 377 397-404.

    Horwitz KB, Jackson TA, Bain DL, Richer JK, Takimaoto GS &Tung L 1996 Nuclear receptor coactivators and corepressors.

    Molecular Endocrinology10 1167-1177.Ignar-Trowbridge DM, Nelson KG, Bidwell MC, Curtis SW,

    Washburn TF, McLachlan JA & Korach KS 1992 Coupling ofdual signaling pathways: epidermal growth factor actioninvolves the estrogen receptor. Proceedings of the National

    Academy of Sciences of the USA 89 4658-4662.Ignar-Trowbridge DM, Teng CT, Ross KA, Parker MG, KorachKS & McLachlan JA 1993 Peptide growth factors elicitestrogen receptor-dependent transcriptional activation of anestrogen-responsive element.Molecular Endocrinology7992-998.

    Ignar-Trowbridge DM, Pimentel M, Parker MG, McLachlan JA& Korach KS 1996 Peptide growth factor cross-talk with theestrogen receptor requires the A/B domain and occursindependently of protein kinase C or estradiol.Endocrinology137 1735-1744.

    Jackson TA, Richer JK, Bain DL, Takimoto GS, Tung L &Horwitz KB 1997 The partial agonist activity of antagonist-

  • 8/6/2019 Steroid Hor Mon

    13/14

    Endocrine-Related Cancer(1998) 5 1-14

    13

    occupied steroid receptors is controlled by a novel hingedomain-binding coactivator L7/SPA and the corepressors N-Cor or SMRT.Molecular Endocrinology 11 693-705.

    Jafrati MD, Karas RH, Aronovitz M, Kim S, Sullivan Jr TR,Lubahn DB, ODonnell Jr TF, Korach KS & Mendelsohn ME1997 Estrogen inhibits the vascular injury response inestrogen receptor -deficient mice.Nature Medicine 3 545-548.

    Janknecht R & Hunter T 1996 A growing coactivator network.Nature 383 22-23.

    Jobling S, Reynolds T, White R, Parker MG & Sumpter JP 1995A variety of environmentally persistent chemicals, includingsome phthalate plasticizers, are weakly estrogenic.

    Environmental Health Perspectives 103 582-587.Jordan VC, Catherino WH & Wolf DM 1995 A mutant receptor

    as a mechanism of drug resistance to tamoxifen treatment.Annals of the New York Academy of Sciences761 138-147.

    Joyeux A, Cavaills V, Balaguer P & Nicolas JC 1996 RIP 140enhances nuclear receptor-dependent transcription in vivo, inyeast.Molecular Endocrinology 11 193-202.

    Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, LinS-C, Heyman RA, Rose DW, Glass CK & Rosenfeld MG 1996A CBP integrator complex mediates transcriptional activationand AP-1 inhibition by nuclear receptors. Cell 85 403-414.

    Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, SasakiH, Masushige S, Gotoh Y, Nishida E, Kawashima H, MetzgerD & Chambon P 1995 Activation of the estrogen-receptorthrough phosphorylation by mitogen-activated protein-kinase.Science 270 1491-1494.

    Kuiper GGJM, Enmark E, Pelto-Huikko M, Nilsson S &Gustafsson J-A 1996 Cloning of a novel estrogen receptor

    expressed in rat prostate and ovary. Proceedings of theNational Academy of Sciences of the USA 93 5925-5930.Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Haggblad J,

    Nilsson S & Gustafsson J-A 1997 Comparison of the ligandbinding specificity and transcript tissue distribution ofestrogen receptors and .Endocrinology 183 863-870.

    Kumar V & Chambon P 1988 The estrogen receptor binds tightlyto its responsive element as a ligand-induced homodimer. Cell55 145-156.

    Le Douarin B, Zechel C, Garnier JM, Lutz Y, Tora L, Pierrat B,Heery D, Gronemeyer H, Chambon P & Losson R 1995 TheN-terminal part of TIF1, a putative mediator of the ligand-dependent activation function (AF-2) of nuclear receptors, isfused to B-raf in the oncogenic protein T18.EMBO Journal14

    2020-2033.Le Douarin B, Nielsen AL, Garnier J-M, Ichinose H, JeanmouginF, Losson R & Chambon P 1996 A possible involvement ofTIF1 and TIF1 in the epigenetic control of transcription bynuclear receptors.EMBO Journal 15 6701-6715.

    Lee JW, Ryan F, Swaffield JC, Johnston SA & Moore DD 1995Interaction of thyroid-hormone receptor with a conservedtranscriptional mediator.Nature 374 91-94.

    Lees JA, Fawell SE & Parker MG 1989 Identification of twotransactivation domains in the mouse oestrogen receptor.

    Nucleic Acids Research 17 5477-5488.LHorset F, Dauvois S, Heery DM, Cavaills V & Parker MG

    1996 RIP-140 interacts with multiple nuclear receptors by

    means of two distinct sites.Molecular and Cellular Biology16 6029-6036.

    Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G,

    Umesono K, Blumberg B, Kastner P, Mark M, Chambon P &Evans RE 1995 The nuclear receptor superfamily: the seconddecade. Cell 83 835-839.

    Metzger D, Berry M, Ali S & Chambon P 1995 Effect ofantagonists on DNA-binding properties of the humanestrogen-receptor in vitro and in vivo. Molecular

    Endocrinology 9 579-591.Mosselman S, Polman J & Dijkema R 1996 ER: identification

    and characterization of a novel human estrogen receptor.FEBS Letters 392 49-53.

    Musgrove EA & Sutherland RL 1994 Cell cycle control bysteroid hormones. In: Seminars in Cancer Biology vol 5,pp 381-389. Ed. M Parker. London: Academic Press.

    Nagy L, Kao H-Y, Chakravarti D, Lin RJ, Hassig CA, Ayer DE,

    Schreiber SL & Evans RM 1997 Nuclear receptor repressionmediated by a complex containing SMRT, mSin3A, andhistone deacetylase. Cell 89 373-380.

    ODonnell AL & Koenig RJ 1990 Mutational analysis identifiesa new functional domain of the thyroid hormone receptor.

    Molecular Endocrinology 4 715-720.Ogryzko VV, Schiltz RL, Russanova V, Howard BH & Nakatani

    Y 1996 The transcriptional coactivators p300 and CBP arehistone acetyltransferases. Cell 87 953-959.

    Onate SA, Tsai SY, Tsai M-J & OMalley BW 1995 Sequenceand characterization of a coactivator for the steroid hormonereceptor superfamily. Science 270 1354-1357.

    Parker MG 1993 Steroid and related receptors. Current Opinionin Cell Biology 5 499-504.

    Pennisi E 1996 Expanding the eukaryotes cast of chaperons.Science 274 1613-1614.

    Pfahl M 1993 Nuclear receptor AP-1 interaction.EndocrineReviews 14 651-658.

    Philips A, Chalbos D & Rochefort H 1993 Estradiol increasesand anti-estrogens antagonize the growth factor-inducedactivator protein-1 activity in MCF7 breast cancer cellswithout affecting c-fos and c-jun synthesis.Journal of

    Biological Chemistry 268 14103-14108.Power RF, Mani SK, Codina J, Conneely OM & OMalley BW

    1991 Dopaminergic and ligand-independent activation ofsteroid hormone receptors. Science 254 1636-1639.

    Pratt WB & Toft DO 1997 Steroid receptor interactions with heatshock proteins and immunophilin chaperons.Endocrine

    Reviews18 306-360.Rea D & Parker MG 1996 Effects of an exon-5 variant of theestrogen-receptor in MCF-7 breast-cancer cells. Cancer

    Research56 1556-1563.Reese JC & Katzenellenbogen BS 1991 Differential DNA-

    binding abilities of estrogen receptor occupied with twoclasses of antiestrogens: studies using human estrogenreceptor overexpressed in mammalian cells.Nucleic Acids

    Research19 6595-6602.Renaud J-P, Rochel N, Ruff M, Vivat V, Chambon P, Gronemeyer

    H & Moras D 1995 Crystal structure of the RAR-ligand-binding domain bound to all-trans retinoic acid.Nature 378681-689.

  • 8/6/2019 Steroid Hor Mon

    14/14

    White and Parker: Molecular mechanisms of steroid hormone action

    14

    Roberts AB & Sporn MB 1992 Mechanistic interrelationshipsbetween two superfamilies: the steroid/retinoid receptors andtransforming growth factor-. In: Cancer Surveys, vol 14, pp

    205-220. Ed. MG Parker. Cold Spring Harbor: Cold SpringHarbor Laboratory Press.

    Saatcioglu F, Bartunek P, Deng T, Zenke M & Karin M 1993 Aconserved C-terminal sequence that is deleted in v-ErbA isessential for the biological activities of c-ErbA (the thyroidhormone receptor).Molecular and Cellular Biology 13 3675-3685.

    Saatcioglu F, Claret F-X & Karin M 1994 Negative trans-criptional regulation by nuclear receptors. In: Seminars inCancer Biology, vol 5, pp 347-359. Ed. M Parker. London:Academic Press.

    Saatcioglu F, Lopez G, West BL, Zandi E, Feng W, Lu H, EsmailiA, Apriletti JW, Kushner PJ, Baxter JD & Karin M 1997Mutations in the conserved C-terminal sequence in the thyroid

    hormone receptor dissociate hormone-dependent activationfrom interference with AP-1 activity.Molecular and CellularBiology17 4687-4695.

    Sadovsky Y, Webb P, Lopez G, Baxter JD, Cavailles V, ParkerMG & Kushner PJ 1995 Transcriptional activators differ intheir responses to overexpression of TATA-box-bindingprotein.Molecular and Cellular Biology 15 1554-1563.

    Shemshedini L, Knauthe R, Sassone-Corsi P, Pornon A &Gronemeyer H 1991 Cell-specific inhibitory and stimulatoryeffects of Fos and Jun on transcription activation by nuclearreceptors.EMBO Journal10 3839-3849.

    Shikama N, Lyon J & La Thangue NB 1997 The p300/CBPfamily: integrating signals with transcription factors andchromatin. Trends in Cell Biology 7 230-236.

    Smith CL, Nawaz Z & OMalley BW 1997 Coactivator andcorepressor regulation of the agonist/antagonist activity of themixed antiestrogen, 4-hydroxytamoxifen.Molecular

    Endocrinology 11 657-666.Smith DF & Toft DO 1993 Steroid-receptors and their associated

    proteins.Molecular Endocrinology7 4-11.Soto AM, Justicia H, Wray JW & Sonnenschein C 1991 p-Nonyl-

    phenol: an estrogenic xenobiotic released from modifiedpolystyrene.Environmental Health Perspectives92 167-173.

    Stein B & Yang MX 1995 Repression of the interleukin-6promoter by estrogen receptor is mediated by NF-and C/EBP.Molecular and Cellular Biology 15 4971-4979.

    Tasset D, Tora L, Fromental C, Scheer E & Chambon P 1990Distinct classes of transcriptional activating domains functionby different mechanisms. Cell 62 1177-1187.

    Tora L, White J, Brou C, Tasset D, Webster N, Scheer E &Chambon P 1989 The human estrogen receptor has twoindependent nonacidic transcriptional activation functions.Cell 59 477-487.

    Torchia J, Rose DW, Inostroza J, Kamei Y, Westin S, Glass CK& Rosenfeld MG 1997 The transcriptional co-activator p/CIPbinds CBP and mediates nuclear-receptor function.Nature387 677-684.

    Tzukerman MT, Esty A, Santisomere D, Danielian P, Parker MG,Stein RPJW & McDonnell DP 1994 Human estrogen-receptor

    transactivational capacity is determined by both cellular andpromoter context and mediated by 2 functionally distinctintramolecular regions.Molecular Endocrinology8 21-30.

    Voegel JJ, Heine MJS, Zechel C, Chambon P & Gronemeyer H1996 TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors.

    EMBO Journal15 101-108.

    Wagner RL, Apriletti JW, McGrath ME, West BL, Baxter JD &Fletterick RJ 1995 A structural role for hormone in the thyroidhormone receptor.Nature 378 690-697.

    Webb P, Lopez GN, Uht RM & Kushner PJ 1995 Tamoxifenactivation of the estrogen receptor/AP-1 pathway: potentialorigin for the cell-specific estrogen-like effects ofantiestrogens.Molecular Endocrinology9 443-456.

    Weis KE, Ekena K, Thomas JA, Lazennec G &Katzenellenbogen BS 1996 Constitutively active human

    estrogen-receptors containing amino-acid substitutions fortyrosine-537 in the receptor protein.Molecular

    Endocrinology10 1388-1398.

    White R, Jobling S, Hoare SA, Sumpter JP & Parker MG 1994Environmentally persistent alkylphenolic compounds areestrogenic.Endocrinology 135 175-182.

    White R, Sjberg M, Kalkhoven E & Parker MG 1997 Ligand-independent activation of the oestrogen receptor by mutationof a conserved tyrosine.EMBO Journal16 1427-1435.

    Wurtz J-M, Bourguet W, Renaud J-P, Vivat V, Chambon P, MorasD & Gronemeyer H 1996 A canonical structure for the ligand-binding domain of nuclear receptors.Nature Structural

    Biology 3 87-94.

    Yang NN, Venugopalan M, Hardikar S & Glasebrook A 1996Identification of an estrogen response element activated bymetabolites of 17-estradiol and raloxifene. Science 2731222-1224.

    Yang X-J, Ogryzko VV, Nishikawa J-I, Howard BH & NakataniY 1996 A p300/CBP-associated factor that competes with theadenoviral oncoprotein E1A.Nature 382 319-324.

    Yao T-P, Ku G, Zhou N, Scully R & Livingston DM 1996 Thenuclear hormone receptor coactivator SRC-1 is a specifictarget of p300. Proceedings of the National Academy ofSciences of the USA 93 10626-10631.

    Yeh S & Chang C 1996 Cloning and characterization of a specificcoactivator, ARA70, for the androgen receptor in prostate

    cells. Proceedings of the National Academy of Sciences of theUSA 93 5517-5521.

    Zhang QX, Borg A, Wolf DM, Oesterreich S & Fuqua SA 1997An estrogen receptor mutant with strong hormone-independent activity from a metastatic breast cancer. Cancer

    Research 57 1244-1249.

    Zwijsen RML, Wientjens E, Klompmaker R, van derSman J, Bernards R & Michalides RJAM 1997 CDK-independent activation of estrogen receptor by cyclin D1. Cell88 405-415.