<<

26 8

Endocrine-Related A Jiménez-Panizo et al. Androgen 26:8 R479–R497 Cancer dimerization in disease REVIEW Non-canonical dimerization of the and other nuclear receptors: implications for human disease

Alba Jiménez-Panizo1, Paloma Pérez2, Ana M Rojas3, Pablo Fuentes-Prior4 and Eva Estébanez-Perpiñá1

1Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB) of the University of Barcelona (UB), Barcelona, Spain 2Instituto de Biomedicina de Valencia (IBV)-CSIC, Valencia, Spain 3Computational Biology and Bioinformatics Group, Andalusian Center for (CABD-CSIC), Sevilla, Spain 4Molecular Bases of Disease, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain

Correspondence should be addressed to P Fuentes-Prior or E Estébanez-Perpiñá: [email protected] or [email protected]

Abstract

Nuclear receptors are factors that play critical roles in development, Key Words and in all multicellular organisms. An important family of ff androgen receptor nuclear receptors comprises those members that respond to steroid , and ff receptor which is subdivided in turn into receptor (ER) isoforms α and β (NR3A1 and ff structure A2, respectively), and a second subfamily of so-called oxosteroid receptors. The latter ff -binding domain includes the androgen receptor (AR/NR3C4), the (GR/NR3C1), ff multimerization the receptor (MR/NR3C2) and the receptor (PR/NR3C3). ff Here we review recent advances in our understanding of the structure-and-function ff androgen insensitivity relationship of steroid nuclear receptors and discuss their implications for the etiology syndromes (AIS) of human diseases. We focus in particular on the role played by AR dysregulation in ff resistance both prostate cancer (PCa) and androgen insensitivity syndromes (AIS), but also discuss conditions linked to of the GR as well as those in a non-steroidal receptor, the (TR). Finally, we explore how these recent results might be exploited for the development of novel and selective therapeutic strategies. Endocrine-Related Cancer (2019) 26, R479–R497

Introduction

Nuclear receptors (NRs) form a superfamily of related (PR/NR3C3). Phylogenetic studies transcription factors that play essential roles in show that AR, GR, MR and PR comprise a subfamily of multicellular organisms through coordination of so-called oxosteroid NRs, which markedly differ from both pivotal signaling networks (Evans & Mangelsdorf 2014). ER isoforms (Bledsoe et al. 2002, Evans & Mangelsdorf One important family of NRs groups together those 2014, Gallastegui et al. 2015, Zennaro & Fernandes-Rosa members that respond to steroid hormones, accordingly 2017, Katzenellenbogen et al. 2018). This phylogenetic termed steroid family. In vertebrates, the steroid NRs separation is also reflected at the level of tertiary and include (ER) isoforms ERα (NR3A1) quaternary structures, as we will discuss below. and ERβ (NR3A2) together with the androgen receptor We note that some members of other subfamilies (AR/NR3C4), the glucocorticoid receptor (GR/NR3C1), of NRs specifically respond to steroid hormones, for the mineralocorticoid receptor (MR/NR3C2) and the instance, the vitamin D3 receptor (VDR/NR1I1), the bile

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access

-19-0132 Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R480 Cancer dimerization in disease acid receptor (FXR/NR1H4), oxysterol receptors α and β advanced into late clinical trials (Sadar 2011, Dalal et al. (LXRα/NR1H3 and LXRβ/NR1H2, respectively) and the 2014, Nadal et al. 2017, Veras Ribeiro Filho et al. 2019). -related orphan receptors α, β and γ (RORα/NR1F1, Over the years, several physiologically relevant protein RORβ/NR1F2 and RORγ/NR1F3, respectively). However, partners of NRs have been reported to selectively fine-tune for space reasons, we will focus in the current review their actions in various tissues. These factors, collectively almost exclusively on the members of the sequence and termed coregulators, can either enhance or inhibit structurally related ‘classical’ steroid family of NRs, most transcription of target and are therefore known notably on the AR. as coactivators or , respectively (Hermanson Similar to other NRs, the members of the steroid et al. 2002, Perissi & Rosenfeld 2005, Dasgupta et al. 2014). family display a marked modular structure: a variable These NR coregulators underlie the tissue-selective actions N-terminal region called the N-terminal domain (NTD) of NR ligands and are also emerging therapeutic targets is followed by a highly conserved DNA-binding domain per se, although they have proved hard to target so far (Yi (DBD) comprising two motifs and a C-terminal et al. 2015, Wang et al. 2016, Ruggero et al. 2018). All NR ligand-binding domain (LBD) that contains the internal domains have been involved in coregulator recruitment ligand-binding pocket (LBP) (Jenster et al. 1995, McEwan and macromolecular complex formation, although the 2012a, Gallastegui et al. 2015, Tien & Sadar 2018, Weikum atomic details still remain elusive (Dasgupta et al. 2014, et al. 2018, Fuentes-Prior et al. 2019, Veras Ribeiro Filho Foley & Mitsiades 2016, Giudici et al. 2016, Lempiäinen et al. 2019). DBD and LBD modules are separated by a et al. 2017). poorly conserved interdomain linker called ‘hinge’, which Although the NTDs are variable in length and harbors a nuclear localization signal (NLS) (Fig. 1 for a intrinsically disordered (i.e., isolated NTDs lack stable schematic representation of the domain organization in secondary (2D) and/or tertiary (3D) structures under steroid NRs). All the NR-comprising domains are proposed physiological conditions), the results of different drug targets, but the LBD remains still the main module biochemical and biophysical studies suggest that they for therapeutic intervention and, in spite of intense undergo at least partial folding (‘helicity induction’) efforts, no molecule targeting other receptor domains has to modulate the formation of competent transcription

Figure 1 Schematic representation of the domain organization in steroid receptors. Note the common modular protein structure of four main elements: the N-terminal domain (NTD), which contains the activating function 1 (AF1), the DNA-binding domain (DBD), a hinge region (H) and the actual ligand- binding domain (LBD), which contains the activating function 2 (AF2) assembled by residues from helices H3, H4, H5 and H12. The so-called ‘F domain’, located at the C-terminal end of the receptor, folds back and covers part of the globular LBD in all oxosteroid receptors, but not in ERα/β, whose F domains differ strongly from the one well-conserved in oxosteroid receptors. Other NRs such as the thyroid receptor (TR) lack an F domain altogether. Note also that the NTDs are highly variable in sequence and length. The AR–NTD can be further dissected into two partly overlapping transcription activation units, TAU1 comprising residues 142–485 and TAU5 including residues 360–528 (Jenster et al. 1995, Hilser et al. 2012). Similarly, studies of translational isoforms with variable lengths of the GR-NTD have also found two coupled but thermodynamically distinct regions (Brinkmann et al. 1989, Michigami et al. 1999, Li et al. 2012). At least in the case of these two oxosteroid receptors, the main determinants for map to their highly similar AF-1 sites (46% sequence identity; Kumar et al. 2004, McEwan et al. 2007, De Mol et al. 2018).

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R481 Cancer dimerization in disease activation complexes around the NRs (Kumar et al. 2004, McEwan et al. 2007, De Mol et al. 2016, 2018). This is particularly relevant for a polypeptide stretch termed activation function-1 (AF-1; residues 142-485 in the human AR), which is a hormone-independent transactivation function (Fig. 1). An important result of early investigations on the role of the NTD was the realization of at least transient contacts with the C-terminal LBD moiety. These intra- or intermolecular amino/carboxy terminal (N-C) interactions play key roles in regulating the functions of some steroid receptors, both by stabilizing the overall protein structure and by modulating interactions with DNA and coregulators, thus ultimately controlling (Langley et al. 1998, He & Wilson 2002, Bai et al. 2005). On the LBD, a solvent-exposed pocket responsible for recognizing both AF-1 and coregulators has been extensively studied, which is called activation function 2 (AF-2) or ligand- dependent -binding site (Figs 1 and 2A). At least in the case of the AR, this is in addition to a nearby pocket that contributes to allosterically regulate receptor functions and which has been termed binding function-3 (BF-3) (A note of caution must be added here, as most of the structural and functional evidence comes from studies performed using short derived from known coregulators, which might not correctly reproduce interactions found in multiprotein complexes in vivo.) (Estébanez-Perpiñá et al. 2005, 2007b, Buzón et al. 2012, Ravindranathan et al. 2013, Badders et al. 2018) (Fig. 2B). In the following section, we will briefly discuss recently presented results on the structure-and-function relationship of the AR, but will refer to and compare with Figure 2 Functional surfaces of the androgen receptor. (A) Surface view of the other steroid NRs when appropriate. AR–LBD dimer structure (derived from PDB 5JJM) highlighting the BF3 pocket (pink surface) with a small molecule ((4-(4-hydroxy-3-iodo- phenoxy)-3,5-diiodo-phenyl)-acetic acid) bound, an UBA3-derived bound to the AF2 pocket, and the hormone (dihydrotestosterone, DHT) occupying the ligand-binding pocket (LBP). Note that the AR–LBD core The ‘life cycle’ of the AR: from dimer is closely packed; it buries a quite extensive interface of ≈ 1000 Å2 chaperone-bound monomer to multimers of solvent-exposed surface from each AR–LBD partner. (B) View of the on AR–LBD crystal structure highlighting the four independent AR–LBD molecules found in the asymmetric unit. Notice that the two middle Newly synthesized AR is present in the AR–LBD monomers (colored gray) form a symmetrical core dimer, while the two peripheral molecules (colored light pink) are associated through associated with major molecular chaperones (Prescott & the BF-3 grooves (highlighted in pink). Coetzee 2006, Centenera et al. 2013a, Guy et al. 2015, Foley & Mitsiades 2016, Kita et al. 2017). AR first binds to (Hernández et al. 2002, Azad et al. 2015). Another the 70 (Hsp70)–Hsp40 complex, which chaperone, Hsp27, is regulated by cell stress and prevents is followed by binding of Hsp90 and co-chaperones and aggregation and degradation of the AR, besides promoting subsequent dissociation of Hsp70–Hsp40. Hsp90 binding nuclear trafficking of the receptor and its binding to DNA ensures that the AR retains a high-affinity conformation (Zoubeidi et al. 2007). for the androgenic hormones, and its Binding of androgens in the cytoplasm induces more potent derivative, dihydrotestosterone (DHT) AR dissociation from the chaperone complex, likely

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R482 Cancer dimerization in disease accompanied by significant conformational changes, nuclear Further, it is worth noting that post-translational translocation of the receptor and its dimerization, which modifications (PTMs) profoundly modulate AR actions, is a critical step within this signaling pathway (Centenera both under physiological and pathological conditions et al. 2008, van Royen et al. 2012, Nadal et al. 2017). Pioneer (Faus & Haendler 2006, Gioeli & Paschal 2012, studies suggested that AR oligomerization precedes binding Treviño & Weigel 2013, van der Steen et al. 2013, to chromatin and demonstrated that DNA binding was Koryakina et al. 2014). redox dependent (Kokontis & Liao 1999). More recently, The intrinsically disordered nature of the long NTD AR dimerization has been shown to enhance chromatin and its unique interactions with the LBD (the ‘N/C interaction and remodeling to modulate gene expression (Jin contacts’) raised over the years the issue of whether et al. 2013). However, it must be stressed that the exact cycle FL-AR dimerizes in a parallel, ‘head-to-head’ manner of AR monomer-to-oligomer transition in the cell has not or alternatively in an anti-parallel, ‘head-to-tail’ been precisely elucidated (Schaufele et al. 2005, Centenera configuration Langley( et al. 1995, 1998, Schaufele et al. et al. 2008, van Royen et al. 2012, Nadal et al. 2017). It is 2005, Centenera et al. 2008, Minges et al. 2013). On the usually assumed that this transition follows exactly the other hand, the crystal structure of rat AR–DBD revealed same steps as in related NRs, although data supporting this a head-to-head homodimer bound to its cognate DNA assumption are incomplete or simply lacking. (Shaffer et al. 2004), in an arrangement quite similar to Recent studies of steroid NR trafficking in living cells that adopted by DBD dimers of GR (Luisi et al. 1991) have dramatically advanced our understanding of this and ER (Schwabe et al. 1993). In this conformation, the process, forcing a paradigm shift in the field. In particular, C-terminal residues of both monomers, corresponding careful quantitation of GR, AR and PR dynamics at the to L627 in human AR, are located far away (about 50 Å). single-molecule level have unexpectedly revealed that the However, the length of the G628–E669 linker does not dimeric forms of these receptors are intermediate states allow for an accurate prediction of the likely arrangement toward their biologically active, tetrameric arrangements of LBD monomers in a hypothetical dimer. In fact, (‘dimers of dimers’) (Presman et al. 2016, Paakinaho et al. limited and contradictory information in support of 2017, Presman & Hager 2017, Presman et al. 2017). Further, AR–LBD dimerization had been presented over the years, it has been established that the regulatory complexes with some authors even suggesting that the LBD–LBD assemble around steroid NRs and undergo a rapid exchange interactions may not significantly contribute to receptor in the time scale of seconds (the ‘hit-and-run’ model; Voss oligomerization (Schaufele et al. 2005, Centenera et al. et al. 2011, Sung et al. 2014, Swinstead et al. 2016, Presman 2008, van Royen et al. 2012, Nadal et al. 2017). & Hager 2017). However, detailed information regarding The long-standing questions of whether the the activities of these signaling complexes in normal and AR–LBD domain dimerized and the relative arrangement tumor cells is still missing. The intricate links between NR of monomers have been answered with the resolution of oligomeric state, affinity for ligands (hormones), binding the crystal structure of human AR–LBD bound to DHT cooperativity and recruitment to specific chromatin sites and a peptide derived from the coregulator, ubiquitin- remain to be worked out (Fuentes-Prior et al. 2019). activating 3 (UBA3) (Nadal et al. 2017). All the AR–LBD structures previously solved by us and others had captured essentially the same conformation of Quaternary structure of the AR–LBD: monomeric AR–LBD (Matias et al. 2000, Sack et al. 2001, functional implications He et al. 2004, Estébanez-Perpiñá et al. 2005, Bohl et al. Several AR features have complicated elucidation of the 2007, Estebanez-Perpina et al. 2007). In striking contrast, physiologically relevant tertiary and quaternary structures our recent crystal structure features four independent, of full-length AR (FL-AR) and other steroid receptors, helically arranged LBD molecules (Nadal et al. 2017) and it is still a matter of debate for example whether (Fig. 2B). Two of these LBD monomers are arranged in a the biological units are dimers or ‘dimers of dimers’ (see symmetrical ‘core dimer’, while the other two peripheral above and Presman et al. 2016, Nadal et al. 2017, Presman partners are associated in a less compact manner to the & Hager 2017, Presman et al. 2017). In addition to the BF-3 sites of each of these monomers (Fig. 2B). The core conformational flexibility that is a prerequisite for the AR–LBD dimer exhibits a head-to-head arrangement and functional versatility of the individual AR modules, they displays the corresponding coactivator-binding sites are known to interact with each other to precisely regulate (AF-2 pockets) facing opposite directions, thus able to the NR functions in a context-dependent manner. independently interact with coregulators (Fig. 2A).

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R483 Cancer dimerization in disease

Helices 5 (H5) from both AR–LBD moieties occupy relevance has been questioned upon careful comparison the center of the dimerization interface (Fig. 3A), which is of currently available GR structures, and alternative GR thus topologically distinct from the ‘canonical’, H10–11- dimeric conformations have been suggested (Bianchetti centered arrangements identified, among others, in ER et al. 2018). Clearly, further experimental work is needed to homodimers and in RXR-mediated heterodimers (Fig. 3C) fully clarify the physiologically relevant conformation(s) (Wurtz et al. 1996, Brzozowski et al. 1997, Bourguet of the GR and other members of the oxosteroid subfamily et al. 2000, Chandra et al. 2008, 2017, Khorasanizadeh to integrate structural, biochemical and cellular studies & Rastinejad 2016, Nadal et al. 2017, Fuentes-Prior et al. (Fig. 3) (Bledsoe et al. 2002, Kauppi et al. 2003, Robertson 2019). The H5-centered dimerization mode of human et al. 2013a,b, Presman et al. 2016, 2017, Paakinaho AR–LBD has been confirmed in solution and in cells Nadal( et al. 2017, Presman & Hager 2017, Weikum et al. 2017, et al. 2017). This non-canonical dimeric conformation Wilkinson et al. 2018). might be shared by other NRs, not only of the oxosteroid Although specific functions have been commonly class (Fuentes-Prior et al. 2019). In fact, a topologically ascribed to the different NR domains (e.g., DNA and equivalent dimer of GR–LBD had been previously reported hormone binding), seminal X-ray crystallography studies (Bledsoe et al. 2002) (Fig. 3B), although its biological of non-steroidal NRs indicate that DBD, hinge and LBD

Figure 3 Crystal structures of homodimeric ligand-binding domains from various nuclear receptors. (A) Overall structure of the AR–LBD core dimer (PDB 5JJM). The two monomers are depicted as cartoons showing helices in blue and loops in pink; helices H5, H9, H10-11 and H12 are marked. Note that the AR–LBD mainly dimerizes through helices H5 of both monomers. The bound hormone (dihydrotestosterone, DHT) and the UBA3 peptide are shown as pink spheres and as a magenta cartoon, respectively. (B) Overall structure of the first reported GR-LBD dimer (PDB 1M2Z). The two monomers are depicted as cartoons showing helices in blue and loops in pink; helices H5, H9 and H10-11 are highlighted. The bound hormone (, DEX) and the coactivator peptide are shown as pink spheres and as a pink cartoon, respectively. Note the topological equivalence to the AR–LBD dimer depicted in panel A, in spite of less intimate contacts at the intermonomer interface. (C) Overall structure of the ER–LBD dimer (PDB 1ERE). The two monomers are depicted as cartoons showing helices in pink and loops in red; helices H9 and H10–11 are indicated. Note that the ER–LBD dimerizes according to the ‘canonical’ mechanism, that is through helices H10–11 of each monomer. The hormone (17beta-, EST) is shown as blue spheres. (D) Overall structure of the TR–LBD dimer (PDB 2PIN). The two monomers are shown as cartoons with helices in green and loops in pink; helices H9, H10–11 and H12 are labeled. The ligand ((4-(4-hydroxy-3-iodo-phenoxy)-3,5-diiodo-phenyl)-acetic acid, 4HY) is shown as pink spheres. Dashed lines indicate parts of the structure that were not visible in the electron density maps.

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R484 Cancer dimerization in disease are more intricately interconnected than initially thought, both structurally and functionally (Chandra et al. 2008, 2013, 2017, Lou et al. 2014, reviewed in Rastinejad et al. 2015, Fuentes-Prior et al. 2019). Regarding the AR and other steroid receptors, current knowledge of their interdomain interactions derives mostly from biochemical and mutagenesis studies. A recent analysis of multidomain ER by small-angle X-ray scattering (SAXS), however, suggests an important cross-talk between DBD and LBD moieties (Huang et al. 2018). These findings, at the light of the results of Rastinejad and coworkers with non-steroidal NRs, point to important DBD-linker-LBD interactions in vivo (Chandra et al. 2008, 2013, 2017, reviewed in Fuentes-Prior et al. 2019). This is in addition to the well-known intra- or intermolecular N–C interactions (see above), which restrict the possible DBD/LBD relative orientations (Fig. 4B).

Allosteric modulation of AR activity

NRs are allosteric par excellence (Kuriyan 2004, del Sol et al. 2006, McEwan 2012b, Mackinnon et al. 2014), and the AR is no exception in this regard (Estebanez- Perpina et al. 2007, Buzón et al. 2012, Grosdidier et al. 2012). Indeed, FL–AR functions as an allosteric switch alternating between inactive, chaperone-bound/ligand- free states and active, hormone- and coactivator-bound conformations. Ligand binding and the exchange of chaperones by coactivators are allosterically coupled, but the sequence of molecular events and detailed conformational changes associated are only partially understood (Hur et al. 2004, Estébanez-Perpiñá et al. 2005). In particular, hormone binding to the LBP has been shown to trigger allosteric remodeling of the AF-2 and BF-3 interaction surfaces. In this manner, occupancy of the LBP by ligands regulates the dynamics and stability of surfaces Figure 4 that recognize coregulators and is thus allosterically Proximity of functionally relevant areas in non-canonical dimers. (A) coupled to the recruitment of these proteins to the AR Close-up of the AR–LBD dimerization interface. Monomers are colored light pink and blue, respectively, and the DHT molecules are shown as (Estébanez-Perpiñá et al. 2005, Estebanez-Perpina et al. color-coded Van der Waals spheres. Residues are shown as color-coded 2007). Albeit a recent proteomics study has identified the sticks (oxygen, red; nitrogen, blue; carbon, yellow or brown) and labeled. essentially overlapping, -specific interactomes of Note that most residues making important contributions to this interface possess hydrophobic/aromatic side chains (e.g., V685, W752, F755, V758, both AR and GR (Lempiäinen et al. 2017), the detailed Y764 and P802). The location of residue R753 is also noteworthy, as its molecular determinants of NR binding to a large number side chain contacts the DHT molecule bound inside the internal of coregulators are far from being well understood. ligand-binding pocket (LBP) from its ‘own’ monomer, while its main chain atoms form part of the dimer interface, and in particular, interact with Noteworthy, in addition to allosteric rearrangements N757 from the second monomer. In this manner, hormone binding and elicited by hormone binding, the target DNA sequences receptor dimerization appear to be intimately coupled in the AR. (B) to which NRs bind also induce important remodeling Close-up of the AR–LBD surface (as seen in PDB 1XOW), highlighting the proximity of the AF-2 pocket (colored blue; the bound NH2-terminal of the receptor structure (Fuentes-Prior et al. 2019). peptide of the receptor is colored cyan) to the LBP (with bound Most impressively, cognate DNA-binding sequences of methyltrienolone/R1881 colored magenta).

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R485 Cancer dimerization in disease the GR have been shown to function as true allosteric metabolism, it is not surprising that dysregulation of modulators, which are capable of affecting conformation their activities is directly responsible for a number of and regulate receptor activity (Meijsing et al. 2009, Love important human conditions (Huang et al. 2010, Evans et al. 2017, Weikum et al. 2017, Frank et al. 2018). Thus, & Mangelsdorf 2014, Luo et al. 2018). For instance, a change in a single base pair in the GR-binding site the ER is a key driver of 70% of breast cancer subtypes resulted in up to 10-fold higher affinity in the activation that require estrogen hormones for progression, and of a glucocorticoid response element (GRE) reporter gene different point mutations in the ER genes are linked in response to ligand. More recently, it has been reported to acquired resistance to commonly used estrogen- that nucleotides directly flanking the core-binding site not blocking drugs such as (Kojetin et al. 2008, only modulate the 3D structure of this site, but also that Katzenellenbogen et al. 2018, Nasrazadani et al. 2018, of the GR–DBD and even the quaternary conformation Reinert et al. 2018). Regarding oxosteroid receptors, of the dimeric receptor (Schöne et al. 2016). This implies single-residue mutations in GR and MR genes have that GR activity can be modulated by both sequence mainly been associated to loss-of-function phenotypes composition and DNA shape to achieve fine-tuning of (Zennaro & Fernandes-Rosa 2017). However, the most the GR structure and activity downstream of binding. conspicuous association between a These features are likely to be shared by other members and human disease links the AR to several biomedical of the oxosteroid class of NRs, at the light of the almost conditions, as we briefly discuss below. identical sequences of DBDs and relative conservation of interdomain hinges. This flanking effect could explain Genetic bases of drug resistance in prostate cancer how NRs predicted to recognize similar binding motifs show distinct DNA-binding preferences in vivo. It is AR is encoded by a ubiquitously expressed gene located in feasible that while two related NRs can bind to the same the X chromosome at Xq11-12 and is particularly important DNA motif, the competent conformation required for an in prostate development and homeostasis (Lubahn et al. optimal transcriptional response is only achieved through 1988). When dysregulated, however, AR activity is central specific flanking sequences. In particular, we assume to the onset, development and progression to metastasis that AR–DBD binding to its cognate DNA sequences will of prostate cancer (PCa), the most common cancer trigger intra- and interdomain conformational changes diagnosed in males worldwide (Matias et al. 2000, Gottlieb that would affect the overall quaternary structure of the et al. 2004, Knudsen & Penning 2010, Arora & Barbieri receptor, including its hormone-binding domain. 2018, Centenera et al. 2018, Cioni et al. 2018, Li et al. 2018, The precise molecular mechanisms underlying Luo et al. 2018, Nevedomskaya et al. 2018, Paschalis et al. allosteric transitions in FL–AR, including the equilibrium 2018). In addition, AR mutations are linked to disorders between different conformational states and the impact of male sexual differentiation and development termed of ligand binding to allosteric modulatory sites (e.g., androgen insensitivity syndromes (AIS) (Hughes et al. AF-2, BF-3) on receptor oligomerization, remain to be 2012, Mongan et al. 2015, Gibson et al. 2018) and to the worked out (Fig. 2A) (Nadal et al. 2017). In multidomain rare adult-onset hereditary neurodegenerative disorder AR, both intra- and interdomain allostery may occur known as spinal and bulbar muscular atrophy (SBMA or simultaneously (Fernandez et al. 2017, Fernandez 2018). Kennedy’s disease; OMIM #313200) (Spada et al. 1991, The first may take place within the NTD, DBD or LBD Badders et al. 2018, Cortes & La Spada 2018, Lieberman moieties, while interdomain appears 2018, Pennuto & Rinaldi 2018). Finally, AR malfunction to be essential in coordinating AR functions (e.g., through is also associated to androgenic alopecia or loss of scalp the well-studied N-C contacts, but also upon as of yet hair and skin malignancies (Ellis et al. 2001, Clocchiatti uncharacterized DBD–LBD and LBD–DNA interactions). et al. 2018). Similar considerations might apply to other steroid NRs. Several altered genetic and epigenetic mechanisms contribute to the development of metastatic and drug- resistant PCa (Knudsen & Penning 2010, Shen & Abate- Shen 2010, Grasso et al. 2012, Robinson et al. 2015, Dysregulation of steroid receptors and Pritchard et al. 2016, Baumgart & Haendler 2017, human disease Armenia et al. 2018, Cotter & Rubin 2018, Li et al. 2018, Given the essential roles of steroid hormones and Quigley et al. 2018, Ruggero et al. 2018, Karthaus & their cognate NRs in development, homeostasis and Sawyers 2019). For example, tandem duplications of an

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R486 Cancer dimerization in disease upstream enhancer of the AR are found in up to 87% encoding truncated AR isoforms. The identification of of metastatic castration-resistant PCa (mCRPC) cases, these constitutively active AR variants (AR-Vs), which lack compared to <2% of primary prostate cancers (Takeda portions or the entire hormone-sensitive LBD, has added et al. 2018, Viswanathan et al. 2018, Wu et al. 2018). In an unanticipated level of complexity to the AR signaling addition, it has been recently reported that deletion of pathways (Dehm & Tindall 2011, Centenera et al. 2013b, the gene encoding the chromatin remodeler, chromatin Ho & Dehm 2017). Many of these truncated AR-Vs seem helicase DNA-binding protein (CHD1), redistributes the to support androgen-independent expression of AR-target AR cistrome in a manner that favors the expression of genes and therefore androgen-independent growth of PCa-specific oncogenic pathways (Augello et al. 2019). PCa cells (Dehm & Tindall 2011). However, the androgen Not surprisingly, CHD1 loss had been previously reported independency of AR-Vs does not imply that expressing as one of the most common and deleterious genetic cells are totally independent of the presence of androgenic alterations in PCa (Grasso et al. 2012, Huang et al. 2012, hormones. The (patho)physiological implications of the Rodrigues et al. 2015, Zhao et al. 2017) and an early event coexistence of full-length AR with one or several AR-Vs in cancer development (Wedge et al. 2018). in the same cell are still a matter of intense debate. The Further, AR gene duplications and AR-mediated likely formation of AR-Vs (hetero)dimers with FL-AR, chromosomal rearrangements are common events. either through DBD-mediated or N–C interactions may Among them, about half of the patients with PCa feature still result in hormone-dependent cell growth leading to fusions of the AR-regulated gene, TMPRSS2, with different PCa progression (Liu et al. 2014, Uo et al. 2017). Further, fragments of the ETS-related gene (ERG), a member of potential differences in the interactomes of the different the ETS family of transcription factors. The presence of AR-Vs in comparison with that of the full-length receptor these TMPRSS2:ERG fusions defines the predominant have not been explored so far. molecular subtype of PCa (Yu et al. 2010, Park et al. 2014, Reig et al. 2016, Stelloo et al. 2018, Berglund et al. The AR–LBD dimerization interface is a hot spot of 2019). The encoded ERG truncated variants are resistant disease-linked mutations to degradation and transform the AR from a factor promoting lineage-specific differentiation of the prostate Inspection of the 3D structure of the AR–LBD homodimer to one that potentiates de-differentiation into a stem cell- immediately revealed that a large number of hitherto like state (Yu et al. 2010). However, TMPRSS2:ERG fusions unexplained mutations of solvent-exposed residues alone do not appear to be transforming (Casey et al. 2012), identified in PCa (Fig. 5A) and AIS patients cluster at and the precise mechanism(s) by which they contribute the dimerization interface (Nadal et al. 2017; see also to PCa initiation and/or progression are still a matter Fig. 5B). We stress that these mutations might have of debate. For instance, it has been reported that these additional consequences for AR protein structure and fusions synergize with deletion of the tumor suppressor, function, such as impaired interaction with chaperones PTEN, to promote prostatic intraepithelial neoplasia (PIN) in the monomeric state (see above). Accordingly, the (Carver et al. 2009, King et al. 2009, Casey et al. 2012). In situation in vivo does not fit a simplistic dichotomy of this regard, and also connecting to the work by Augello gain-of-function mutations causing PCa, while loss-of- and coworkers cited above, it has been proposed that function variants of the AR gene are linked to AIS (Hay & ETS factors alter the AR cistrome to prime the prostate McEwan 2012). epithelium to respond to aberrant signals such as Nevertheless, there are several correlations between PTEN loss, thus ultimately resulting in prostate-specific for example the nature of the mutant residue and disease transformation (Chen et al. 2013). Carver and coworkers severity, which strongly point to impaired homodimer also observed that two genes strongly associated with formation as a more likely cause of the observed cell migration, ADAMTS1 and CXCR4, were upregulated phenotype. This is in particular the case of repeatedly upon ERG overexpression (Carver et al. 2009). Besides, identified mutations that affect residues such as F755, overexpression of MMP9 and plexin B correlates with the N757, V758, N759, R761 and P767 (Figs 4A and 5A, C). presence of the fusion in samples of PCa patients and has Most notably, the mutant Y764C has been found both been linked to migration and invasion of prostate cancer in PCa and AIS. Furthermore, residue F755 has been cells (Liu et al. 2017). found conservatively replaced by either leucine, in some On the other hand, several studies have described patients with partial AIS, or by another aliphatic residue, the synthesis in vivo of alternatively spliced transcripts valine, in cases of complete androgen insensitivity.

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R487 Cancer dimerization in disease

Figure 5 Mutations that affect the non-canonical, AR-like dimerization interface are linked to various conditions. The LBD dimers of all four analyzed receptors are represented as gray cartoons. (A and B) AR–LBD (PDB 5JJM) with the side chains of all mutated interface residues shown with all their non-hydrogen atoms as sticks and surface, colored red for mutations reported in PCa patients (A) or blue for those linked to AIS (B). Mutations have been retrieved from the Androgen Receptor Gene Mutations Database (http://androgendb.mcgill.ca/). (C) GR-LBD (PDB 1M2Z) with the side chains of all mutated interface residues shown with all their non-hydrogen atoms as sticks and surface, colored pink to highlight those residues linked to glucocorticoid hormone resistance. Some of these residues have been associated with primary generalized Chrousos syndrome. (D) Cartoon representation of the TR-LBD dimer (gray, PDB 2PIN) with the side chains of all mutated interface residues shown with all their non-hydrogen atoms as sticks and surface, colored green to highlight those residues associated with thyroid hormone resistance.

The side chain of F755 makes important Van der Waals result in impaired tissue-specific sensitivity to GCs, which interactions with P802 from the second monomer may manifest as GC resistance or hypersensitivity, both of (Fig. 4A), and modeling experiments indicate that a which are associated with significant morbidity (Nicolaides leucine residue at position 755 might still interact with & Charmandari 2015, Nicolaides et al. 2015a,b, Wilkinson this proline, supporting homodimer formation. However, et al. 2018). a less bulky valine would not be able to contact P802, with For instance, in primary generalized GC resistance concomitantly impaired dimerization. A second example (PGGR or Chrousos syndrome, MIM #615962), NR3C1 of strong genotype–phenotype correlation is given by heterozygous variants (point mutations, insertions or mutant p.V758I identified in PCa patients. Again, a fully deletions) result in impaired GR function and thus conservative replacement of an aliphatic residue leading decreased sensitivity to GCs in all organs, although to PCa most likely reflects the higher stability of the with a high degree of variability among different tissues. homodimer formed by mutant AR–LBD molecules, as Alterations in the GR function include dysregulation of the bulkier I758 side chain fills better the intermonomer the GC-mediated negative feedback mechanisms, which space. These findings, among others, lend extraordinary results in compensatory activation of the hypothalamic– support to the (patho)physiological relevance of the AR pituitary–adrenal (HPA) axis. In turn, HPA overactivation homodimer interface and suggest that this area might be leads to hypersecretion of ACTH and (highly targeted for pharmacological intervention (see below). variable, ranging from severe symptoms to subclinical hypercortisolism) and may result in increased production of other adrenal steroids such as androgens and NR3C1/GR mutations linked to either glucocorticoid . Patients affected by this syndrome resistance or hypersensitivity syndromes are thus commonly diagnosed by clinical signs of (GCs) are major regulators of many mineralocorticoid and/or androgen excess (hypertension physiological functions, and thus contribute substantially due to hyperaldosteronism and/or hyperandrogenic to tissue homeostasis. Dysfunction of GC-mediated signs), rather than those associated with GC deficiency actions underlies two human pathologic conditions, (Kino & Chrousos 2001, Kino et al. 2002, Nicolaides et al. Cushing syndrome and Addison’s disease, which are due 2010, 2015a,b, Charmandari et al. 2013, Nicolaides & to GC excess or deficiency, respectively. On the other Charmandari 2015, Wilkinson et al. 2018). hand, alterations in GR function (either due to mutations So far, 26 mutations have been described in the human or polymorphisms of the encoding gene, NR3C1, or to NR3C1 gene, most of which are missense mutations other molecular changes along the GR signaling pathway) and affect the GR–LBD (Vitellius et al. 2016, 2018,

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R488 Cancer dimerization in disease

Nicolaides & Charmandari 2017) (Fig. 5C). Functional LBP (p.S810L) has been associated with a severe form of studies indicate that most NR3C1 mutations impair several inherited hypertension (Kino & Chrousos 2001, Zennaro steps of GC signaling including hormone recognition, GR & Fernandes-Rosa 2017). Finally, PR mutations are less nuclear translocation, DNA binding and interactions with frequent but they have been associated to an increased the coactivator, GRIP1, which ultimately leads to reduced risk of breast, endometrial and colon cancer (Agoulnik or absent GR-dependent transactivation. Structural et al. 2004). studies have contributed to a better understanding of how conformational changes in the receptor cause GC Impact of mutations affecting the non-canonical resistance, although a systematic analysis of all reported dimerization interface in other non-steroidal NRs mutants is lacking. Computer simulations suggest that the primary cause of GC resistance in Chrousos syndrome We finish this section on the linkage between dysregulation is an overall destabilization of the LBD, as the result of of NR activities and pathologic conditions by presenting single replacements that affect LBP, AF-2 or the case of a non-steroidal NR that has also been shown both (Hurt et al. 2016). The possible role of impaired to dimerize in an AR-like, non-canonical conformation, receptor multimerization on this syndrome, however, has the thyroid receptor (TR; Estébanez-Perpiñá et al. 2007a, not been explored so far. Jouravel et al. 2009 and Fig. 5D). Vertebrates possess actually In addition, it has been recently reported that NR3C1 two TR isoforms, termed TRα (NR1A1) and TRβ (NR1A2), mutations also cause another pathological condition and which are encoded by two independent genes, THRA called incidentaloma (incidentally discovered bilateral and THRB, respectively. In spite of their close structural adrenal hyperplasia), with clinical features similar to similarity (70% sequence identity), these two isoforms differ those of Chrousos syndrome such as asymptomatic strongly both in expression profile. TRα is ubiquitously hypercortisolism and/or hypertension. Five novel expressed, while TRβ is mainly expressed in liver, pituitary, heterozygous NR3C1 mutations that cause impaired GR inner ear and some brain areas (Chatonnet et al. 2013), but signaling were identified, which represents a relatively in particular because they regulate different sets of genes. high prevalence of 5% in the analyzed cohort. These TRs respond to iodinated biomolecules known as thyroid findings suggest that the overall prevalence of NR3C1 hormones (THs), mostly to the active form 3,3′,5-triiodo- mutations may have been previously underestimated l-thyronine (T3), which is generated in turn from the and advise to perform GR screening in patients prohormone, l-thyroxine (T4) (Holzer et al. 2017). with adrenal incidentalomas (Vitellius et al. 2016, 2018). Since THs have fundamental functions in Although most of the identified NR3C1 mutations mapped development, growth and metabolic homeostasis, at the GR–LBD, other (p.R477S, p.R477C, p.R477H and alterations in TR structure affect these functions at many p.Y478C) were located at the C-terminal end of the second different levels. This is in particular the case of generalized zinc finger, implicated in the dimerization of the receptor. thyroid hormone resistance (GTHR), a syndrome By contrast, in the rare syndrome known as primary characterized by elevated serum TH but impaired action generalized glucocorticoid hypersensitivity (PGGH), of these hormones within the hypotalamic–pituitary– patients exhibit clinical signs of metabolic syndrome thyroid axis and variable tissue hyposensitivity to them without hypercortisolism, due to compensatory (Huber et al. 2003a,b, Onigata & Szinnai 2014). GTHR hypoactivation of the HPA axis (Nicolaides & Charmandari is linked to mutations located in the LBD of TRβ (Weiss 2017). This disease mostly correlates with activating NR3C1 et al. 1993, Adams et al. 1994). Similar to the results polymorphisms while to date, a single patient harboring discussed above for GR, most of these mutations result a point mutation in the NTD (p.D401H) exhibited clinical in a lower affinity for T3 or a defective interaction symptoms of GC hypersensitivity including obesity, type with TR coregulators. Whether are least some of these 2 diabetes and hypertension (Charmandari et al. 2008). variants would impair dimer formation has not been Further, loss-of-function mutations in the NR3C2/MR experimentally confirmed. However, inspection of the gene are responsible for renal pseudohypoaldosteronism 3D structure of the TRβ homodimer (Estébanez-Perpiñá type 1 (MIM #177735), a rare disease of mineralocorticoid et al. 2007a, Jouravel et al. 2009) immediately reveals resistance characterized by sodium and potassium that some of the reported missense mutations actually imbalances that manifests at birth with weight loss and map to the homodimer interface and are therefore dehydration despite elevated plasma levels of . likely to disrupt receptor homodimerization (Fig. 5D). Conversely, an activating MR mutation that reshapes its Particularly interesting mutations in this regard affect

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R489 Cancer dimerization in disease

Table 1 Mutations within the non-canonical dimerization interface are linked to various human conditions.

Androgen receptor Glucocorticoid receptor Estrogen receptor Thyroid receptor Residue Disease Residue Disease Residue Disease Residue Disease Q671R PCa I673T PCa V685I CAIS V543 I326 A234T THR C687R PAIS Y545 Y328 R243Q THR R243W D691E PAIS D549 X K244 D691V CAIS D696H CAIS D554 F337 I250 THR D696N CAIS/PAIS/M I250T D696V CAIS D696Y CAIS A749T PCa A749D PAIS

W752R PCa W610 W393 V319 R753P CAIS R611L GCR R394H Estrogen R320C THR R753Q CAIS insensitivity R320H F755L PCa Y613 M396 D322H THR F755S PAIS/MAIS F755V CAIS T756A PCa R614 E397 L328S THR N757D PCa Q615 X E324 N757S PAIS/MAIS S760F CAIS A618 H398 P323 S760P PCa S760Y CAIS R761? PCa N619 G400 P323/D324 R761S PAIS M762T PAIS L620 K401 E326 L763F CAIS L621 L402 T327P THR T327A Y764C PCa/PAIS C622 L403 T329N THR Y764H CAIS T329I F765L CAIS F623 F404 L330S THR P767A CAIS P625 P406 G332R THR P767S CAIS G332E D768E CAIS N626 N407 E333D THR D768Y CAIS E773 E631 R412 R338W THR R338L THR W797del CAIS H655 R437 S362 Q799E PCa/PAIS/M Q657 N439 N364 Q803R PAIS E661 E443 T368 R847G PCa E705 G494 X N849K CAIS T496 H210 R856C CAIS R714Q Chrousos R503W BCa syndrome R856H CAIS/PAIS/M

Residues from the non-canonical dimerization interface of the AR–LBD were aligned to those of the corresponding domains in GR, ER and TR. Missense mutations that affect these topologically equivalent residues are given, along with the associated disease. Note in particular that several mutations affect topologically equivalent residues in AR–LBD and TR–LBD. Note also that replacement of the highly conserved residue R753 of AR–LBD, which corresponds to R611 of GR–LBD, R394 of ER–LBD and R320 of TR–LBD, causes hormone resistance in all receptors. GCR, glucocorticoid resistance; THR, thyroid hormone resistance. arginine residues at positions 320 (p.R320C, p.R320H) GC resistance (p.R611L in GR) or estrogen insensitivity and 338 (p.R338W, p.R338L) in TRβ, as mutations of (p.R394H in ER) (Table 1) (Weiss et al. 1993). the topologically equivalent residues in several steroid In summary, residues responsible for non-canonical receptors result in either AIS (p.R753P, p.R753Q in AR), homodimer formation in NRs play a critical functional

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R490 Cancer dimerization in disease role, leading to various pathological conditions when (so-called ‘LBP-focused strategies’). However, the recent dimerization is disrupted. A list of mutations that are advances in the structure and function of NRs discussed likely to interfere with non-canonical homodimer above have largely expanded the space for pharmacological formation of some NRs and their associated conditions intervention. First, we mention that AR chaperones had is given in Table 1. been proposed as potential therapeutic targets against PCa and, after initial failures, heat shock proteins, and in particular Hsp90, are currently considered viable targets to treat PCa (Eskew et al. 2011, Centenera et al. 2012, 2013a, Implications for the development of novel He et al. 2013). Indeed, inhibitors of Hsp90 C-terminal therapeutic strategies domain have been recently shown to inhibit growth of All steroid NRs are major therapeutic targets to treat various PCa cell lines without inducing the expression of several endocrine-related diseases (Moore et al. 2006, other chaperones (Armstrong et al. 2016). Evans & Mangelsdorf 2014, Carroll 2016, Nasrazadani Novel strategies that directly target the NR include et al. 2018). Most notably, the dependence of PCa tumors blocking receptor interactions with specific coactivators on the AR protein and its cognate endogenous hormones (Azad et al. 2015, Guy et al. 2015, Foley & Mitsiades 2016), is the basis for its pharmacological exploitation as a drug directly targeting NR binding to cognate DNA sequences target. Indeed, inhibition of AR functions by means or interfering with DBD dimerization (Dalal et al. 2017), of ligand depletion and/or the use of AR antagonists but also modulating ligand-independent functions () is the first line of therapeutic intervention (reviewed in Caboni & Lloyd 2013). Also along these against PCa (Chen et al. 2008, Mohler et al. 2012, lines, the AR–NTD has been explored as drug target for Lorente et al. 2015, Aggarwal et al. 2017, Ponnusamy AR function control, in particular for management of its et al. 2017, Narayanan et al. 2018, Nevedomskaya et al. ligand-independent truncated variants (De Mol et al. 2016, 2018). However, recurrent resistant and incurable tumors Imamura & Sadar 2016, Kumar et al. 2016, Monaghan & arise as a result of inappropriately restored AR function McEwan 2016). associated with various genetic and epigenetic accidents. It is believed that conformational changes elicited In particular, emergence of point mutations as a response upon ligand binding are ultimately responsible for to therapy is a quite common event, which the different activity profiles exhibited by therapeutic may result in antiandrogens acting as rather than compounds in different cell types, similar to what has been antagonists with severe clinical consequences (Knudsen shown for other NRs (Srinivasan et al. 2013, Nwachukwu & Penning 2010, Balbas et al. 2013, Joseph et al. 2013, et al. 2016). Given that these signals might propagate Schrecengost & Knudsen 2013, Lorente et al. 2015, Watson through the whole multidomain protein, controlling et al. 2015, Jernberg et al. 2017, Giacinti et al. 2018). The NR functions by targeting novel unexplored sites is an identification of constitutively active, truncated splice AR attractive but challenging alternative. Most importantly, variants (AR-Vs) lacking parts or the entire LBD also poses apart from the classical modulators targeting the LBP important pharmacological challenges (Dehm & Tindall site, synthetic allosteric modulators are currently under 2011, Centenera et al. 2013b, Ho & Dehm 2017, Paschalis investigation, thus substantially broadening the chemical et al. 2018). This is in addition to the cross-reactivity of space for novel antiandrogens (Buzón et al. 2012). In this therapeutic steroidal androgens with other, highly related manner, the AR LBP shall no longer be seen as the only steroid NRs (i.e., GR) in PCa patients, which derives in pocket suitable for pharmacological intervention, but all unwanted side effects that pose additional limits to their NR domains and their protein–protein interaction sites are clinical use (Arora et al. 2013, Karamouzis et al. 2016, envisioned as potential drug targets (Estébanez-Perpiñá Narayanan et al. 2016). Altogether, there is an unmet et al. 2007b, Buzón et al. 2012, Nadal et al. 2017, Badders need for the development of novel therapeutic strategies et al. 2018). In this regard, we recall that both AF-2 and including tissue-selective AR modulators (SARMs; BF-3 sites are druggable and are ideal candidates for off- Estébanez-Perpiñá et al. 2007b, Dalton 2017, Narayanan LBP strategies (Estébanez-Perpiñá et al. 2007b, Buzón et al. 2018) that may overcome the problems encountered et al. 2012, Ravindranathan et al. 2013, Badders et al. by currently used drugs. 2018). Most notably, AF2-targeting compounds have been Most therapeutic strategies to date have focused on recently validated as potent androgen-sparing strategies the development of small-molecule compounds that for SBMA therapy (Badders et al. 2018). Last but not least, compete with receptor binding to natural hormones interfering with the non-canonical dimerization interfaces

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R491 Cancer dimerization in disease of the AR and other oxosteroid receptors (Nadal et al. alters AR binding at lineage-specific enhancers and modulates 2017, Fuentes-Prior et al. 2019) offers hitherto unforeseen distinct transcriptional programs to drive prostate tumorigenesis. Cancer Cell 35 603.e8–617.e8. possibilities for the regulation of NR activities, and thus, Azad AA, Zoubeidi A, Gleave ME & Chi KN 2015 Targeting heat shock for the development of novel pharmaceutical drugs. proteins in metastatic castration-resistant prostate cancer. Nature Reviews: Urology 12 26–36. (https://doi.org/10.1038/ nrurol.2014.320) Badders NM, Korff A, Miranda HC, Vuppala PK, Smith RB, Winborn BJ, Declaration of interest Quemin ER, Sopher BL, Dearman J, Messing J, et al. 2018 Selective The authors declare that there is no conflict of interest that could be modulation of the androgen receptor AF2 domain rescues perceived as prejudicing the impartiality of this review. degeneration in spinal bulbar muscular atrophy. Nature Medicine 24 427–437. (https://doi.org/10.1038/nm.4500) Bai S, He B & Wilson EM 2005 Melanoma antigen gene protein MAGE- 11 regulates androgen receptor function by modulating the Funding interdomain interaction. Molecular and Cellular Biology 25 E E P expresses her deepest gratitude to the generous and continuous 1238–1257. (https://doi.org/10.1128/MCB.25.4.1238-1257.2005) private donations from Gemma E Carretero. E E P, P F P and P P acknowledge Balbas MD, Evans MJ, Hosfield DJ, Wongvipat J, Arora VK, Watson PA, the RED Nacional de Receptores Nucleares (NurCaMeIn: SAF2017-71878- Chen Y, Greene GL, Shen Y & Sawyers CL 2013 Overcoming REDT and SAF2015-71878-REDT). E E P acknowledges the BFU-Retos2017- mutation-based resistance to antiandrogens with rational drug 86906-R, P F P the SAF2014-57994-R, P P the SAF2017-88046-R and A R design. eLife 2 e00499. (https://doi.org/10.7554/eLife.00499) the RTI2018-096735-B-100 grants from the MINECO. E E P and P F P are Baumgart SJ & Haendler B 2017 Exploiting epigenetic alterations in thankful for the CaixaImpulse2017 (ARDIs) from La Caixa (Fundación prostate cancer. International Journal of Molecular Sciences 18 Bancaria Caja de Ahorros y Pensiones de Barcelona, Obra Social La Caixa y 1017–1017. (https://doi.org/10.3390/ijms18051017) Caixa Risc) and SGR2017 and LLAVOR2018 grants from the Generalitat de Berglund AE, Rounbehler RJ, Gerke T, Awasthi S, Cheng CH, Takhar M, Catalunya. E E P acknowledges the F2I-FVAL and F2I-MiR grants from the Davicioni E, Alshalalfa M, Erho N, Klein EA, et al. 2019 Distinct EU FEDER and the Fundació Bosch i Gimpera (FBG). transcriptional repertoire of the androgen receptor in ETS fusion- negative prostate cancer. Prostate Cancer and Prostatic Diseases 22 292–302. (https://doi.org/10.1038/s41391-018-0103-4) Bianchetti L, Wassmer B, Defosset A, Smertina A, Tiberti ML, Stote RH & Dejaegere A 2018 Alternative dimerization interfaces in the References glucocorticoid receptor-α ligand binding domain. Biochimica et Biophysica Acta. General Subjects 1862 1810–1825. (https://doi. Adams M, Matthews C, Collingwood TN, Tone Y, Beck-Peccoz P & org/10.1016/j.bbagen.2018.04.022) Chatterjee KK 1994 Genetic analysis of 29 kindreds with generalized Bledsoe RK, Montana VG, Stanley TB, Delves CJ, Apolito CJ, McKee DD, and pituitary resistance to thyroid hormone. Identification of Consler TG, Parks DJ, Stewart EL, Willson TM, et al. 2002 Crystal thirteen novel mutations in the beta structure of the glucocorticoid receptor ligand binding domain gene. Journal of Clinical Investigation 94 506–515. (https://doi. reveals a novel mode of receptor dimerization and coactivator org/10.1172/JCI117362) recognition. Cell 110 93–105. (https://doi.org/10.1016/S0092- Aggarwal RR, Feng FY & Small EJ 2017 Emerging categories of disease in 8674(02)00817-6) advanced prostate cancer and their therapeutic implications. Bohl CE, Wu Z, Miller DD, Bell CE & Dalton JT 2007 Crystal structure Oncology 31 467–474. of the T877A human androgen receptor ligand-binding domain Agoulnik IU, Tong XW, Fischer DC, Körner K, Atkinson NE, Edwards DP, complexed to cyproterone acetate provides insight for ligand- Headon DR, Weigel NL & Kieback DG 2004 A germline variation in induced conformational changes and structure-based drug design. the progesterone receptor gene increases transcriptional activity and Journal of Biological Chemistry 282 13648–13655. (https://doi. may modify ovarian cancer risk. Journal of Clinical Endocrinology and org/10.1074/jbc.M611711200) Metabolism 89 6340–6347. (https://doi.org/10.1210/jc.2004-0114) Bourguet W, Vivat V, Wurtz JM, Chambon P, Gronemeyer H & Moras D Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, 2000 Crystal structure of a heterodimeric complex of RAR and RXR Chatila WK, Chakravarty D, Han GC, Coleman I, et al. 2018 The ligand-binding domains. Molecular Cell 5 289–298. (https://doi. long tail of oncogenic drivers in prostate cancer. Nature Genetics 50 org/10.1016/S1097-2765(00)80424-4) 645–651. (https://doi.org/10.1038/s41588-018-0078-z) Brinkmann AO, Faber PW, van Rooij HCJ, Kuiper GGJM, Ris C, Armstrong HK, Koay YC, Irani S, Das R, Nassar ZD, Australian Prostate Klaassen P, van der Korput JAGM, Voorhorst MM, van Laar JH & Cancer BioResource, Selth LA, Centenera MM, McAlpine SR & Mulder E 1989 The human androgen receptor: domain structure, Butler LM 2016 A novel class of Hsp90 C-terminal modulators have genomic organization and regulation of expression. Journal of Steroid pre-clinical efficacy in prostate tumor cells without induction of a Biochemistry 34 307–310. (https://doi.org/10.1016/0022- heat shock response. Prostate 76 1546–1559. (https://doi. 4731(89)90098-8) org/10.1002/pros.23239) Brzozowski AM, Pike ACW, Dauter Z, Hubbard RE, Bonn T, Engström O, Arora K & Barbieri CE 2018 Molecular subtypes of prostate cancer. Öhman L, Greene GL, Gustafsson JA & Carlquist M 1997 Molecular Current Oncology Reports 20 58–58. (https://doi.org/10.1007/s11912- basis of agonism and antagonism in the oestrogen receptor. Nature 018-0707-9) 389 753–758. (https://doi.org/10.1038/39645) Arora V K, Schenkein E, Murali R, Subudhi S K, Wongvipat J, Balbas M Buzón V, Carbó LR, Estruch SB, Fletterick RJ & Estébanez-Perpiñá E 2012 D, Shah N, Cai L, Efstathiou E, Logothetis C, et al. 2013 A conserved surface on the ligand binding domain of nuclear Glucocorticoid receptor confers resistance to antiandrogens by receptors for allosteric control. Molecular and Cellular Endocrinology bypassing androgen receptor blockade. Cell 155 1309–1322. (https:// 348 394–402. (https://doi.org/10.1016/j.mce.2011.08.012) doi.org/10.1016/j.cell.2013.11.012) Caboni L & Lloyd DG 2013 Beyond the ligand-binding pocket: targeting Augello MA, Liu D, Deonarine LD, Robinson BD, Huang D, Stelloo S, alternate sites in nuclear receptors. Medicinal Research Reviews 29 Blattner M, Doane AS, Wong EWP, Chen Y, et al. 2019 CHD1 loss 1292–1327.

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R492 Cancer dimerization in disease

Carroll JS 2016 Mechanisms of oestrogen receptor (ER) gene regulation response to PTEN loss. Nature Medicine 19 1023–1029. (https://doi. in breast cancer. European Journal of Endocrinology 175 R41–R49. org/10.1038/nm.3216) (https://doi.org/10.1530/EJE-16-0124) Cioni B, Zwart W & Bergman AM 2018 Androgen receptor Carver BS, Tran J, Gopalan A, Chen Z, Shaikh S, Carracedo A, moonlighting in the prostate cancer microenvironment. Endocrine- Alimonti A, Nardella C, Varmeh S, Scardino PT, et al. 2009 Aberrant Related Cancer 25 R331–R349. (https://doi.org/10.1530/ERC-18-0042) ERG expression cooperates with loss of PTEN to promote cancer Clocchiatti A, Ghosh S, Procopio MG, Mazzeo L, Bordignon P, Ostano P, progression in the prostate. Nature Genetics 41 619–624. (https://doi. Goruppi S, Bottoni G, Katarkar A, Levesque M, et al. 2018 Androgen org/10.1038/ng.370) receptor functions as transcriptional repressor of cancer-associated Casey OM, Fang L, Hynes PG, Abou-Kheir WG, Martin PL, Tillman HS, fibroblast activation. Journal of Clinical Investigation 128 5531–5548. Petrovics G, Awwad HO, Ward Y, Lake R, et al. 2012 TMPRSS2-driven (https://doi.org/10.1172/JCI99159) ERG expression in vivo increases self-renewal and maintains Cortes CJ & La Spada AR 2018 X-linked spinal and bulbar muscular expression in a castration resistant subpopulation. PLoS ONE 7 atrophy: From clinical genetic features and molecular pathology to e41668. (https://doi.org/10.1371/journal.pone.0041668) mechanisms underlying disease toxicity. Advances in Experimental Centenera MM, Harris JM, Tilley WD & Butler LM 2008 The Medicine and Biology 1049 103–133. (https://doi.org/10.1007/978-3- contribution of different androgen receptor domains to receptor 319-71779-1_5) dimerization and signaling. Molecular Endocrinology 22 2373–2382. Cotter KA & Rubin MA 2018 Sequence of events in prostate cancer. (https://doi.org/10.1210/me.2008-0017) Nature 560 557–559. (https://doi.org/10.1038/d41586-018-06029-5) Centenera MM, Gillis JL, Hanson AR, Jindal S, Taylor RA, Risbridger GP, Dalal K, Roshan-Moniri M, Sharma A, Li H, Ban F, Hessein MD, Sutherland PD, Scher HI, Raj GV, Knudsen KE, et al. 2012 Evidence Hsing M, Singh K, LeBlanc E, Dehm S, et al. 2014 Selectively for efficacy of new Hsp90 inhibitors revealed by ex vivo culture of targeting the DNA-binding domain of the androgen receptor as a human prostate tumors. Clinical Cancer Research 18 3562–3570. prospective therapy for prostate cancer. Journal of Biological Chemistry (https://doi.org/10.1158/1078-0432.CCR-12-0782) 289 26417–26429. (https://doi.org/10.1074/jbc.M114.553818) Centenera MM, Fitzpatrick AK, Tilley WD & Butler LM 2013a Hsp90: Dalal K, Che M, Que NS, Sharma A, Yang R, Lallous N, Borgmann H, still a viable target in prostate cancer. Biochimica et Biophysica Acta Ozistanbullu D, Tse R, Ban F, et al. 2017 Bypassing drug-resistance 1835 211–218. (https://doi.org/10.1016/j.bbcan.2012.12.005) mechanisms of prostate cancer with small-molecules that target Centenera MM, Raj GV, Knudsen KE, Tilley WD & Butler LM 2013b Ex androgen receptor chromatin interactions. Molecular Cancer vivo culture of human prostate tissue and drug development. Nature Therapeutics 16 2281–2291. (https://doi.org/10.1158/1535-7163.MCT- Reviews: Urology 10 483–487. (https://doi.org/10.1038/ 17-0259) nrurol.2013.126) Dalton JT 2017 The long and winding road for selective androgen Centenera MM, Selth LA, Ebrahimie E, Butler LM & Tilley WD 2018 receptor modulators. British Journal of Clinical Pharmacology 83 New opportunities for targeting the androgen receptor in prostate 2131–2133. (https://doi.org/10.1111/bcp.13345) cancer. Cold Spring Harbor Perspectives in Medicine 8 a030478. (https:// Dasgupta S, Lonard DM & O'Malley BW 2014 Nuclear receptor doi.org/10.1101/cshperspect.a030478) coactivators: master regulators of human health and disease. Annual Chandra V, Huang P, Hamuro Y, Raghuram S, Wang Y, Burris TP & Review of Medicine 65 279–292. (https://doi.org/10.1146/annurev- Rastinejad F 2008 Structure of the intact PPAR-γ-RXR-α nuclear med-051812-145316) receptor complex on DNA. Nature 456 350–356. (https://doi. De Mol E, Fenwick RB, Phang CTW, Buzón V, Szulc E, De La Fuente A, org/10.1038/nature07413) Escobedo A, García J, Bertoncini CW, Estébanez-Perpiñá E, et al. 2016 Chandra V, Huang P, Potluri N, Wu D, Kim Y & Rastinejad F 2013 EPI-001, a compound active against castration-resistant prostate Multidomain integration in the structure of the HNF-4α nuclear cancer, targets transactivation unit 5 of the androgen receptor. ACS receptor complex. Nature 495 394–398. (https://doi.org/10.1038/ Chemical Biology 11 2499–2505. (https://doi.org/10.1021/ nature11966) acschembio.6b00182) Chandra V, Wu D, Li S, Potluri N, Kim Y & Rastinejad F 2017 The De Mol E, Szulc E, Di Sanza C, Martínez-Cristóbal P, Bertoncini CW, quaternary architecture of RARβ-RXRα heterodimer facilitates Fenwick RB, Frigolé-Vivas M, Masín M, Hunter I, Buzón V, et al. domain-domain signal transmission. Nature Communications 8 2018 Regulation of androgen receptor activity by transient 868–868. (https://doi.org/10.1038/s41467-017-00981-y) interactions of its with general transcription Charmandari E, Ichijo T, Jubiz W, Baid S, Zachman K, Chrousos GP & regulators. Structure 26 145–152.e3. (https://doi.org/10.1016/j. Kino T 2008 A novel point mutation in the amino terminal domain str.2017.11.007) of the human glucocorticoid receptor (hGR) gene enhancing hGR- Dehm SM & Tindall DJ 2011 Alternatively spliced androgen receptor mediated gene expression. Journal of Clinical Endocrinology and variants. Endocrine-Related Cancer 18 R183–R196. (https://doi. Metabolism 93 4963–4968. (https://doi.org/10.1210/jc.2008-0892) org/10.1530/ERC-11-0141) Charmandari E, Kino T & Chrousos GP 2013 Primary generalized del Sol A, Fujihashi H, Amoros D & Nussinov R 2006 Residues crucial familial and sporadic glucocorticoid resistance (Chrousos syndrome) for maintaining short paths in network communication mediate and hypersensitivity. In Hormone Resistance and Hypersensitivity. From signaling in proteins. Molecular Systems Biology 2 2006.0019. (https:// Genetics to Clinical Management. Eds M Maghnie, S Loche, M Cappa, doi.org/10.1038/msb4100063) L Ghizzoni & R Lorini. vol 24, pp 67–85. (https://doi. Ellis JA, Stebbing M & Harrap SB 2001 Polymorphism of the androgen org/10.1159/000342505) receptor gene is associated with male pattern baldness. Journal of Chatonnet F, Guyot R, Benoit G & Flamant F 2013 Genome-wide Investigative Dermatology 116 452–455. (https://doi. analysis of thyroid hormone receptors shared and specific functions org/10.1046/j.1523-1747.2001.01261.x) in neural cells. PNAS 110 E766–E775. (https://doi.org/10.1073/ Eskew JD, Sadikot T, Morales P, Duren A, Dunwiddie I, Swink M, pnas.1210626110) Zhang X, Hembruff S, Donnelly A, Rajewski RA, et al. 2011 Chen Y, Sawyers CL & Scher HI 2008 Targeting the androgen receptor Development and characterization of a novel C-terminal inhibitor pathway in prostate cancer. Current Opinion in Pharmacology 8 of Hsp90 in androgen dependent and independent prostate cancer 440–448. (https://doi.org/10.1016/j.coph.2008.07.005) cells. BMC Cancer 11 468. (https://doi.org/10.1186/1471-2407-11- Chen Y, Chi P, Rockowitz S, Iaquinta PJ, Shamu T, Shukla S, Gao D, 468) Sirota I, Carver BS, Wongvipat J, et al. 2013 ETS factors reprogram Estebanez-Perpina E, Arnold LA, Nguyen P, Rodrigues ED, Mar E, the androgen receptor cistrome and prime prostate tumorigenesis in Bateman R, Pallai P, Shokat KM, Baxter JD, Guy RK, et al. 2007 A

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R493 Cancer dimerization in disease

surface on the androgen receptor that allosterically regulates Grosdidier S, Carbó LR, Buzón V, Brooke G, Nguyen P, Baxter JD, coactivator binding. PNAS 104 16074–16079. (https://doi. Bevan C, Webb P, Estébanez-Perpiñá E & Fernández-Recio J 2012 org/10.1073/pnas.0708036104) Allosteric conversation in the androgen receptor ligand-binding Estébanez-Perpiñá E, Moore JMR, Mar E, Delgado-Rodrigues E, Nguyen P, domain surfaces. Molecular Endocrinology 26 1078–1090. (https://doi. Baxter JD, Buehrer BM, Webb P, Fletterick RJ & Guy RK 2005 The org/10.1210/me.2011-1281) molecular mechanisms of coactivator utilization in ligand-dependent Guy NC, Garcia YA & Cox MB 2015 Therapeutic targeting of the FKBP52 transactivation by the androgen receptor. Journal of Biological co-chaperone in receptor-regulated physiology and Chemistry 280 8060–8068. (https://doi.org/10.1074/jbc.M407046200) disease. Current Molecular Pharmacology 9 109–125. (https://doi.org/1 Estébanez-Perpiñá E, Arnold LA, Jouravel N, Togashi M, Blethrow J, 0.2174/1874467208666150519114115) Mar E, Nguyen P, Phillips KJ, Baxter JD, Webb P, et al. 2007a Hay CW & McEwan IJ 2012 The impact of point mutations in the Structural insight into the mode of action of a direct inhibitor of human androgen receptor: classification of mutations on the basis of coregulator binding to the thyroid hormone receptor. Molecular transcriptional activity. PLoS ONE 7 e32514. (https://doi.org/10.1371/ Endocrinology 21 2919–2928. (https://doi.org/10.1210/me.2007-0174) journal.pone.0032514)

Estébanez-Perpiñá E, Jouravel N & Fletterick RJ 2007b Perspectives on He B & Wilson EM 2002 The NH2-terminal and carboxyl-terminal designs of antiandrogens for prostate cancer. Expert Opinion on in the human androgen receptor. Molecular Genetics and Discovery 2 1341–1355. (https://doi. Metabolism 75 293–298. (https://doi.org/10.1016/S1096- org/10.1517/17460441.2.10.1341) 7192(02)00009-4) Evans RM & Mangelsdorf DJ 2014 Nuclear receptors, RXR, and the big He B, Gampe RT, Kole AJ, Hnat AT, Stanley TB, An G, Stewart EL, bang. Cell 157 255–266. (https://doi.org/10.1016/j.cell.2014.03.012) Kalman RI, Minges JT & Wilson EM 2004 Structural basis for Faus H & Haendler B 2006 Post-translational modifications of steroid androgen receptor interdomain and coactivator interactions suggests receptors. Biomedicine and Pharmacotherapy 60 520–528. (https://doi. a transition in nuclear receptor activation function . org/10.1016/j.biopha.2006.07.082) Molecular Cell 16 425–438. (https://doi.org/10.1016/j. Fernandez EJ 2018 Allosteric pathways in nuclear receptors – Potential molcel.2004.09.036) targets for drug design. Pharmacology and Therapeutics 183 152–159. He S, Zhang C, Shafi AA, Sequeira M, Acquaviva J, Friedland JC, Sang J, (https://doi.org/10.1016/j.pharmthera.2017.10.014) Smith DL, Weigel NL, Wada Y, et al. 2013 Potent activity of the Fernandez EJ, Gahlot V, Rodriguez C & Amburn J 2017 DNA-induced Hsp90 inhibitor ganetespib in prostate cancer cells irrespective of unfolding of the thyroid hormone receptor α A/B domain through androgen receptor status or variant receptor expression. International allostery. FEBS Open Bio 7 854–864. (https://doi.org/10.1002/2211- Journal of Oncology 42 35–43. (https://doi.org/10.3892/ijo.2012.1698) 5463.12229) Hermanson O, Glass CK & Rosenfeld MG 2002 Nuclear receptor Foley C & Mitsiades N 2016 Moving beyond the androgen receptor coregulators: multiple modes of modification. Trends in Endocrinology (AR): targeting AR-interacting proteins to treat prostate cancer. and Metabolism 13 55–60. (https://doi.org/10.1016/S1043- Hormones and Cancer 7 84–103. (https://doi.org/10.1007/s12672-015- 2760(01)00527-6) 0239-9) Hernández MP, Chadli A & Toft DO 2002 HSP40 binding is the first step Frank F, Okafor CD & Ortlund EA 2018 The first crystal structure of a in the HSP90 chaperoning pathway for the progesterone receptor. DNA-free nuclear receptor DNA binding domain sheds light on Journal of Biological Chemistry 277 11873–11881. (https://doi. DNA-driven allostery in the glucocorticoid receptor. Scientific Reports org/10.1074/jbc.M111445200) 8 13497. (https://doi.org/10.1038/s41598-018-31812-9) Hilser VJ, Wrabl JO & Motlagh HN 2012 Structural and energetic basis Fuentes-Prior P, Rojas A, Hagler AT & Estébanez-Perpiñá E 2019 Diversity of allostery. Annual Review of Biophysics 41 585–609. (https://doi. of quaternary structures regulates nuclear receptor activities. Trends org/10.1146/annurev-biophys-050511-102319) in Biochemical Sciences 44 2–6. (https://doi.org/10.1016/j. Ho Y & Dehm SM 2017 Androgen receptor rearrangement and splicing tibs.2018.09.005) variants in resistance to endocrine therapies in prostate cancer. Gallastegui N, Mackinnon JAG, Fletterick RJ & Estébanez-Perpiñá E 2015 Endocrinology 158 1533–1542. (https://doi.org/10.1210/en.2017- Advances in our structural understanding of orphan nuclear 00109) receptors. Trends in Biochemical Sciences 40 25–35. (https://doi. Holzer G, Roux N & Laudet V 2017 Evolution of ligands, receptors and org/10.1016/j.tibs.2014.11.002) metabolizing of thyroid signaling. Molecular and Cellular Giacinti S, Poti G, Roberto M, Macrini S, Bassanelli M, Di Pietro F, Endocrinology 459 5–13. (https://doi.org/10.1016/j.mce.2017.03.021) Aschelter AM, Ceribelli A, Ruggeri EM & Marchetti P 2018 Molecular Huang P, Chandra V & Rastinejad F 2010 Structural overview of the basis of drug resistance and insights for new treatment approaches nuclear receptor superfamily: insights into physiology and in mCRPC. Anticancer Research 38 6029–6039. (https://doi. therapeutics. Annual Review of Physiology 72 247–272. (https://doi. org/10.21873/anticanres.12953) org/10.1146/annurev-physiol-021909-135917) Gibson DA, Saunders PTK & McEwan IJ 2018 Androgens and androgen Huang S, Gulzar ZG, Salari K, Lapointe J, Brooks JD & Pollack JR 2012 receptor: above and beyond. Molecular and Cellular Endocrinology 465 Recurrent deletion of CHD1 in prostate cancer with relevance to cell 1–3. (https://doi.org/10.1016/j.mce.2018.02.013) invasiveness. Oncogene 31 4164–4170. (https://doi.org/10.1038/ Gioeli D & Paschal BM 2012 Post-translational modification of the onc.2011.590) androgen receptor. Molecular and Cellular Endocrinology 352 70–78. Huang W, Peng Y, Kiselar J, Zhao X, Albaqami A, Mendez D, Chen Y, (https://doi.org/10.1016/j.mce.2011.07.004) Chakravarthy S, Gupta S, Ralston C, et al. 2018 Multidomain Giudici M, Goni S, Fan R & Treuter E 2016 Nuclear receptor coregulators architecture of estrogen receptor reveals interfacial cross-talk in metabolism and disease. Handbook of Experimental Pharmacology between its DNA-binding and ligand-binding domains. Nature 233 95–135. (https://doi.org/10.1007/164_2015_5) Communications 9 3520–3520. (https://doi.org/10.1038/s41467-018- Gottlieb B, Beitel LK, Wu J, Elhaji YA & Trifiro M 2004 Nuclear receptors 06034-2) and disease: androgen receptor. Essays in Biochemistry 40 121–136. Huber BR, Desclozeaux M, West BL, Cunha-Lima ST, Nguyen HT, (https://doi.org/10.1042/bse0400121) Baxter JD, Ingraham HA & Fletterick RJ 2003a Thyroid hormone Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, receptor-β mutations conferring hormone resistance and reduced Quist MJ, Jing X, Lonigro RJ, Brenner JC, et al. 2012 The mutational release exhibit decreased stability in the N-terminal landscape of lethal castration-resistant prostate cancer. Nature 487 ligand-binding domain. Molecular Endocrinology 17 107–116. (https:// 239–243. (https://doi.org/10.1038/nature11125) doi.org/10.1210/me.2002-0097)

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R494 Cancer dimerization in disease

Huber BR, Sandler B, West BL, Cunha Lima ST, Nguyen HT, Apriletti JW, Kino T, Vottero A, Charmandari E & Chrousos GP 2002 Familial/ Baxter JD & Fletterick RJ 2003b Two resistance to thyroid hormone sporadic glucocorticoid resistance syndrome and hypertension. mutants with impaired hormone binding. Molecular Endocrinology 17 Annals of the New York Academy of Sciences 970 101–111. (https://doi. 643–652. (https://doi.org/10.1210/me.2002-0095) org/10.1111/j.1749-6632.2002.tb04416.x) Hughes IA, Werner R, Bunch T & Hiort O 2012 Androgen insensitivity Kita K, Shiota M, Tanaka M, Otsuka A, Matsumoto M, Kato M, syndrome. Seminars in Reproductive Medicine 30 432–442. (https://doi. Tamada S, Iwao H, Miura K, Nakatani T, et al. 2017 Heat shock org/10.1055/s-0032-1324728) protein 70 inhibitors suppress androgen receptor expression in Hur E, Pfaff SJ, Payne ES, Grøn H, Buehrer BM & Fletterick RJ 2004 LNCaP95 prostate cancer cells. Cancer Science 108 1820–1827. Recognition and accommodation at the androgen receptor (https://doi.org/10.1111/cas.13318) coactivator binding interface. PLoS Biology 2 E274–E274. (https://doi. Knudsen KE & Penning TM 2010 Partners in crime: deregulation of AR org/10.1371/journal.pbio.0020274) activity and androgen synthesis in prostate cancer. Trends in Hurt DE, Suzuki S, Mayama T, Charmandari E & Kino T 2016 Structural Endocrinology and Metabolism 21 315–324. (https://doi.org/10.1016/j. analysis on the pathologic mutant glucocorticoid receptor ligand- tem.2010.01.002) binding domains. Molecular Endocrinology 30 173–188. (https://doi. Kojetin DJ, Burris TP, Jensen EV & Khan SA 2008 Implications of the org/10.1210/me.2015-1177) binding of tamoxifen to the coactivator recognition site of the Imamura Y & Sadar MD 2016 Androgen receptor targeted therapies in estrogen receptor. Endocrine-Related Cancer 15 851–870. (https://doi. castration-resistant prostate cancer: bench to clinic. International org/10.1677/ERC-07-0281) Journal of Urology 23 654–665. (https://doi.org/10.1111/iju.13137) Kokontis JM & Liao S 1999 Molecular action of androgen in the normal Jenster G, van der Korput HA, Trapman J & Brinkmann AO 1995 and neoplastic prostate. Vitamins and Hormones 55 219–307. (https:// Identification of two transcription activation units in the N-terminal doi.org/10.1016/S0083-6729(08)60937-1) domain of the human androgen receptor. Journal of Biological Koryakina Y, Ta HQ & Gioeli D 2014 Androgen receptor Chemistry 270 7341–7346. (https://doi.org/10.1074/jbc.270.13.7341) phosphorylation: biological context and functional consequences. Jernberg E, Bergh A & Wikström P 2017 Clinical relevance of androgen Endocrine-Related Cancer 21 T131–T145. (https://doi.org/10.1530/ receptor alterations in prostate cancer. Endocrine Connections 6 ERC-13-0472) R146–R161. (https://doi.org/10.1530/EC-17-0118) Kumar R, Betney R, Li J, Thompson EB & McEwan IJ 2004 Induced Jin HJ, Kim J & Yu J 2013 Androgen receptor genomic regulation. α-helix structure in AF1 of the androgen receptor upon binding Translational Andrology and Urology 2 157–177. (https://doi. TFIIF. Biochemistry 43 3008–3013. (https://doi. org/10.3978/j.issn.2223-4683.2013.09.01) org/10.1021/bi035934p) Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Kumar A, Coleman I, Morrissey C, Zhang X, True LD, Gulati R, Maneval EC, Chen I, Darimont B, et al. 2013 A clinically relevant Etzioni R, Bolouri H, Montgomery B, White T, et al. 2016 Substantial androgen receptor mutation confers resistance to second-generation interindividual and limited intraindividual genomic diversity among antiandrogens enzalutamide and ARN-509. Cancer Discovery 3 tumors from men with metastatic prostate cancer. Nature Medicine 1020–1029. (https://doi.org/10.1158/2159-8290.CD-13-0226) 22 369–378. (https://doi.org/10.1038/nm.4053) Jouravel N, Sablin E, Togashi M, Baxter JD, Webb P & Fletterick RJ 2009 Kuriyan J 2004 Allostery and coupled sequence variation in nuclear Molecular basis for dimer formation of TRβ variant D355R. Proteins hormone receptors. Cell 116 354–356. (https://doi.org/10.1016/ 75 111–117. (https://doi.org/10.1002/prot.22225) S0092-8674(04)00125-4) Karamouzis MV, Papavassiliou KA, Adamopoulos C & Papavassiliou AG Langley E, Zhou ZX & Wilson EM 1995 Evidence for an anti-parallel 2016 Targeting androgen/estrogen receptors crosstalk in cancer. orientation of the ligand-activated human androgen receptor dimer. Trends in Cancer 2 35–48. (https://doi.org/10.1016/j. Journal of Biological Chemistry 270 29983–29990. (https://doi. trecan.2015.12.001) org/10.1074/jbc.270.50.29983) Karthaus WR & Sawyers CL 2019 Strategies to identify and target cells of Langley E, Kemppainen JA & Wilson EM 1998 Intermolecular NH2-/ origin in prostate cancer. Journal of the National Cancer Institute 111 carboxyl-terminal interactions in androgen receptor dimerization 221–223. (https://doi.org/10.1093/jnci/djy146) revealed by mutations that cause androgen insensitivity. Journal of Katzenellenbogen JA, Mayne CG, Katzenellenbogen BS, Greene GL & Biological Chemistry 273 92–101. (https://doi.org/10.1074/ Chandarlapaty S 2018 Structural underpinnings of oestrogen jbc.273.1.92) receptor mutations in endocrine therapy resistance. Nature Reviews: Lempiäinen JK, Niskanen EA, Vuoti KM, Lampinen RE, Göös H, Cancer 18 377–388. (https://doi.org/10.1038/s41568-018-0001-z) Varjosalo M & Palvimo JJ 2017 Agonist-specific protein interactomes Kauppi B, Jakob C, Färnegårdh M, Yang J, Ahola H, Alarcon M, Calles K, of glucocorticoid and androgen receptor as revealed by proximity Engström O, Harlan J, Muchmore S, et al. 2003 The three- mapping. Molecular and Cellular Proteomics 16 1462–1474. (https:// dimensional structures of antagonistic and agonistic forms of the doi.org/10.1074/mcp.M117.067488) glucocorticoid receptor ligand-binding domain: RU-486 induces a Li J, Motlagh HN, Chakuroff C, Thompson EB & Hilser VJ 2012 transconformation that leads to active antagonism. Journal of Thermodynamic dissection of the intrinsically disordered N-terminal Biological Chemistry 278 22748–22754. (https://doi.org/10.1074/jbc. domain of human glucocorticoid receptor. Journal of Biological M212711200) Chemistry 287 26777–26787. (https://doi.org/10.1074/jbc. Khorasanizadeh S & Rastinejad F 2016 Visualizing the architectures and M112.355651) interactions of nuclear receptors. Endocrinology 157 4212–4221. Li Q, Deng Q, Chao HP, Liu X, Lu Y, Lin K, Liu B, Tang GW, Zhang D, (https://doi.org/10.1210/en.2016-1559) Tracz A, et al. 2018 Linking prostate cancer cell AR heterogeneity to King JC, Xu J, Wongvipat J, Hieronymus H, Carver BS, Leung DH, distinct castration and enzalutamide responses. Nature Taylor BS, Sander C, Cardiff RD, Couto SS, et al. 2009 Cooperativity Communications 9 3600–3600. (https://doi.org/10.1038/s41467-018- of TMPRSS2-ERG with PI3-kinase pathway activation in prostate 06067-7) oncogenesis. Nature Genetics 41 524–526. (https://doi.org/10.1038/ Lieberman AP 2018 Spinal and bulbar muscular atrophy. Handbook of ng.371) Clinical Neurology 148 625–632. (https://doi.org/10.1016/B978-0-444- Kino T & Chrousos GP 2001 Glucocorticoid and mineralocorticoid 64076-5.00040-5) resistance/hypersensitivity syndromes. Journal of Endocrinology 169 Liu LL, Xie N, Sun S, Plymate S, Mostaghel E & Dong X 2014 437–445. (https://doi.org/10.1677/joe.0.1690437) Mechanisms of the androgen receptor splicing in prostate cancer

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R495 Cancer dimerization in disease

cells. Oncogene 33 3140–3150. (https://doi.org/10.1038/ of Biological Chemistry 288 1939–1952. (https://doi.org/10.1074/jbc. onc.2013.284) M112.428409) Liu B, Gu X, Huang T, Luan Y & Ding X 2017 Identification of Mohler ML, Coss CC, Duke CB, Patil SA, Miller DD & Dalton JT 2012 TMPRSS2-ERG mechanisms in prostate cancer invasiveness: Androgen receptor antagonists: a patent review (2008–2011). Expert involvement of MMP-9 and plexin B1. Oncology Reports 37 201–208. Opinion on Therapeutic Patents 22 541–565. (https://doi.org/10.1517/1 (https://doi.org/10.3892/or.2016.5277) 3543776.2012.682571) Lorente D, Mateo J, Zafeiriou Z, Smith AD, Sandhu S, Ferraldeschi R & Monaghan AE & McEwan IJ 2016 A sting in the tail: the N-terminal De Bono JS 2015 Switching and withdrawing hormonal agents for domain of the androgen receptor as a drug target. Asian Journal of castration-resistant prostate cancer. Nature Reviews: Urology 12 37–47. Andrology 18 687–694. (https://doi.org/10.4103/1008-682X.181081) (https://doi.org/10.1038/nrurol.2014.345) Mongan NP, Tadokoro-Cuccaro R, Bunch T & Hughes IA 2015 Androgen Lou X, Toresson G, Benod C, Suh JH, Philips KJ, Webb P & Gustafsson J- insensitivity syndrome. Best Practice and Research: Clinical A 2014 Structure of the α- β Endocrinology and Metabolism 29 569–580. (https://doi.org/10.1016/j. (RXRα-LXRβ) heterodimer on DNA. Nature Structural and Molecular beem.2015.04.005) Biology 21 277–281. (https://doi.org/10.1038/nsmb.2778) Moore JT, Collins JL & Pearce KH 2006 The nuclear receptor superfamily Love MI, Huska MR, Jurk M, Schöpflin R, Starick SR, Schwahn K, and drug discovery. ChemMedChem 1 504–523. (https://doi. Cooper SB, Yamamoto KR, Thomas-Chollier M, Vingron M, et al. org/10.1002/cmdc.200600006) 2017 Role of the chromatin landscape and sequence in determining Nadal M, Prekovic S, Gallastegui N, Helsen C, Abella M, Zielinska K, cell type-specific genomic glucocorticoid receptor binding and gene Gay M, Vilaseca M, Taulès M, Houtsmuller AB, et al. 2017 Structure of regulation. Nucleic Acids Research 45 1805–1819. (https://doi. the homodimeric androgen receptor ligand-binding domain. Nature org/10.1093/nar/gkw1163) Communications 8 14388. (https://doi.org/10.1038/ncomms14388) Lubahn DB, Joseph DR, Sullivan PM, Willard HF, French FS & Narayanan S, Srinivas S & Feldman D 2016 Androgen-glucocorticoid Wilson EM 1988 Cloning of human androgen receptor interactions in the era of novel prostate cancer therapy. Nature complementary DNA and localization to the X chromosome. Science Reviews: Urology 13 47–60. (https://doi.org/10.1038/nrurol.2015.254) 240 327–330. (https://doi.org/10.1126/science.3353727) Narayanan R, Coss CC & Dalton JT 2018 Development of selective Luisi BF, Xu WX, Otwinowski Z, Freedman LP, Yamamoto KR & Sigler PB androgen receptor modulators (SARMs). Molecular and Cellular 1991 Crystallographic analysis of the interaction of the Endocrinology 465 134–142. (https://doi.org/10.1016/j. glucocorticoid receptor with DNA. Nature 352 497–505. (https://doi. mce.2017.06.013) org/10.1038/352497a0) Nasrazadani A, Thomas RA, Oesterreich S & Lee AV 2018 Precision Luo J, Attard G, Balk SP, Bevan C, Burnstein K, Cato L, Cherkasov A, De medicine in hormone receptor-positive breast cancer. Frontiers in Bono JS, Dong Y, Gao AC, et al. 2018 Role of androgen receptor Oncology 8 144–144. (https://doi.org/10.3389/fonc.2018.00144) variants in prostate cancer: report from the 2017 mission androgen Nevedomskaya E, Baumgart SJ & Haendler B 2018 Recent advances in receptor variants meeting. European Urology 73 715–723. (https://doi. prostate cancer treatment and drug discovery. International Journal of org/10.1016/j.eururo.2017.11.038) Molecular Sciences 19 E1359. (https://doi.org/10.3390/ijms19051359) Mackinnon JAG, Gallastegui N, Osguthorpe DJ, Hagler AT & Estébanez- Nicolaides NC & Charmandari E 2015 Chrousos syndrome: from Perpiñá E 2014 Allosteric mechanisms of nuclear receptors: insights molecular pathogenesis to therapeutic management. European Journal from computational simulations. Molecular and Cellular Endocrinology of Clinical Investigation 45 504–514. (https://doi.org/10.1111/ 393 75–82. (https://doi.org/10.1016/j.mce.2014.05.017) eci.12426) Matias PM, Donner P, Coelho R, Thomaz M, Peixoto C, Macedo S, Nicolaides NC & Charmandari E 2017 Novel insights into the molecular Otto N, Joschko S, Scholz P, Wegg A, et al. 2000 Structural evidence mechanisms underlying generalized glucocorticoid resistance and for ligand specificity in the binding domain of the human androgen hypersensitivity syndromes. Hormones 16 124–138. (https://doi. receptor: implications for pathogenic gene mutations. Journal of org/10.14310/horm.2002.1728) Biological Chemistry 275 26164–26171. (https://doi.org/10.1074/jbc. Nicolaides NC, Galata Z, Kino T, Chrousos GP & Charmandari E 2010 M004571200) The human glucocorticoid receptor: molecular basis of biologic McEwan IJ 2012a Intrinsic disorder in the androgen receptor: function. Steroids 75 1–12. (https://doi.org/10.1016/j. identification, characterisation and drugability. Molecular Biosystems steroids.2009.09.002) 8 82–90. (https://doi.org/10.1039/c1mb05249g) Nicolaides NC, Geer EB, Vlachakis D, Roberts ML, Psarra AM, McEwan IJ 2012b Nuclear hormone receptors: allosteric switches. Moutsatsou P, Sertedaki A, Kossida S & Charmandari E 2015a A Molecular and Cellular Endocrinology 348 345–347. (https://doi. novel mutation of the hGR gene causing Chrousos syndrome. org/10.1016/j.mce.2011.08.035) European Journal of Clinical Investigation 45 782–791. (https://doi. McEwan IJ, Lavery D, Fischer K & Watt K 2007 Natural disordered org/10.1111/eci.12470) sequences in the amino terminal domain of nuclear receptors: Nicolaides NC, Lamprokostopoulou A, Polyzos A, Kino T, Katsantoni E, lessons from the androgen and glucocorticoid receptors. Nuclear Triantafyllou P, Christophoridis A, Katzos G, Dracopoulou M, Receptor Signaling 5 e001–e001. (https://doi.org/10.1621/nrs.05001) Sertedaki A, et al. 2015b Transient generalized glucocorticoid Meijsing SH, Pufall MA, So AY, Bates DL, Chen L & Yamamoto KR 2009 hypersensitivity. European Journal of Clinical Investigation 45 DNA binding site sequence directs glucocorticoid receptor structure 1306–1315. (https://doi.org/10.1111/eci.12554) and activity. Science 324 407–410. (https://doi.org/10.1126/ Nwachukwu JC, Srinivasan S, Zheng Y, Wang S, Min J, Dong C, Liao Z, science.1164265) Nowak J, Wright NJ, Houtman R, et al. 2016 Predictive features of Michigami T, Suga A, Yamazaki M, Shimizu C, Cai G, Okada S & ligand-specific signaling through the estrogen receptor. Molecular Ozono K 1999 Identification of amino acid sequence in the hinge Systems Biology 12 864–864. (https://doi.org/10.15252/ region of human receptor that transfers a cytosolic msb.20156701) protein to the nucleus. Journal of Biological Chemistry 274 Onigata K & Szinnai G 2014 Resistance to thyroid hormone. In 33531–33538. (https://doi.org/10.1074/jbc.274.47.33531) Paediatric Thyroidology. Ed G Szinnai. Basel, Switzerland: Karger. Minges JT, Su S, Grossman G, Blackwelder AJ, Pop EA, Mohler JL & (https://doi.org/10.1159/000363159) Wilson EM 2013 Melanoma antigen-a11 (MAGE-a11) enhances Paakinaho V, Presman DM, Ball DA, Johnson TA, Schiltz RL, Levitt P, transcriptional activity by linking androgen receptor dimers. Journal Mazza D, Morisaki T, Karpova TS & Hager GL 2017 Single-molecule

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R496 Cancer dimerization in disease

analysis of steroid receptor and action in living cells. Nature translocation and distribution. Steroids 78 182–194. (https://doi. Communications 8 15896. (https://doi.org/10.1038/ncomms15896) org/10.1016/j.steroids.2012.10.016) Park K, Dalton JT, Narayanan R, Barbieri CE, Hancock ML, Bostwick DG, Robertson S, Rohwer JM, Hapgood JP & Louw A 2013b Impact of Steiner MS & Rubin MA 2014 TMPRSS2:ERG gene fusion predicts glucocorticoid receptor density on ligand-independent dimerization, subsequent detection of prostate cancer in patients with high-grade cooperative ligand-binding and basal priming of transactivation: a prostatic intraepithelial neoplasia. Journal of Clinical Oncology 32 cell culture model. PLoS ONE 8 e64831. (https://doi.org/10.1371/ 206–211. journal.pone.0064831) Paschalis A, Sharp A, Welti JC, Neeb A, Raj GV, Luo J, Plymate SR & de Robinson D, Van Allen E M, Wu Y-M, Schultz N, Lonigro R J, Bono JS 2018 in prostate cancer. Nature Reviews: Mosquera J-M, Montgomery B, Taplin M-E, Pritchard C C, Attard G, Clinical Oncology 15 663–675. (https://doi.org/10.1038/s41571-018- et al. 2015 Integrative clinical of advanced prostate cancer. 0085-0) Cell 161 1215–1228. (https://doi.org/10.1016/j.cell.2015.05.001) Pennuto M & Rinaldi C 2018 From gene to therapy in spinal and bulbar Rodrigues LU, Rider L, Nieto C, Romero L, Karimpour-Fard A, Loda M, muscular atrophy: are we there yet? Molecular and Cellular Lucia MS, Wu M, Shi L, Cimic A, et al. 2015 Coordinate loss of Endocrinology 465 113–121. (https://doi.org/10.1016/j. MAP3K7 and CHD1 promotes aggressive prostate cancer. Cancer mce.2017.07.005) Research 75 1021–1034. (https://doi.org/10.1158/0008-5472.CAN-14- Perissi V & Rosenfeld MG 2005 Controlling nuclear receptors: the 1596) circular logic of cofactor cycles. Nature Reviews: Molecular Cell Biology Ruggero K, Farran-Matas S, Martinez-Tebar A & Aytes A 2018 Epigenetic 6 542–554. (https://doi.org/10.1038/nrm1682) regulation in prostate cancer progression. Current Ponnusamy S, Coss CC, Thiyagarajan T, Watts K, Hwang DJ, He Y, Reports 4 101–115. (https://doi.org/10.1007/s40610-018-0095-9) Selth LA, McEwan IJ, Duke CB, Pagadala J, et al. 2017 Novel selective Sack JS, Kish KF, Wang C, Attar RM, Kiefer SE, An Y, Wu GY, Scheffler JE, agents for the degradation of androgen receptor variants to treat Salvati ME, Krystek SR, et al. 2001 Crystallographic structures of the castration-resistant prostate cancer. Cancer Research 77 6282–6298. ligand-binding domains of the androgen receptor and its T877A (https://doi.org/10.1158/0008-5472.CAN-17-0976) mutant complexed with the natural agonist dihydrotestosterone. Prescott J & Coetzee GA 2006 Molecular chaperones throughout the life PNAS 98 4904–4909. (https://doi.org/10.1073/pnas.081565498) cycle of the androgen receptor. Cancer Letters 231 12–19. (https:// Sadar MD 2011 Small molecule inhibitors targeting the ‘Achilles’ heel’ doi.org/10.1016/j.canlet.2004.12.037) of androgen receptor activity. Cancer Research 71 1208–1213. Presman DM & Hager GL 2017 More than meets the dimer: what is the (https://doi.org/10.1158/0008-5472.CAN_10-3398) quaternary structure of the glucocorticoid receptor? Transcription 8 Schaufele F, Carbonell X, Guerbadot M, Borngraeber S, Chapman MS, 32–39. (https://doi.org/10.1080/21541264.2016.1249045) Ma AAK, Miner JN & Diamond MI 2005 The structural basis of Presman DM, Ganguly S, Schiltz RL, Johnson TA, Karpova TS & androgen receptor activation: intramolecular and intermolecular Hager GL 2016 DNA binding triggers tetramerization of the amino-carboxy interactions. PNAS 102 9802–9807. (https://doi. glucocorticoid receptor in live cells. PNAS 113 8236–8241. (https:// org/10.1073/pnas.0408819102) doi.org/10.1073/pnas.1606774113) Schöne S, Jurk M, Helabad MB, Dror I, Lebars I, Kieffer B, Imhof P, Presman DM, Ball DA, Paakinaho V, Grimm JB, Lavis LD, Karpova TS & Rohs R, Vingron M, Thomas-Chollier M, et al. 2016 Sequences Hager GL 2017 Quantifying transcription factor binding dynamics at flanking the core-binding site modulate glucocorticoid receptor the single-molecule level in live cells. Methods 123 76–88. (https:// structure and activity. Nature Communications 7 12621. (https://doi. doi.org/10.1016/j.ymeth.2017.03.014) org/10.1038/ncomms12621) Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, Schrecengost R & Knudsen KE 2013 Molecular pathogenesis and Garofalo A, Gulati R, Carreira S, Eeles R, et al. 2016 Inherited DNA- progression of prostate cancer. Seminars in Oncology 40 244–258. repair gene mutations in men with metastatic prostate cancer. New (https://doi.org/10.1053/j.seminoncol.2013.04.001) England Journal of Medicine 375 443–453. (https://doi.org/10.1056/ Schwabe JW, Chapman L, Finch JT & Rhodes D 1993 The crystal NEJMoa1603144) structure of the estrogen-receptor DNA-binding domain bound to Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, DNA – how receptors discriminate between their response elements. Foye A, Kothari V, Perry MD, Bailey AM, et al. 2018 Genomic Cell 75 567–578. (https://doi.org/10.1016/0092-8674(93)90390-C) hallmarks and structural variation in metastatic prostate cancer. Cell Shaffer PL, Jivan A, Dollins DE, Claessens F & Gewirth DT 2004 174 758.e9–769.e9. (https://doi.org/10.1016/j.cell.2018.06.039) Structural basis of androgen receptor binding to selective androgen Rastinejad F, Ollendorff V & Polikarpov I 2015 Nuclear receptor full- response elements. PNAS 101 4758–4763. (https://doi.org/10.1073/ length architectures: confronting myth and illusion with high pnas.0401123101) resolution. Trends in Biochemical Sciences 40 16–24. (https://doi. Shen MM & Abate-Shen C 2010 Molecular Genetics of prostate cancer: org/10.1016/j.tibs.2014.10.011) new prospects for old challenges. Genes and Development 24 Ravindranathan P, Lee TK, Yang L, Centenera MM, Butler L, Tilley WD, 1967–2000. (https://doi.org/10.1101/gad.1965810) Hsieh JT, Ahn JM & Raj GV 2013 Peptidomimetic targeting of critical Spada ARL, Wilson EM, Lubahn DB, Harding AE & Fischbeck KH 1991 androgen receptor-coregulator interactions in prostate cancer. Nature Androgen receptor gene mutations in X-linked spinal and bulbar Communications 4 1923–1923. (https://doi.org/10.1038/ muscular atrophy. Nature 352 77–79. (https://doi. ncomms2912) org/10.1038/352077a0) Reig Ò, Marín-Aguilera M, Carrera G, Jiménez N, Paré L, García-Recio S, Srinivasan S, Nwachukwu JC, Parent AA, Cavett V, Nowak J, Hughes TS, Gaba L, Pereira MV, Fernández P, Prat A, et al. 2016 TMPRSS2-ERG in Kojetin DJ, Katzenellenbogen JA & Nettles KW 2013 Ligand-binding blood and docetaxel resistance in metastatic castration-resistant dynamics rewire cellular signaling via estrogen receptor-α. Nature prostate cancer. European Urology 70 709–713. (https://doi. Chemical Biology 9 326–332. (https://doi.org/10.1038/ org/10.1016/j.eururo.2016.02.034) nchembio.1214) Reinert T, Gonçalves R & Bines J 2018 Implications of ESR1 mutations Stelloo S, Nevedomskaya E, Kim Y, Schuurman K, Valle-Encinas E, in hormone receptor-positive breast cancer. Current Treatment Options Lobo J, Krijgsman O, Peeper DS, Chang SL, Feng FY-C, et al. 2018 in Oncology 19 24–24. (https://doi.org/10.1007/s11864-018-0542-0) Integrative epigenetic taxonomy of primary prostate cancer. Nature Robertson S, Hapgood JP & Louw A 2013a Glucocorticoid receptor Communications 9 4900–4900. (https://doi.org/10.1038/s41467-018- concentration and the ability to dimerize influence nuclear 07270-2)

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access Endocrine-Related A Jiménez-Panizo et al. Androgen receptor 26:8 R497 Cancer dimerization in disease

Sung MH, Guertin MJ, Baek S & Hager GL 2014 DNase footprint therapeutic targets. Hormones and Cancer 7 229–235. (https://doi. signatures are dictated by factor dynamics and DNA sequence. org/10.1007/s12672-016-0261-6) Molecular Cell 56 275–285. (https://doi.org/10.1016/j. Watson PA, Arora VK & Sawyers CL 2015 Emerging mechanisms of molcel.2014.08.016) resistance to androgen receptor inhibitors in prostate cancer. Swinstead EE, Miranda TB, Paakinaho V, Baek S, Goldstein I, Nature Reviews: Cancer 15 701–711. (https://doi.org/10.1038/ Hawkins M, Karpova TS, Ball D, Mazza D, Lavis LD, et al. 2016 nrc4016) Steroid receptors reprogram FoxA1 occupancy through dynamic Wedge DC, Gundem G, Mitchell T, Woodcock DJ, Martincorena I, chromatin transitions. Cell 165 593–605. (https://doi.org/10.1016/j. Ghori M, Zamora J, Butler A, Whitaker H, Kote-Jarai Z, et al. 2018 cell.2016.02.067) Sequencing of prostate cancers identifies new cancer genes, routes of Takeda DY, Spisák Sn, Seo JH, Bell C, O'Connor E, Korthauer K, Ribli D, progression and drug targets. Nature Genetics 50 682–692. (https:// Csabai I, Solymosi N, Szállási Z, et al. 2018 A somatically acquired doi.org/10.1038/s41588-018-0086-z) enhancer of the androgen receptor is a noncoding driver in Weikum ER, Knuesel MT, Ortlund EA & Yamamoto KR 2017 advanced prostate cancer. Cell 174 422–432.e13. (https://doi. Glucocorticoid receptor control of transcription: precision and org/10.1016/j.cell.2018.05.037) plasticity via allostery. Nature Reviews: Molecular Cell Biology 18 Tien AH & Sadar MD 2018 Order within a disordered structure. Structure 159–174. (https://doi.org/10.1038/nrm.2016.152) 26 4–6. (https://doi.org/10.1016/j.str.2017.12.007) Weikum ER, Liu X & Ortlund EA 2018 The nuclear receptor superfamily: Treviño LS & Weigel NL 2013 Phosphorylation: a fundamental regulator a structural perspective. Protein Science 27 1876–1892. (https://doi. of steroid receptor action. Trends in Endocrinology and Metabolism 24 org/10.1002/pro.3496) 515–524. (https://doi.org/10.1016/j.tem.2013.05.008) Weiss RE, Marcocci C, Bruno-Bossio G & Refetoff S 1993 Multiple Uo T, Plymate SR & Sprenger CC 2017 Allosteric alterations in the genetic factors in the heterogeneity of thyroid hormone resistance. androgen receptor and activity in prostate cancer. Endocrine-Related Journal of Clinical Endocrinology and Metabolism 76 257–259. (https:// Cancer 24 R335–R348. (https://doi.org/10.1530/ERC-17-0108) doi.org/10.1210/jcem.76.1.8421095) van der Steen T, Tindall DJ & Huang H 2013 Posttranslational Wilkinson L, Verhoog NJD & Louw A 2018 Disease- and treatment- modification of the androgen receptor in prostate cancer. associated acquired glucocorticoid resistance. Endocrine Connections International Journal of Molecular Sciences 14 14833–14859. (https:// 12 R328–R349. (https://doi.org/10.1530/EC-18-0421) doi.org/10.3390/ijms140714833) Wu YM, Cieslik M, Lonigro RJ, Vats P, Reimers MA, Cao X, Ning Y, van Royen ME, van Cappellen WA, de Vos C, Houtsmuller AB & Wang L, Kunju LP, de Sarkar N, et al. 2018 Inactivation of CDK12 Trapman J 2012 Stepwise androgen receptor dimerization. Journal of delineates a distinct immunogenic class of advanced prostate cancer. Cell Science 125 1970–1979. (https://doi.org/10.1242/jcs.096792) Cell 173 1770.e14–1782.e14. (https://doi.org/10.1016/j. Veras Ribeiro Filho H, Tambones IL, Mariano Gonçalves Dias M, cell.2018.04.034) Bernardi Videira N, Bruder M, Amorim Amato A & Migliorini Wurtz JM, Bourguet W, Renaud JP, Vivat Vr, Chambon P, Moras D & Figueira AC 2019 Modulation of nuclear receptor function: targeting Gronemeyer H 1996 A canonical structure for the ligand-binding the protein-DNA interface. Molecular and Cellular Endocrinology 484 domain of nuclear receptors. Nature Structural Biology 3 87–94. 1–14. (https://doi.org/10.1016/j.mce.2019.01.023) (https://doi.org/10.1038/nsb0196-87) Viswanathan SR, Ha G, Hoff AM, Wala JA, Carrot-Zhang J, Whelan CW, Yi P, Wang Z, Feng Q, Pintilie G D, Foulds C E, Lanz R B, Ludtke S J, Haradhvala NJ, Freeman SS, Reed SC, Rhoades J, et al. 2018 Schmid M F, Chiu W & O'Malley B W 2015 Structure of a Structural alterations driving castration-resistant prostate cancer biologically active estrogen receptor-coactivator complex on DNA. revealed by linked-read genome sequencing. Cell 174 433–447.e19. Molecular Cell 57 1047–1058. (https://doi.org/10.1016/j. (https://doi.org/10.1016/j.cell.2018.05.036) molcel.2015.01.025) Vitellius G, Fagart J, Delemer B, Amazit L, Ramos N, Bouligand J, Le Yu J, Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J, Billan F, Castinetti F, Guiochon-Mantel A, Trabado S, et al. 2016 Hu M, et al. 2010 An integrated network of androgen receptor, Three novel heterozygous point mutations of NR3C1 causing Polycomb, and TMPRSS2-ERG gene fusions in prostate cancer glucocorticoid resistance. Human Mutation 37 794–803. (https://doi. progression. Cancer Cell 17 443–454. (https://doi.org/10.1016/j. org/10.1002/humu.23008) ccr.2010.03.018) Vitellius G, Trabado S, Hoeffel C, Bouligand J, Bennet A, Castinetti F, Zennaro MC & Fernandes-Rosa F 2017 30 YEARS OF THE Decoudier B, Guiochon-Mantel A, Lombes M, Delemer B, et al. 2018 MINERALOCORTICOID RECEPTOR: Mineralocorticoid receptor Significant prevalence of NR3C1 mutations in incidentally mutations. Journal of Endocrinology 234 T93–T106. (https://doi. discovered bilateral adrenal hyperplasia: results of the French org/10.1530/JOE-17-0089) Muta-GR Study. European Journal of Endocrinology 178 411–423. Zhao D, Lu X, Wang G, Lan Z, Liao W, Li J, Liang X, Chen JR, Shah S, (https://doi.org/10.1530/EJE-17-1071) Shang X, et al. 2017 Synthetic essentiality of chromatin remodelling Voss TC, Schiltz RL, Sung MH, Yen PM, Stamatoyannopoulos JA, factor CHD1 in PTEN-deficient cancer. Nature 542 484–488. (https:// Biddie SC, Johnson TA, Miranda TB, John S & Hager GL 2011 doi.org/10.1038/nature21357) Dynamic exchange at regulatory elements during chromatin Zoubeidi A, Zardan A, Beraldi E, Fazli L, Sowery R, Rennie P, Nelson C & remodeling underlies assisted loading mechanism. Cell 146 544–554. Gleave ME 2007 Cooperative interactions between androgen (https://doi.org/10.1016/j.cell.2011.07.006) receptor (AR) and heat-shock protein 27 facilitate AR transcriptional Wang L, Lonard DM & O’Malley BW 2016 The role of steroid receptor activity. Cancer Research 67 10455–10465. (https://doi. coactivators in hormone dependent cancers and their potential as org/10.1158/0008-5472.CAN-07-2057)

Received in final form 17 June 2019 Accepted 26 June 2019 Accepted Preprint published online 26 June 2019

https://erc.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/ERC-19-0132 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/06/2021 07:02:00AM via free access