Hannelore V. Heemers and Donald J. Tindall*


Running title : Functionally diverse AR coregulators


Acknowledgements : This work was supported by NIH grants CA121277, CA91956, CA15083, CA125747, DK65236 and the T.J. Martell Foundation.


Disclosure summary : H.V.H. has nothing to declare. D.J.T. comsults for GlaxoSmithKline, Inc. and received lecture fees from GTx, Inc. and Takeda Pharma. Co.


NIH statement : This is an un-copyedited author manuscript copyrighted by the Endocrine Society. This may not be duplicated or reproduced, other than for personal use or within the rule of “Fair Use of Copyrighted Materials” (section 107, Title 17, U.S. Code) without permission of the copyright owner, The Endocrine Society. From the time of acceptance following peer review, the full text of this manuscript is made freely available by The Endocrine Society at http://www.endoiournals.org/. The final copy edited article can be found at http:/www.endojournals.org/. The Endocrine Society disclaims any responsibility or liability for errors or omissions in this version of the manuscript or in any version derived from it by the National Institutes of Health or other parties. The citation of this article must include the following information:author(s), article title, journal title, year of publication and DOI.


Dr. Donald J. Tindall, Departments of Urology Research/Biochemistry and Molecular Biology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905. Phone : 507-284 8139 Fax : 507-284-2384 ; E-mail: tindall.donald@mayo.edu.


Abstract

Androgens, acting through the androgen receptor (AR), are responsible for the development of the male phenotype during embryogenesis, the achievement of sexual maturation at puberty and the maintenance of male reproductive function and behaviour in adulthood. In addition, androgens affect a wide variety of non-reproductive tissues. Moreover, aberrant androgen action plays a critical role in multiple pathologies, including prostate cancer and androgen insensitivity syndromes. The formation of a productive AR transcriptional complex requires the functional and structural interaction of the AR with its coregulators. In the last decade, an overwhelming and ever increasing number of proteins have been proposed to possess AR coactivating or corepressing characteristics. Intriguingly, a vast diversity of functions has been ascribed to these proteins, indicating that a multitude of cellular functions and signals converge on the AR to regulate its function. The current review aims to provide an overview of the AR coregulator proteins identified to date and to propose a classification of these AR coregulator proteins according to the function(s) ascribed to them. Taken together, this approach will increase our understanding of the cellular pathways that converge on the AR to ensure an appropriate transcriptional response to androgens.


Overview

  • I. Introduction
  • II. The androgen receptor (AR)
  • III. AR-interacting proteins
  • 1. General transcription factors
  • 2. AR coregulators
  • 2a. Components of the chromatin remodelling complex
  • 2b. Histone modifiers : acetyltransferases and deacetylases
  • 2c. Histone modifiers : methyltransferases and demethylases
  • 2d. Components of the ubiquitination/proteasome pathway
  • 2e. Components of the SUMOylation pathway
  • 2f. Proteins involved in splicing and RNA metabolism
  • 2g. Proteins involved in DNA repair
  • 2h. Chaperones and co-chaperones
  • 2i. Cytoskeletal proteins
  • 2j. Proteins involved in endocytosis
  • 2k. Signal integrators and transducers, scaffolds and adaptors
  • 2l. Cell cycle regulators
  • 2m. Regulators of apoptosis
  • 2n. Viral oncoproteins
  • 2o. Other, functionally diverse proteins
  • 3. Specific transcription factors
  • IV. Implications and significance of the convergence of a multitude of diverse functions on the AR
  • a. Accurate transcriptional output by the AR requires the concerted action of numerous cellular pathways and processes
  • b. Assembly of the AR transcriptional complex
  • c. Accomodation of coregulators by the AR
  • V. AR coregulators in (patho)physiology
  • VI. Conclusions and future directions

Key words : androgens, coactivator, corepressor, transcription.



Introduction


Androgens, which are the main male sex steroids, are responsible for the development of the male phenotype during embryogenesis and for male sexual maturation at puberty. In adulthood, androgens remain essential for the maintenance of male reproductive function and behaviour. In addition to their effects on reproduction, androgens affect a wide variety of non-reproductive tissues including skin, bone, muscle and adipose tissues. As a consequence, deregulations in the production or action of androgens can affect different organ systems with a variable degree of severity. This is reflected in pathologies ranging from androgen insensitivity syndromes and prostate cancer, to an increased risk and susceptibility to gender-related diseases such as hepatocellular carcinomas, to relatively mild conditions as acne and male pattern alopecia (1-6).


Testosterone, the principal androgen in the male circulation, is synthesized by the testes. The remaining androgens in the bloodstream (~5-10%, including dehydroepiandrosterone (DHEA), androstenediol and androstenedione) are either produced by the adrenal cortex and can be converted into testosterone in peripheral tissues or are derived from peripheral conversion from testosterone (dihydrotestosterone, DHT) (7-8). Synthesis of androgens is tightly regulated by the hypothalamic-pituitary-gonadal axis. Pulsatile secretion of luteinizing hormone (LH)- releasing hormone (LHRH) by the hypothalamus stimulates secretion of LH by the anterior pituitary, which in turn induces production of testosterone by the testicular Leydig cells. Testosterone acts through a negative feedback loop to prevent LHRH release by the hypothalamus and to decrease the sensitivity of the pituitary to LHRH. The majority of circulating testosterone is bound to the carrier proteins: sex hormone-binding globulin (SHBG) or albumin. Therefore, only 1-2% of testosterone exists in an unbound, free form. The major androgens in women include DHEA sulphate, DHEA, androstenedione, testosterone and DHT. Androgen biosynthesis occurs both in the adrenal and in the ovary, and is regulated by adrenocorticotropic hormone (ACTH) (adrenal synthesis) and LH (ovarian synthesis) (9,10).


Upon transportation by the blood to its target tissues, unbound, lipophilic testosterone diffuses into its target cell where it can be rapidly and irreversibly converted into its more potent metabolite DHT by action of 5α- reductase in some but not all target cells (type I or II, depending on the target tissue) (11). Both testosterone and DHT (either locally produced or from the circulation) exert their activities by binding to a cognate receptor, the androgen receptor (AR), a 110 kDa member of the nuclear receptor superfamily of ligand- activated transcription factors. DHT binds the AR with higher affinity, and its biological activity exceeds that of testosterone up to 10 times (12). DHT dissociates from the AR more slowly than testosterone, and AR bound to DHT is more stable, persisting in cells for greater lengths of time (13). In its basal, unliganded state, the AR resides primarily in the cytoplasmic compartment where it exists in a complex with heat shock proteins (Hsps) and immunophilin chaperones such as Hsp70, 90, 56 and p23. Upon ligand binding, alterations occur in the composition of this Hsp complex and the AR undergoes a conformational change, allowing nuclear translocation of the AR and AR homodimer formation (14). Inside the nucleus, the activated AR binds to specific recognition sequences known as androgen reponse elements (AREs) in the promoter and enhancer regions of target genes. The ARE-bound AR dimer can either interact directly with components of the transcription pre-initiation complex or recruit other components that promote such a functional interaction (15-17). Recruited elements may be other transcription factors binding to recognition elements in the vicinity of AREs and forming more complex androgen response units, or they can be coregulator proteins. As a general definition, AR coregulators are proteins that are recruited by the AR and either enhance (i.e. coactivators) or reduce (i.e. corepressors) its transactivation, but do not significantly alter the basal transcription rate and do not typically possess DNA binding ability. Instead, coregulators influence AR-mediated transcription by acting at the target gene promoter region to facilitate DNA occupancy, chromatin remodelling and/or recruitment of general transcription factors associated with RNA polymerase II, or by assuring the competency of the AR to directly enhance gene expression. The latter can be achieved by modulation of the proper folding of the AR, ensuring its stability or correct subcellular localization (16-17). In the last decade, an overwhelming and ever increasing number of proteins have been identified as AR coregulators. In the current review, we provide an overview of the AR coregulators that have been identified to date. In view of the remarkable functional diversity displayed by these proteins and the vast number of cellular pathways with which they are involved, we propose a classification of AR coregulatory proteins according to their intrinsic primary function. Finally, we discuss the importance of these factors in the regulation of tissue-selective androgen- dependent gene expression under physiological and pathological conditions.



The androgen receptor


Like other members of the nuclear receptor superfamily, the AR is characterized by a modular structure consisting of 4 functional domains : an N-terminal domain (NTD), a DNA-binding domain (DBD), a hinge region and a ligand-binding domain (LBD) (18,19). The AR NTD is relatively long and displays the most sequence variability among nuclear receptors. It is very flexible and displays a high degree of intrinsic disorder, which has hampered studies into its three-dimensional structure (20). The AR NTD contains the major transactivation function (AF) of the AR, termed AF-1. When separated from the LBD, AF-1 gives rise to a constitutively active AR. Two motifs in the AR NTD, 23-FQNLF-27 and to a lesser extent 433-WHTLF-473 have been shown to interact with the LBD, resulting in an NH2-COOH terminal intra- and/or intermolecular AR interaction that has been proposed to be important for the transcriptional activation of some, but not all, AR target genes (21). AF-1 is highly modular and consists of 2 transactivation units (TAUs), TAU 1 and TAU 5, which participate in transcriptional activation (22). The AF-1 domain undergoes induced folding when contacted by basal transcription factors such as TFIIF, resulting in a more compact and active conformation that enables further co-regulator recruitment and transcription (20). In addition, the NTD harbors a variable number of homopolymeric repeats, the most important of which is a polyglutamine repeat that ranges from 8 to 31 repeats in normal individuals, with an average length of 20. Expansion of the glutamine repeats up to 40 residues or more results in spinal and bulbar muscular athrophy (SBMA or Kennedy’s disease), an X- linked pathology characterized by neurological features and late onset symptoms of mild androgen insensitivity (1,23,24). Shortening of the polyglutamine stretch on the other hand, gives rise to a more transcriptionally active AR, which has been suggested to be associated with a predisposition to prostatic neoplasia (25).


The centrally located DBD is the most conserved region within the nuclear receptor family. This region harbors 9 cysteine residues, of which 8 are involved in forming 2 zinc fingers, and a C-terminal extension. The first zinc finger, most proximal to the NTD, determines the specificity of DNA recognition, while residues in the second zinc finger are involved in AR dimerization. Two AR monomers in a head-to head conformation bind as a homodimer to AREs (26), which are direct or indirect repeats of the core 5’- TGTTCT-3’, or more complex response elements harboring diverse arrangements of AREs (27,28). The C-terminal extension is important for the overall three-dimensional structure of the DBD and plays a role in mediating the AR selectivity of DNA interaction (27).


The hinge region has long been considered to be a flexible linker between the DBD and LBD. More recently, however, this region was shown to be involved in DNA binding as well as AR dimerization and was suggested to attenuate transcriptional activity of the AR (29,30). Moreover, a ligand-dependent bipartite nuclear localization signal (NLS) is located in the carboxyterminal part of the DBD and the hinge region, implicating the hinge region in AR nuclear translocation (31,32).


X-ray crystallographic studies indicate that the AR LBD structure is similar to that of the other members of the nuclear receptor superfamily (33-36). The LBD in nuclear receptors consists of 12 discrete α-helices. Insertion of the agonist into the AR ligand-binding pocket has been suggested to change the conformation of the LBD in such a way that helix-12 is stabilized. This leads to the formation of a shallow hydrophobic groove at the top of the ligand binding pocket, generally referred to as AF-2. AF-2 is the major protein-protein interaction surface used by nuclear receptors to recruit LXXLL-motif containing coactivators (37). The AR, however, differs from other nuclear receptors in this respect and interacts with coactivators in a unique manner. The hydrophobic pocket in the AR-LBD binds preferentially to FXXLF motifs, including the 23-FQNLF-27 found in its NTD and interacts poorly with LXXLL motifs commonly found in coactivators (38-42). Consequently, the hydrophobic pocket within the AR LBD faciltates intramolecular and intermolecular interaction between the AR NTD and its C-terminus and is apparently not readily available for coactivator binding. Recent data suggest that the AR N/C terminal interactions occur predominantly when the AR is not bound to DNA (43). Interestingly, several AR-associated coactivators that contain FXXLF motifs have been isolated (44), suggesting that competition exists between these regulatory proteins and the NTD for binding to the AF-2. The implications of such competition and the association of NTD and LBD is not clear, but suggest that additional surfaces outside this well-defined coactivator pocket enable the AR to interact with its coactivators and that different classes of coactivators may interact with different AR surfaces. These observations explain why the AF-2 in the AR LBD displays relatively weak ligand- dependent transactivating properties when compared to the AF2 of other nuclear receptors. Nonetheless, mutation or deletion of AF-2 markedly reduces transcriptional activation in response to ligand. Apart from forming the ligand binding pocket, the AR LBD mediates interaction between the AR and Hsps (14).


AR-interacting proteins


In general, proteins that interact with the AR can be divided into 3 general classes : 1) components of the general transcriptional machinery, 2) functionally diverse proteins with AR coactivating or corepressing properties and 3) specific transcription factors. AR coregulators differ from general and specific transcription factors in that they do not affect the basal rate of transcription and typically do not bind to DNA.


1. General transcription factors.

As is the case for other transcription factors, enhanced transcription by the AR depends on the recruitment of RNA polymerase II to promoters of its target genes. This is achieved by the assembly of general transcription factors that make up the preinitiation complex (PIC). A detailed description of transcription initiation is beyond the scope of this manuscript and has been reviewed previously (45). Briefly, formation of the PIC is accomplished by binding of TFIID, which is composed of TATA-binding protein (TBP) and TBP-associated factors (TAFs), in the proximity of the transcriptional start site. TFIIB then binds TBP and recruits RNA polymerase II and TFIIF, which ensures specific interaction of RNA polymerase II at the promoter. TFIIE and TFIIH are recruited to RNA polymerase II to facilitate strand separation, which allows transcription initiation. Although many AR-associated coregulators facilitate and mediate communication between the AR and the general transcriptional machinery, the AR has also been shown to interact directly with components of the basal transcriptional machinery. For instance, the AR NTD interacts with RAP74, a large subunit of TFIIF. Binding of RAP74 induces α-helical structure in AF-1 and facilitates interaction between AR and the p160 coactivator SRC-1 (20,46-50). Modest binding between the RAP30 subunit of TFIIF and TBP has also been described (46). Moreover, AR has been shown to interact with TFIIH, and overexpression of the cdk-activating kinase (CAK) subunit of TFIIH markedly stimulates AR-mediated transcription (50). AR interaction with TFIIH may enhance phosphorylation of the RNA polymerase COOH terminal domain (CTD), an event necessary to transition from transcription initiation to transcriptional elongation, suggesting that AR may increase the efficiency of transcriptional elongation of AR target genes. Consistent with this hypothesis, an interaction between AR and positive transcription elongation factor b (p-TEFb) has been described (51). The small subunit of p- TEFb, PITALRE (also known as cdk9), harbors protein kinase activity that is able to phosphorylate the CTD of the largest subunit of RNA polymerase II, which is necessary to progress from PIC formation on the promoter to transcriptional elongation. Remarkably, both TFIIH and p-TEFb possess CTD kinase activity, but these activities act at different stages of transcription (15). In addition to its contacts with several general transcription factors, AR also interacts directly with RNA polymerase II through association with its subunit RPB2. Co-expression of RPB2, which is involved in transcriptional elongation, stimulates AR-mediated transcription of target genes (52). Interaction of the AR with and regulation of AR-mediated gene expression by other subunits of RNA polymerase II has not been observed. Taken together, these findings indicate that AR may regulate transcription of target genes by regulating both transcriptional initiation and elongation events.


2. AR coregulators

More than 200 nuclear receptor coregulators have been identified since the identification of the first nuclear receptor coactivator, steroid receptor coactivator 1 (SRC-1), in 1995 (53). By mining peer-reviewed literature accessible through PubMed (www.pubmed.gov) and consulting specialized websites dedicated to AR function (the androgen receptor gene mutation database, androgendb.mcgill.ca and the Nuclear Receptor Signaling Atlas (NURSA) database, www.nursa.org), we have attempted to provide an up-to-date overview of proteins that have been listed as putative coregulators for the AR. As of May 2007, the list of proteins that have been classified as potential AR coregulators contains 169 members. Remarkably, these coregulators display a diverse array of functions and are involved in multiple cellular pathways. There are many ways one could categorize or group these proteins. We have chosen to arrange these proteins according to their apparent primary function, i.e. the function for which they are best recognized, even if, in some cases, this particular activity may not be critical for their effect on AR action. Overall, we feel that such a classification, rather than by their function as a coactivator or a corepressor will provide a broader picture of the cellular events that converge on and regulate the transactivation properties of the AR. As will be discussed below, this does not preclude the possibility that some multifunctional AR-coregulators can be assigned to multiple categories. For reasons of simplicity and to keep our overview comprehensive, we chose to not include information on the specific cell systems or specific target genes used to ascertain the coregulator properties of the AR cofactors in this section. This information will be addressed in a subsequent section.


a. Components of the chromatin remodeling complex

Transcription occurs on a chromatin template, in which DNA is wound around a core of 4 basic histone proteins (H2A, H2B, H3 and H4) to form nucleosomes. DNA-histone interactions limit the accessibility of the nucleosomal DNA to transcription factors and form a major obstacle to transcription. Chromatin remodelling complexes alter and unwrap the histone-DNA contacts in an ATP- dependent manner catalyzed by ATPases, leading to reorganization of the nucleosomal structure and eventually to a chromatin status that is more permissive to transcription (54,55). Several AR coregulator proteins have been identified as components of the chromatin remodelling complex. One of the first indications that components of the chromatin remodelling complex may play a role in AR-mediated transcription came from the identification of ARIP4, a nuclear ATPase that belongs to the SNF2-like family of chromatin remodelling proteins. ARIP4 interacts with the AR zinc-finger region and stimulates AR-dependent transactivation in cotransfection experiments (56). Alhough ARIP4 displays DNA dependent ATPase activity, its specific activity was subsequently shown to be considerably lower than that of SNF2-family members, suggesting that it may not be a classical chromatin remodelling protein. Nonetheless, ARIP4 mutants that do not possess ATPase activity behave as dominant negative regulators of AR function (56,57). Subsequently, the ATPases BRG1 and hBRM, two core components required for nucleosome repositioning by the SWI/SNF chromatin remodeling complex, were shown to potently stimulate AR activity (58). Depending on the gene context, however, different requirements for these ATPases were noted, with some genes relying solely on hBRM and others relying on both hBRM and Brg1 for androgen-regulation (58). The BAF57 subunit, an accessory component of this remodelling complex, is also required for AR-dependent transactivation (59). The AR coactivation function of BAF57 depends on SWI/SNF ATPase activity and cooperates with other classes of coactivators. BAF57 directly binds to the AR and is recruited to AR target genes upon ligand stimulation (59). Interestingly, the SWI3-related gene product (SRG3/BAF155), another component of the SWI/SNF complex, is also able to enhance transactivation by the AR. SRG3 interacts with the AR DBD-hinge region and exists in a complex with the AR on promoters of AR targets genes (60). SRG3 appears to initiate at least some of its coactivation properties by enlisting the SRC-1 co-activator. Remarkably, SRG3 function may not be entirely dependent on the presence of BRG1 or hBRM (60). Furthermore, the SNF2-related CBP activator protein (SRCAP) is able to coactivate transcription by the AR (61). Direct interaction of SRCAP with the AR, however, has not been reported. Similarly, hOsa1 (BAF250) and hOsa2, the largest subunits of the SWI/SNF complex, stimulate transcription by the AR, but association of these proteins with AR has not been demonstrated (62).


Overall, the recruitment of these chromatin remodeling proteins to the AR transcriptional complex is consistent with the altered DNA topology and the loss of canonical nucleosomal ladder that is observed at AR target genes following exposure to androgens (63).


b. Histone modifiers : acetyltransferases and deacytelases

In addition to chromatin remodelling, which represents a higher order level of chromatin reorganization and involves repositioning of components of the nucleosome structure, modification of histone residues can affect transcription efficiency and provide a more localized control over transcriptional events in chromatin (54,55). Modifications such as acetylation, methylation, phosphorylation, ubiquitination, ADP-ribosylation and glycosylation of histone residues have been described. In most cases, modification of a histone residue changes the net charge of the nucleosome, which results in loosening or tightening of the DNA-histone interactions. For example, acetylation of a histone lysine residue attenuates its posititive charge, and abrogates its interaction with the negatively charged DNA. Some of these histone modifications are associated with transcriptional activation (e.g. acetylation), others are indicative of both active or repressed genes (e.g. methylation). Also, the position of the modified histone residue can affect the activation status of a gene. The concept that a combination of such marks affect binding of transcriptionally effector proteins lies at the basis of the histone code (54,55).


In keeping with this notion, several dynamic changes in the covalent histone modification status have been associated with androgen- stimulated transcription. These include activating modifications at histone 3 such as acetylation of lysine 9 and lysine 14, dimethylation of arginine 17, phosphorylation of serine 10 and di- as well as trimethylation of lysine 4 (64). In addition, removal of repressive marks has been described, such as demethylation of mono-, di- and trimethyl marks at lysine 9 of histone 3, and decrease in the dimethylation status of lysine 20 at histone 4 (65-68). The significance of these modifications will be discussed in more detail in the sections of the text describing enzymes executing these events. A summary of the histone modifications associated with androgen action can be found in Fig.1.


Recruitment of histone acetylase (HAT) activity to chromatin is associated with transcriptional activation. Conversely, deacetylation of these marks by histone deacetylase activity (HDAC) renders the chromatin environment transcriptionally repressed. Several HATs and HDACs have been shown interact with the AR and modulate its transactivating properties.


For example, two members of the p160 family of steroid receptor coactivator (SRC) gene family, SRC-1 and SRC-3 (p300/CBP interacting protein (p/CIP), receptor-associated coactivator-3 (RAC3), acetyltransferase (ACTR), amplified in breast cancer-1 (AIB1) or thyroid hormone receptor activator molecule-1 (TRAM1)) have been reported to possess HAT activity. Similar to SRC-2 (also known as glucocorticoid receptor-interacting protein-1 (GRIP1) or transcriptional intermediary factor-2 (TIF2)), the third member of this family that does not possess HAT activity, SRC-1 and -3 interact directly and ligand-dependently with the AR to enhance AR-mediated transcription (16, 53, 69-77). Although the LXXLL motif containing p160 family members have been shown to interact with AR AF-2, they interact primarily with the AR N-terminus and possibly the DBD. Moreover, all 3 SRC family members function as scaffold proteins that attract additional coactivator proteins including factors with histone modifying potential (78). Indeed, SRCs have been shown to recruit p300, the p300 homologue CREB- binding protein (CBP) as well as p300/CBP- associated factor (P/CAF), all coactivator proteins that possess HAT functions that are intrinsically stronger than those found in SRC- 1 and -3. Moreover, in vitro experiments have demonstrated a direct, SRC-independent interaction between p300, CBP and P/CAF and the AR (79). The potentiation of ligand- induced AR transactivation by these three coactivators (79-81) relies on the presence of a functional HAT domain. In addition to their effects on histones, CBP, p300 and P/CAF can acetylate proteins such as transcription factors and coregulators. Noteworthy, p300 as well as P/CAF acetylate the AR at three lysine residues in its DBD-hinge region (79). Point mutations in these AR acetylation sites selectively prevent androgen-induction of androgen-responsive genes, hampers coactivation of the AR by SRC-1, p300, Tip 60 and Ubc9, and results in a 10-fold increase in the binding of the co-repressor NCoR (81). It should, however, be noted that the lysine resisues that are acetylated by p300 and p/CAF are part of the AR NLS, and that mutations of these sites may therefore be expected to disrupt AR activity regardless of acetylation events at these sites. Nonetheless, histone acetylation by p300 and CBP facilitates recruitment of the SWI/SNF and Mediator coactivator complexes (63). Furthermore, CBP and p300 function as a direct bridge between DNA-bound AR and the basal transcriptional machinery (79). They may also serve as a scaffold interacting with and assembling a number of other transcriptional regulators (79).


Another coregulator that harbors HAT activity is Tat interactive protein 60 kDa (Tip60). Tip 60 interacts with the AR LBD and enhances AR-mediated transcription by acetylating histones as well as the AR. Acetylation of AR lysine residues in the AR hinge region by Tip60 is a requisite for Tip60-mediated coactivation of the AR. Remarkably, action of Tip60 on AR transactivation is counteracted by HDAC-1 (82-84).


Human origin recognition complex interacting protein (HBO1) is another HAT protein that ligand-dependently associates with the AR DBD-LBD region. Contrary to the HAT protein listed above, HBO1 acts as an AR corepressor, inhibiting hormone-dependent AR activation (85). The direct involvement of HBO1’s HAT function in these events is not clear, as HBO1 has been reported to harbor a relatively weak HAT function. Moreover, histone acetylation by HBO1 has not been demonstrated. Therefore, HBO1 has been proposed to acetylate histones as part of a multisubunit complex (86).


The importance of acetylation and deacetylation of histone and non-histone proteins in AR-mediated transcription is further emphasized by the functional antagonism between the NAD-dependent HDAC SIRT1 and the AR at sites of p300 acetylation. SIRT1 (Sir2, a class III HDAC) represses androgen-induced AR signaling, by a mechanism that involves direct binding to the AR hinge and requires both the NAD- dependent catalytic function of SIRT1 and deacetylation of the lysine residues 630/632/633 in the AR hinge region that are targeted by p300 and P/CAF (87). Moreover, SIRT1 inhibits p300-mediated interaction between the AR N- and C-terminus. On the other hand, ligand-induced repression of AR function by the class II HDAC HDAC-7 is independent of these acetylation sites. Moreover, the deacetylase activity of HDAC7 is at least partly dispensable in the repression of AR function (88). In addition to SIRT1, several HDACs of class I AND II negatively affect AR transactivation. Contrary to SIRT1 and HDAC-7 that can interact directly with the AR, recruitment of HDACs to the AR transcriptional complex is usually indirect through association with multisubunit corepressor complexes such NCoR and SMART or as binding partners for other AR corepressors (17).


Reflecting the importance of the action of HATs and HDAC enzymes at genomic sites mediating AR transcriptional activity, the acetylation status of histone 3 and 4 residues is often assessed as a marker for the transcriptional activation status of a particular AR target gene.


c. Histone modifiers : methyltransferases and demethylases

While histone acetylation is generally associated with active gene transcription, histone methylation can be indicative of both the active and repressed transcriptional states of the chromatin (54,55). The position of the histone residue affected by this modification is important to make this distinction. For example, methylation of lysine 4 on histone 3, arginines 2, 17 and 26 on histone 3 and arginine 3 on histone 4 is associated with active genes, whereas methylation of lysine 9 on histone 4 is predominantly associated with repressed genes. Moreover, lysine residues can be monomethylated, dimethylated or trimethylated, and the extent of the methylation serves as an important indication of its transcriptional status. Although methylation has long been considered to be an irreversible epigenetic mark, recently, demethylases that mediate active demethylation of repressive histone segments have been identified (54,55).


AR-dependent transcription relies on both methyltransferase and demethylase activities. Coactivator-associated arginine methyltransferase 1 (CARM1 or protein arginine methyltransferase (PRMT)-5), a histone methyltransferase acting at H3R17, was identified initially by its ability to interact with SRC co-activators. Due to its indirect recruitment to ligand-bound nuclear receptors including the AR, CARM-1 has been classified as a secondary coactivator (89). Androgen stimulation leads to recruitment of CARM-1 to androgen responsive enhancers (90). CARM-1 stimulation of AR transactivation depends entirely on the presence of SRC family members. Moreover, the presence of CARM-1 enhances AR transactivation by p160 family members. Loss of CARM-1 reduces transcription of androgen- responsive genes. Mutation of its S-adenosyl methionine binding site abrogates its methyltransferase activity and prevents CARM-1 from exerting its coactivator function. In addition to its effects on histones, CARM-1 methylates proteins in the transcriptional complex including CBP/p300 and several RNA binding proteins (89,90). Noteworthy, CARM-1 may also be recruited to the AR transcriptional complex through p44 (MEP50), a component of the methylosome. p44 interacts directly with the AR and CARM- 1, is found on promoters of AR target genes upon androgen stimulation, and stimulates the transcription of some AR-target genes. In addition, p44 and CARM-1 synergistically cooperate to enhance transactivation by the AR (91). Similar to CARM-1, PRMT1 is recruited to the AR transcriptional complex and stimulates AR dependent gene expression via SRC proteins (92). PRMT1 however preferentially methylates H4R3, which facilitates subsequent acetylation of histone 4 tails by p300. Remarkably, acetylation of histone 4 inhibits its methylation by PRMT1. Like CARM-1, PRMT1 depends on an intact S-adenosyl methionine binding site to fulfil its cofactor function.


Methylation of lysine-9 on histone-3 by the methyltransferase G9a is predominantly associated with repression of transcription. However, for some nuclear receptors including the AR, G9a functions as a coactivator, although weakly (93). Nonetheless, G9a cooperates synergistically with TIF-2, CARM-1 and p300 in activating transcription by the AR. This synergy is strongly dependent on the arginine-specific protein methyltransferase activity of CARM1, suggesting a link between histone arginine and lysine methylation in AR- mediated transcription. Noteworthy, PRMT methyltransferases can not substitute for CARM-1 in this respect. On the other hand, cooperation between G9a, CARM-1 and SRC-2 does not absolutely require the enzymatic activity of G9a. The dependency of G9a on SRC-2 indicates that SRC-2 may function as a scaffold to recruit G9a. G9a associates with regulatory regions in AR target genes in the presence as well as the absence of androgens, and loss of G9a expression hampers androgen- induced AR-dependent stimulation of target genes (93).


Recently, histone methyltransferase activity with a specificity for H3-K36 and H4-K20 has been attributed to nuclear receptor-binding SET domain-containing protein-1 (NSD1, also known as AR-associated (ARA) protein 267α) (94). Based on its ability to interact with the AR DBD-LBD region, to stimulate AR transactivation in an androgen-dependent manner, and to cooperate with other AR coregulators, ARA267 has been classified as an AR-associated coactivator (95). While this possibility remains to be proven, it is tempting to speculate that the AR coactivating properties of ARA267 are mediated by its histone methyltransferase moiety.


The concept that demethylation of histones is involved in transcriptional activation by the AR has emerged only recently. Interest was triggered by the observation that lysine- specific demethylase 1 (LSD1), which specifically demethylates monomethylated and dimethylated H3K9, interacts with the AR (NTD, DBD as well as LBD) and stimulates androgen-receptor-dependent transcription (65). Down-regulation of LSD1 expression abrogates androgen-induced transcriptional activation. Chromatin immunoprecipitation (ChIP) analysis demonstrated that AR and LSD1 form chromatin-associated complexes in a ligand-dependent manner. Androgen exposure leads to a robust decrease in mono,- di- as well as trimethyl H3K9 marks at the promoter of AR target genes. siRNA- mediated loss of LSD1 prevents ligand- induced changes in mono- and dimethyl- but does not affect trimethyl-H3K9 (65). Similar to LSD1, JHDM2A, which demethylates mono- and dimethylated-H3K9, interacts directly with the AR and coactivates androgen-mediated transcription (66). Contrary to LSD1, which is constitutively present at AR target genes, JHDM2A exhibits hormone-dependent recruitment.


Overexpression of JHDM2A greatly reduces the H3K9 methylation level. A knockdown of JHDM2A expression results in increased dimethyl-K9 levels at the promoter region of AR target genes concomitant with a decrease in their expression. Loss of LSD1 does not affect the binding of AR to promoter regions of target genes, nor the ligand-induced recuitment of JHDM2A, but does partially impair hormone induced reduction of dimethyl H3K9 (66). In addition to LSD1 and JHDM2A, a third demethylase, JMJD2C, interacts with and functions as a coactivator for the AR (67). Interestingly, JMJD2C is a histone tridemethylase able to remove mono-, di- and trimethyl marks from H3K9. Like LSD1, JMJD2C is constitutively present at promoter regions of AR target genes. Both JMJD2C and LSD1 interact with and stimulate AR dependent gene transcription in a cooperative manner. Upon androgen treatment, AR, LSD1 and JMJD2C assemble on chromatin, resulting in demethylation of mono-, di- and trimethyl-H3K9 and stimulation of AR-dependent transcription. Conversely, knockdown of JMJD2C inhibits androgen-induced removal of trimethyl H3K9 and transcriptional activation by the AR (67). Taken together, these observations indicate that androgen-dependent gene transcription requires the assembly and coordinate action of methyl transferases and demethylases with distinct substrate specificities.


d. Components of the ubiquitination/proteasome pathway

Ubiquitination is a reversible posttranslational modification of cellular proteins, in which a 76-amino acid polypeptide, ubiquitin, is attached to lysines in target proteins. Ubiquitination of a substrate involves the action of an activating E1 enzyme that transfers ubiquitin to a conjugating E2 enzyme, which in turn enlists an E3 ligase to deliver the ubiquitin tag to the intended target protein. Target proteins can be either poly- ubiquitinated or mono-ubiquitinated. The former usually serves as a signal for degradation of the substrate protein by the 26S proteasome, whereas the latter tends to function as a signal that regulates protein stability, protein-protein recognition, activity and intracellular localization. Both modes of ubiquitination play vital roles in transcriptional regulation as they allow proper progression through rounds of transcription, appropriate assembly of the necessary protein complexes, and modulate the activation status of transcription factors and coregulators (96,97). Not surprisingly, several AR coregulator proteins function in the ubiquitin-proteasome pathway. Most of these coregulators demonstrate E3 ligase activity ; this is the case for instance for E6-AP, Mdm2, PIRH2, SNURF/RNF4 and ChIP. The E3 ligase E6- associated protein (E6-AP) interacts with the AR NTD in a hormone-dependent manner, demonstrates hormone-dependent recruitment to the promoter region of AR target genes and enhances the transactivation function of AR (98). E6-AP may also modulate the protein level of the AR as E6-AP null mice demonstrate increased AR levels in androgen- responsive prostate tissues, and overexpression of E6-AP markedly reduces AR protein expression in cells in culture (98). In addition, Mdm2 interacts with the AR and catalyzes its ubiquitination and proteolysis. Mdm2’s interaction with the AR NTD and DBD is at least in part dependent on its E3 ubiquitin ligase activity (99). Following androgen stimulation, Mdm2 complexes with AR and HDAC1 at active AR target gene promoter promoter and attenuates AR activity. Both AR and HDAC1 are ubiquitinated in response to androgen. HDAC1 and Mdm2 cooperate to reduce AR-mediated transcription, and this functional interaction is attenuated by the HAT activity of the AR co-activator Tip60 (100). However, PIRH2 enhances AR- mediated transcription by reducing HDAC1 protein levels and inhibition of HDAC1- mediated transcriptional repression. PIRH2 is recruited to AREs in AR target genes and is required for optimal expression of these genes. The E3 ubiquitin ligase small nuclear RING finger protein (SNURF/RNF4) was isolated as an AR coregulator based on its ability to interact with the AR NLS in a hormone- dependent manner (102). SNURF does not influence the binding of AR to cognate DNA sequences, but appears to enhance transactivation by the AR by facilitating its import into the cell nucleus (103). SNURF also retards AR nuclear export on hormone withdrawal (104). To date, there is no evidence of SNURF-mediated changes in the ubiquitination pattern of the AR. In addition to its E3 ubiquitin ligase function, C-terminal Hsp-interacting protein (Chip) acts as a molecular chaperone involved in the folding and hormone binding of nuclear receptors. Interaction between Chip and the AR NTD, which occurs in a highly specific and sequence-dependent manner, suggests a functional link between these processes in the regulation of AR activity (105). Chip negatively regulates AR transcriptional activity by promoting AR ubiquitination and degradation (105,106). Noteworthy, these effects of Chip are not fully reversed by proteasome inhibitors, suggesting that mechanisms different from proteasome- mediated degradation are involved. Indeed, Chip overexpression reduces the rate of AR degradation, which is consistent with an effect on AR folding (107). Thus, its effect on AR action might not be entirely dependent on its ubiquitin ligase activity. These findings suggest a role for molecular chaperones.


Other proteins that have been identified as AR coregulators and have been suggested to possess E3 ubiquitin ligase function are ARNIP, ARA54 and MKRN1 (108-111). AR N-terminal-interacting protein (ARNIP) interacts with the AR NTD, although it does not affect AR ligand-binding kinetics, or AR transcriptional activation. However, AR N- terminal-C-terminal interaction is reduced in the presence of ARNIP (108). ARA54 has been isolated based on its ability to ligand- dependently associate with the AR LBD and was subsequently shown to enhance AR- mediated transactivation (109). The ubiquitously expressed Makorin RING zinc finger protein 1 (MKRN1) inhibits the transcriptional activity of the AR (111). Interestingly, disruption of the ubiquitin ligase activity of MKRN1 does not affect its inhibitory transcriptional activity. Whether ARNIP, ARA54 and MKRN1 affect the ubiquitination status of the AR or its associated complex has not been assessed.


In addition to these E3 ubiquitin ligases, proteins with other functions in the ubiquitin/proteasome pathway have been shown to modulate AR-mediated transcription. For instance, the ubiquitin-specific protease USP10 interacts directly with the AR and is part of DNA-bound AR complexes (112). USP10, but not an enzymatically inactive mutant USP10, stimulates AR-dependent transcription. Conversely, loss of USP10 expression impairs responsiveness to androgens. Similarly, the E2 ubiquitin- conjugating enzyme UBCH7 stimulates AR- mediated transactivation in a hormone- dependent manner (113). The ubiquitin conjugation activity of UBCH7 is required for this response. Tumor susceptibility gene 101 (TSG101), an E2-like enzyme deprived of ubiquitin conjugase activity, which has been proposed to act as a dominant-negative inhibitor of polyubiquitination, also modulates AR transactivating potential. TSG101 was originally shown to repress ligand-dependent AR transcriptional activation (114).


Interestingly, in an independent study TSG101 was reported to act as a coactivator for AR- mediated transcription (115). TSG101 enhances monoubiquitination of the androgen receptor in a ligand-dependent manner, and this correlates with enhanced transactivating capacity. A dominant-negative mutant of ubiquitin preventing polyubiquitination also stimulates AR-mediated transcription, which cannot be enhanced by TSG101 (115).


These reports establish multiple effectors of distinct steps in the ubiquitinylation pathway as important regulators of the AR transactivating function. Interestingly, the role of the proteasome in these events does not appear to be restricted solely to degradation of the AR or its associated proteins in the cytoplasm (116, discussed also below). Instead, the proteasome itself may be actively involved in AR-governed transcriptional events in the cell nucleus. This hypothesis is supported by the observation that overexpression of the proteosomal subunit PSMA7 enhances AR transactivation (117). Moreover, inhibition of the proteasome prevents the nuclear translocation of the AR, blocks interaction between the AR and several of its coregulators and ultimately abolishes androgen-induced target gene expression (117). In addition, following androgen treatment, the 19S proteasomal subcomplex is recruited to AR target genes, where its occupancy parallels that of the AR (118).


e. Components of the SUMOylation pathway

The small ubiquitin-related modifier SUMO post-transcriptionally modifies several proteins involved in regulation of transcription and chromatin structure. The machinery responsible for the sumoylation of target genes displays remarkable similarity to that mediating protein ubiquitination, as it consists of E1 activating enzymes, an E2 conjugating enzyme and several E3 ligases. SUMO E1, -2 and -3 enzymes are however distinct from their counterpart enzymes in the ubiquitin/proteasome pathway. Similar to the non-proteasomal roles of ubiquitin, SUMO modification of a protein primarily regulates localization and activity (119). Multiple proteins involved in several aspects of the sumoylation pathway can modulate the AR transcriptosome. First, SUMO homologues have been shown to affect AR-mediated transcription. SUMO-1 decreases, whereas SUMO-2 and -3 enhance AR transcriptional activity (120,121). SUMO-3 interacts with the AR NTD. The positive effect of SUMO-3 on AR-transcriptional activity does not depend on either the sumoylation sites of the AR or the sumoylation function of SUMO-3 (121). Sumoylation of the AR NTD by SUMO-1 is stimulated by androgens, and mutation of the sumoylation sites in the AR NTD increases AR transactivation, indicating that sumoylation serves to attenuate AR function (120). Second, the SUMO E2 conjugating enzyme Ubc9 interacts with the NLS in the AR hinge region and acts as a AR coactivator (122). Interestingly, the effects of Ubc9 on AR activity occur independently of its SUMO- 1 conjugating catalytic activity (122,123). Third, protein inhibitors of activated STAT (PIAS) family members that function as SUMO E3 ligases can either positively or negatively affect transcription by the AR (124-132). PIAS proteins (PIAS-1, -3, -xα or ARIP3, -xβ and -y) bind the AR DBD (126). Although homologuous, PIAS proteins can differ markedly in their ability to sumoylate the AR and/or AR-associated coactivators such as TIF-2 (131). PIAS-y, which acts as a corepressor for the AR, does not rely on its E3 ligase activity to influence AR-mediated transcription (132). Moreover, the decision for a particular PIAS protein to function as a corepressor or coactivator depends on the cell type and the target gene (127). Furthermore, the PIAS-like SUMO E3 ligases Zimp7 and Zimp10 both function as AR coactivators (133-135). Zimp10 was shown to interact with the AR NTD (133). AR sumoylation is enhanced in the presence of Zimp10, and mutation of the AR sumoylation sites abrogates the augmentation of AR activity by Zimp10 (133, 135). Fourth, AR-mediated transcription is markedly enhanced by SENP1, a member of SUMO-specific protease family (136). While AR is a target for SENP1, the ability of SENP1 to enhance AR-dependent transcription is not mediated through desumoylation of AR, but rather through its ability to deconjugate HDAC-1, thereby reducing its deacetylase activity. The repressive effect of HDAC-1 on AR- dependent transcription can be reversed by SENP1 and by deletion of its sumoylation sites. In contrast, SENP2 and SENP3 have only modest effects on AR transactivation (136).


In addition to the ubiquitination/proteasome and sumoylation pathways, activity of the related neddylation pathway appears to be involved in AR-mediated transcription. Ubiquitin-activating enzyme 3 (Uba3), the catalytic subunit of the activating enzyme of the ubiquitin-like NEDD8 (neural precursor cell expressed developmentally down- regulated) conjugation pathway, inhibits transactivation by the AR. The neddylation activity of Uba3 is required for its inhibition of steroid receptor transactivation. Direct interaction between Uba3 and the AR has not been reported (137).


Overall, the identification of numerous proteins with functions in the ubiquitination, SUMOylation as well as NEDDylation pathways as AR coregulators points towards the critical importance of tight regulation of the turnover, stability, degradation and subcellular relocalization of components of the AR transcriptional machinery in androgenregulated gene transcription.


f. Proteins involved in splicing and RNA metabolism

Primary transcripts undergo several modifications before a mature mRNA is generated that can serve as a template for translation. As the steps from transcription to translation are mechanistically and functionally coupled (138,139), it is not surprising that several proteins, which demonstrate AR coregulator characteristics, belong to or share high functional or structural homology to members of the family of RNA processing enzymes. For instance, the pre- mRNA splicing proteins PSF (polypyrimidine tract-binding protein-associated splicing factor), PSP1 (paraspeckle protein 1) and PSP2 interact with AF-1 of the AR (140). p54nrb (p54 nuclear RNA binding protein, NonO), another component of the U1snRNP prespliceasome, interacts with the AR NTD in a ligand-dependent manner and potentiates AF-1 function (140). Since p54nrb and PSF also directly interact with the RNA polymerase II CTD, a molecular link between the AR transcriptional and splicing machinery is apparent (138). Furthermore, p102 U5snRNP has been isolated as a protein interacting with AR AF-1, termed ANT-1 (AR N-terminal domain transactivating protein-1). ANT-1 enhances the ligand-independent AF-1 function of AR but does not affect ligand- dependent AF-2 activity (141,142).


The splicing factor hRNPA1 is recruited to the AR through association with the AR interacting coactivator ARA54 and selectively suppresses ARA54-enhanced AR transactivation via interruption of AR-ARA54 interaction (143). Finally, the previously discussed methyltransferases CARM-1 and p44 (MEP50) are components of the methylosome complex that methylates snRNP complex proteins, suggesting they also fulfill roles in the splicing events.


g. Proteins involved in DNA repair

In a targeted approach to discover components that comprise the AR apo- and holoreceptor complex using tandem mass spectroscopy analysis, the trimeric DNA-dependent protein kinase (DNA-PK) complex was isolated (144). The DNA-PK complex is best-known for its role in DNA repair and has emerged as a part of the transcriptional machinery. The AR- LBD interacts directly with the Ku70 and 80 regulatory subunits of DNA-PK in a DNA-independent manner. Interaction between the AR and the catalytic DNA-PK subunit has not been observed. Ku proteins bind the AR both in the cytoplasm and nucleus. Ku proteins are recruited in an androgen-dependent manner to the promoter of AR target genes. Ku70 and Ku 80 as well as DNA-PK enhance AR activity in transactivation assays. Ku70 and Ku80 have been shown to exert these effects through recycling of transcriptional factors (144). Other proteins with roles in DNA repair and damage control have been demonstrated to modulate AR-mediated transcription. For instance, the checkpoint protein Rad9 acts as a corepressor to suppress androgen-AR transactivation (145). AR interacts with the C terminus of Rad9 via its LBD. The FXXLF motif within the C terminus of Rad9 interrupts the androgen-induced interaction between the N terminus and C terminus of AR. Moreover, the tumor suppressor genes BRCA1 and BRCA2 are AR coactivators (146-148). BRCA2 is an integral component of the homologous recombination machinery, while BRCA1 possesses both E3 ubiquitin ligase activity and DNA repair activity. BRCA1 interacts with the AR NTD and enhances transcription of AR target genes. BRCAl-enhanced AR transactivation can be further induced synergistically with AR coregulators SRCs, CBP, ARA55, and ARA70 (146,147). BRCA2, but not a truncated mutant of BRCA2, synergizes with SRC-2 to enhance transcriptional activation by the AR. BRCA2 associates with the AR NTD and LBD, as well as SRC-2 and further cooperates with P/CAF and BRCA1 to enhance AR- and SRC-2-mediated transactivation (148).


In general, components of the DNA repair machinery are recruited when the transcriptional machinery runs into obstacles or DNA lesions that prevent proper transcription of target genes. The identification of several proteins with functions in DNA repair as coregulators for the AR indicates that this mechanism holds true also for AR-mediated transcription.


h. Chaperones and cochaperones.

In the absence of androgens, the molecular chaperone complex is critical to maintain the AR in a stable, inactive, intermediate configuration that has a high affinity for androgenic ligands. Upon binding of androgens and folding of the AR into an active conformation, selective molecular chaperones remain associated with the AR and are important for downstream events such as AR translocation, AR transcriptional activity, disassembly of the AR transcriptional complex and AR degradation. The sustained importance of these proteins in overall AR activity is reflected in the interactions between the AR and multiple components of chaperone complexes throughout the life cycle of the AR (14). In the early stages of the AR activation process, AR LBD interacts transiently with Hsp40 (Ydj1), Hsp70 (HSc70), Hip, Hps90, Hop and p23, leading to an equilibrium in which the AR is maintained in an overall high- affinity ligand-binding state. In this respect, Hsp40 is necessary for hormone binding to the AR (149). Mutations in Hsp40 result in a reduction of AR-Hsp70 complex formation and defects in AR folding (150). In contrast, loss of the Hsp70 co-chaperone DjA1 in a knock-out mouse model leads to increases in AR protein levels and enhanced transcription of several androgen-responsive genes inSertoli cells, giving rise to severe defects in spermatogenesis (151). In the same study DjA1 was suggested to function as a negative regulator of transactivation by the AR. Hormone binding causes AR to undergo a sequential loss of chaperones (14). With the assistance of Hsp90, the AR is transformed into a DNA-binding competent state. Receptor activation leads to unmasking of the NLS, resulting in Hsp90-dependent translocation of the AR to the nucleus (14). Cdc37 (p50) also functions down-stream of hormone-binding as a Hsp90-associated protein involved in AR trafficking. Mutant forms of cdc37 induce defects in AR transactivation while leaving AR protein levels unaltered (152). Hsp70 and Hsp40 are also believed to re-associate with the AR in the presence of ligand and to facilate transport of the receptor into the nucleus (14). Hsp90 binding co-chaperone FKBP52 (FK506 binding protein of 52kDa) interacts with AR complexes (153). In vivo studies using FKBP52 deficient mice indicate that FKBP52 does not affect hormone binding by the AR or AR nuclear translocation, but is critical for AR transactivation (153,154). FKBP52 enhances AR-mediated transcription, and this effect depends on its ability to interact with Hsp90 (153). FKBP52 may also be necessary to maintain AR protein levels (153). The related co-chaperone FKBP51 forms complexes with AR and stimulates AR transactivation, although these observations could not be confirmed by a second, independent study (154,155). Apart from their effects on AR folding and trafficking, experimental evidence supports a direct involvement of molecular chaperones in the transcriptional activation of AR target genes. Bag-1 Hsp70 co-chaperones, in particular the Bag-1L isoform, bind directly to the tau5 domain in the AR NTD and function as a coactivator for the AR (156,157). Bag-1L relies on its association with Hsp70 to interact with the AR and loss of this interaction domain markedly suppresses its abilitity to stimulate AR-mediated transactivation. Moreover, Bag-1L as well as Hsp70 are recuited with the AR to promoter regions of AR target genes (156). Noteworthy, BAG-1L harbors a ubiquitin-like domain that facilitates association of Bag-1L with the proteasome, enabling BAG-1L to function as a coupling factor between the chaperone and proteolytic complex (158).


This is reminescent of the dual of function of Chip as both an E3 ubiquitin ligase and a co-chaperone. In fact, AR degradation by a Hsp70-Chip governed system has been reported (158). BAG-1 and Chip interact directly and cooperate with each other during the sorting of chaperone substrates to the proteasome. Taken together, these findings suggest an interdependency between AR- mediated transcription, AR degradation and folding events.


i. Cytoskeletal proteins

Actin is a major component of the cytoskeleton. While the cytoplasmic roles of actin and actin organization in the cytoplasm have been well established, the possibility for a role for actin in the nucleus has been controversial. However, it is now generally accepted that actin plays a role in transcriptional events. Actin as well as actin binding proteins have been shown to mediate nuclear translocation of transcriptional regulators. Moreover, actin is found as part of chromatin remodeling complexes and ribonucleoprotein particles, and interacts directly with RNA polymerases (159). Actin binding proteins and actin monomers bind to the AR, indicating that they also play an important role in AR-mediated transcription. For example, supervillin, an actin-binding protein, is able to interact with the AR NTD and DBD-LBD (160). This association is enhanced in the presence of androgens. Supervillin increases AR transactivation and cooperates with other AR coregulators, such as ARA55 or ARA70. Moreover, three different actin isoforms cooperate with supervillin to further stimulate AR transactivation in an additive manner (161). Conversely, an actin chelator that reduces the availability of monomer actin attenuates the coactivator properties of supervillin (161). Supervillin has no effect on cytoplasmic-nuclear translocation of the AR, nor does it affect the half-life of the AR (160). Similar to their effect on supervillin, androgens increase the interaction between the AR DBD-LBD and gelsolin, another actin-binding protein, in a dose- dependent manner. Gelsolin interacts with the AR during nuclear translocation and to enhance ligand-dependent AR activity (162). In addition, the F-actin crosslinking protein filamin, which was originally identified as a protein that facilitates nuclear transport of the AR, interacts with the AR DBD-LBD in a ligand-independent manner. However, this interaction is enhanced in the presence of androgens. The absence of filamin hampers androgen-induced AR transactivation (163). A second, smaller 90-100 kDa (instead of the 280 kDa form) fragment of filamin (termed filamin A) is capable of nuclear translocation and colocalizes with AR to the nucleus (164,165). This naturally occurring filamin fragment represses AR transactivation and disrupts AR interdomain interactions as well as hampers SRC-2-activated AR function. Another cytoskeletal protein, α-actinin-2, enhances the transactivation activity of SRC-2 and serves as a primary coactivator for the AR, acting in synergy with SRC-2 to increase AR transactivation function (166). α-actinin-4 also binds to the AR and exhibits coregulating properties (167). Finally, the actin binding protein transgelin suppresses AR function via interruption of AR-ARA54 heterodimerization, resulting in the cytoplasmic retention of AR and ARA54. Transgelin does not directly interact with the AR but exerts its effects through recruitment to ARA54 (168).


Like actin, microtubules constitute a principal component of the cytoskeleton and have been proposed to play an important role in nuclear receptor function. In the case of AR-mediated transcription, ARA67/PAT1/APPBP which shows homology to kinesin light chain and binds microtubules, interacts with the AR NTD. ARA67/PAT1 functions as a corepressor for the AR. Interruption of AR cytoplasmic-nuclear shuttling may play a major role in ARA67/PAT1 mediated suppression on AR (169).


j. Proteins involved in endocytosis

Endocytosis mediates the selective uptake of specific macromolecules into the cell. The best characterized form of endocytosis is that mediated by the budding of clathrin-coated vesicles from specialized regions of the plasma membrane. Clathrin-coated vesicles fuse with endosomes, and the content of the vesicle is sorted for intracellular transport or recycled to the plasma membrane (170). Endocytosis can also be achieved in a clathrin- independent manner by uptake of molecules in small invaginations of the plasma membrane termed caveolae, that are coated with caveolin (171). Interestingly, multiple proteins involved in both mechanisms of endocytosis have been shown to interact with the AR and to influence AR mediated transcription. One of the adaptor proteins involved in clathrin- mediated uptake, huntingtin interacting protein 1 (HIP1), associates with the AR and reduces the rate of AR protein degradation (172). Moreover, HIP1 is recruited to AREs upon androgen stimulation. Overexpression of HIP1 enhances AR-mediated transcription. Conversely, transcription by the AR is significantly repressed following knockdown of HIP1 expression. Androgen stimulation results in nuclear translocation of HIP1, an event that relies on a nuclear localization signal at the COOH terminus of HIP1 (172). Remarkably, another endocytic protein termed APPL (adapter protein containing PH domain, PTB domain and leucine zipper motif), that translocates to the nucleus upon growth factor stimulation, down-regulates AR-mediated transcription in a dose-dependent manner. Interaction between the AR and APPL is mediated by Akt (173). In addition, HAP1 (huntingtin-associated protein 1), which functions in endocytosis of membrane receptors and endosomal trafficking, interacts with AR through its LBD (174). This interaction is dependent on the length of the AR polyglutamine stretch (stronger with increasing length), and the addition of androgens diminishes the strength of this association. Cyclin G-associated kinase (GAK) or auxilin 2, is an essential cofactor for Hsp70-dependent uncoating of clathrin-coated vesicles. GAK interacts with the AR NTD and enhances the AF-1 function of AR activity in a ligand-dependent manner (175). Caveolin is a principal component of caveolae membranes that serve as a scaffold protein of many signal transduction pathways. Caveolin-1 ligand- dependently interacts with the AR NTD and LBD (176). Overexpression of caveolin-1 significantly increases nuclear localization of the AR and potentiates ligand-dependent AR activation (177). In contrast, down-regulation of caveolin-1 expression diminishes androgen- induced AR-mediated transcription (176).


Overall, these findings support the concept that several proteins involved in different aspects of endocytosis exert AR coregulatory characteristics.


k. Signal integrators and transducers, scaffolds and adaptors.

In line with the AR coregulator properties of endocytic scaffold proteins such as caveolin-1, several proteins involved in cell-cell and cell- substrate adhesion complexes are able to interact with the AR and regulate its activity. This is the case also for a number of proteins that function as scaffolds and adaptors for multiple signal transduction pathways. Among these are LIM domain-containing proteins that can associate with focal adhesions, such as ARA55/Hic, paxillin and FHL2. ARA55 binds to the AR-LBD in a ligand-dependent manner, and relies on 3 LIM domains in its C-terminal half for this interaction. ARA55 enhances transcriptional activity of the AR (178). Interestingly, the focal adhesion kinase (FAK) Pyk2 is able to repress AR transactivation by interacting with and phosphorylating ARA55 (179). The ARA55-related protein paxillin also localizes within focal adhesions and can participate in a number of signal transduction pathways. Paxillin directly interacts with the AR, and overexpression of paxillin results in increased targeting of the AR to the nuclear matrix (180). Paxillin functions as a coactivator for the AR. Moreover, like ARA55, paxillin relies on its COOH-terminal LIM domain to interact with the AR. The four-and-a-half LIM domain protein FHL2, that also functions at focal adhesions as well as in the nucleus, directly associates with full length AR, and stimulates AR-mediated transcription in an agonist- and AF-2-dependent manner (181). FHL2 has been found to interact with PELP1/MNAR (proline-, glutamic acid-, and leucine-rich protein-1/modulator of nongenomic actions of the estrogen receptor), which serves as a scaffolding protein that couples nuclear receptors with various signaling complexes (182). The PELP/MNAR interactome harbors transcriptional regulators, chromatin regulators, splicing factors, cell cycle proteins, cytoskeletal regulators and proteins involved in non-genomic signaling. PELP1/MNAR interacts with the AR and enhances FHL2-mediated AR transactivation function. Conversely, knockdown of PELP1/MNAR reduces FHL2-induced AR transactivation (182). Thus, PELP1 functions as a molecular adaptor, coupling FHL2 with the AR. Another focal adhesion protein, vinexin a, has been identified as a vinculin- binding protein that plays a key role in cell spreading and cytoskeletal organization. The AR binds to vinexin a, and the ligand-induced transactivation function of the AR is stimulated by vinexin a (183).


In addition to focal adhesion components, several proteins involved in G-coupled receptor signaling affect AR-induced transcription and/or interact with the AR. The Rho GTPase guanine nucleotide exchange factor (GEF) Vav3 activates Rho family GTPases by promoting the exchange of GDP for GTP. Vav3 potentiates AR transcriptional activity (184,185). In contrast, knock-down of Vav3 results in decreased AR transactivation. The increase in AR activity by Vav3 involves AF-1 of AR. However, Vav3 does not interact with the AR nor does it increase AR levels. Whether its GEF function is required for the stimulation of AR transactivation is still under debate (184,185). Rho guanine nucleotide dissociation inhibitor (Rho GDI) was originally identified as a negative regulator of the Rho family of GTP binding proteins. Overexpression of Rho GDI increases AR transcriptional activation, suggesting an AR coactivator role (186). Physical interaction between RhoGDI and the AR has not been assessed. Interplay between activated Cdc42- associated tyrosine kinase Ack1 and AR has also been described. Ack1 binds the AR and phosphorylates several tyrosine residues in its NTD (187). Activated Ack1 is recuited to AREs, and promotes induction of AR target gene expression both in the presence and absence of androgen. Moreover, knock-down of Ack1 decreases androgen-stimulated recruitment of the AR to AREs in target genes, suggesting that Ack1 is required for optimal androgen-regulated DNA binding of the AR (187). PRK1/PKN is a member of the protein kinase C superfamily of serine/threonine kinases and is one of the first identified effectors for RhoA GTPases. The AR interacts with PRK1 through the TAU-5 domain (188). Blocking of endogenous PRK signaling severely impairs agonist-dependent AR transactivation. Similar effects are seen for the related PRK2. Conversely, stimulation of the PRK signalling cascade results in a ligand- dependent superactivation of AR. Furthermore, PRK1 promotes a functional complex of AR with the coactivator SRC-2 (188). RanBPM (Ran-binding protein in the microtubule-organizing center) was originally identified by its interaction with the small Ras- like GTPase Ran. RanBPM interacts directly with the AR-NTD and DBD in the presence of ligand and enhances androgen-dependent transcription by the AR (189). RanBPM may also play a role in Ran-dependent nuclear transport. Noteworthy, Ran/ARA24 interacts with the AR NTD and acts as a coactivator for the AR (190), suggesting that RanBPM, Ran and the AR work in a multi-protein complex. The p21-activated kinase (PAK6) that contains a putative amino-terminal Cdc42/Rac interactive binding motif and a carboxyl- terminal kinase domain interacts either with the AR hinge region or LBD (191-193). In contrast to most PAKs, PAK6 kinase activity is not stimulated by Cdc42 or Rac, but can be stimulated by AR binding. In response to androgens, PAK6 cotranslocates into the nucleus with AR and represses AR-mediated transcription. This suppression requires its kinase activity, but does not depend upon GTPase binding to PAK6 and is not mimicked by the closely related PAK1 and PAK4 isoforms. Active PAK6 inhibits nuclear translocation of the stimulated AR, suggesting a possible mechanism for inhibition of AR responsiveness (191-193). Interaction with PAK6 could provide a mechanism for the AR to cross-talk with other signal transduction pathways. In keeping with this concept, the adaptor/scaffolding protein receptor for activated C kinase 1 (RACK1) interacts with the AR through its LBD (194). RACK1 facilitates ligand-independent AR nuclear translocation upon PKC activation and suppresses both ligand-dependent and - independent AR transactivation through PKC activation. ChIP assays reveal a decrease in AR recruitment to AR target genes following stimulation of PKC. These observations support a role for RACK1 as a scaffold for the association and modification of AR by PKC, enabling translocation of AR to the nucleus but rendering AR unable to activate transcription of its target genes (194). An independent study confirmed the repressive effect of RACK1 on androgen-dependent gene expression and showed that androgens can enhance the association between RACK1 and the AR (195). Moreover, RACK1 facilitates the interaction between the AR and Src kinase, which results in increased tyrosine phosphorylation of AR (195). Noteworthy, tyrosine phosphorylation by Src is important for AR nuclear translocation (196).


In line with the concept that adaptors for multiple signal transduction pathways can modulate AR-driven transcription, several effectors of such cascades are able to interact with the AR and positively or negatively alter the transcriptional events it mediates. For instance, signal transducer and activator of transcription-3 (STAT3) can bind the AR and enhance AR transactivation (125, 197-199). STAT3 has been described to stimulate the transcriptional activity of AR in a hormone- dependent manner acting synergistically with SRC-1, pCAF, CBP, and SRC-2 (197). Conversely, DHT-induced AR activity is increased by IL-6, a cytokine activating and signaling through STAT3 (198). In addition, AR’s association with STAT3 enhances the activity of STAT3 (199). AR activation overcomes the inhibitory effect on STAT3- mediated transcription by PIAS3. AR relieves STAT3 from STAT3-PIAS3 complex formation (199). Similarly, Smad3, a key component in the transforming growth factor beta (TGF beta) signaling cascade modulates AR-mediated transcription (200-203). Depending on the experimental setting and the target gene studied, Smad3 can act as an AR coactivator or corepressor. A protein-protein interaction between AR and Smad3 involves the AR DBD-LBD region. Moreover, ligand- bound AR inhibits TGF-beta transcriptional responses through selectively repressing Smad3 signaling (203).


Ligand-enhanced binding of Ebp1, an ErbB-3 binding protein, to the AR NTD suggests a link also between ErbB receptor and AR signalling (204). Indeed, ectopic expression of Ebp1 inhibits ligand-mediated transcriptional activation of AR target genes (204-206). Ebp1 participates in the transcriptional regulation by the AR via its interaction with the corepressors HDAC and Sin3 (207).


Supporting the possibility for cross-talk between Notch and androgen-signaling pathways, Hairy/Enhancer of split related with YRPW motif 1 (Hey1), a member of the basic helix-loop-helix-orange family of transcriptional repressors that mediate Notch signaling, interacts with the AR in a ligand-independent manner (208). Androgen- dependent AR transcriptional activity is inhibited by Heyl and expression of a constitutively active form of Notch represses transactivation by the AR. Noteworthy, Hey2, another member of the Hey family, is also able to repress AR transcription. The inhibiting effects of Heyl on AR transactivation are mediated by AF1 (208). Similarly, AR (through its LBD) and the interferon-activated RNase L interact in a ligand-dependent manner (209). In addition, overexpression of RNase L in the presence of interferon reduces androgen-mediated AR transcriptional activity. Conversely, androgens are able to interfere with induction of gene expression by interferon, indicating a functional cross-talk between DHT and interferon signalling (209). Finally, the Wnt signaling pathway also modulates androgen signaling at multiple levels. Androgens promote the cytoplasmic- nuclear translocation of beta-catenin, a critical component of this signaling pathway. Moreover, beta-catenin is able to bind to the AR LBD in a ligand-dependent manner, is recruited to AREs in AR target genes and enhances transcriptional activity by the AR (210-214). Furthermore, glycogen synthase kinase-3beta (GSK-3beta), a protein serine/threonine kinase that regulates beta- catenin degradation, phosphorylates and interacts with the AR and suppresses its ability to activate transcription (215,216). In contrast, some reports mention a GSK-3beta-mediated increase in AR transactivation (217). Furthermore, TCF4, one of the targets of Wnt signaling that relies on beta-catenin for transcriptional activation, interacts with the AR DBD and functions as a corepressor for the AR (214,218).


l. Cell cycle regulators

Interestingly, several proteins involved in the regulation of cell cycle progression also associate with the AR and modulate its transcriptional activity. For example, cyclin E increases the transactivation activity of the AR in the presence of DHT. Cyclin E binds directly to the NTD of the AR, and enhances its AF-1 transactivation function. Interaction with the AR does not require complex formation with cdk2 nor does it involve phosphorylation of the AR (219). Cdc25B is a cell cycle regulator that functions as a dualspecific phosphatase to mediate cell cycle progression by activating the cyclin-dependent kinases. Cdc25B interacts directly with the AR and stimulates AR-dependent transcription independent of its protein phosphatase activity (220-222). Moreover, p/CAF and CBP interact and synergize with Cdc25B and further enhance its coactivation activity (220). Furthermore, cyclin-dependent kinase 6 (CDK6) binds to and is activated by cyclin D1 and as such enhances the transition of cells through the G1 phase of the cell cycle. CDK6 also associates with the AR and stimulates its transcriptional activity in the presence of DHT. This effect does not require its kinase activity and is inhibited by cyclin D1 and p16INK4a (223). Moreover, CDK6 is present in association with the AR at the promoter region of AR target genes. Contrary to the coactivator properties exhibited by these cell cycle regulators, cyclin D1 functions as a corepressor for the AR (224-229). Ligand-mediated transcriptional activation of AR target genes is inhibited by cyclin D1 as well as cyclin D3 (224). This function of cyclin D1 is independent of its role in cell cycle progression. Cyclin D1 directly binds to the AR in a ligand-independent manner. Cyclin D1 targets the AF-1 transactivation function. Corepressor activity of cyclin D1 can be explained by its ability to recruit HDACs and its inhibition of AR N-C terminal interactions (224-229). A second isoform of cyclin D1, termed cyclin D1b is compromised in its ability to regulate AR activity, although it retains the ability to associate with the AR (230). The retinoblastoma protein (Rb) functions as a tumor suppressor by controlling progression through the cell cycle. Rb has been described to bind to the AR in an androgen-independent manner and enhances AR transcriptional activity in the presence of DHT (231,232). Rb and ARA70 cooperate with each other to activate transcription by the AR (231). Interestingly, pp32, which interacts with Rb, also functions as an AR coactivator (233). The presence of Rb modulates this activity. The splicing factors p54nrb and PSF are additional components of the pp32-Rb complex (233). Furthermore, retinoblastoma- associated Krüppel protein (RbaK) interact with the AR LBD in a ligand-dependent manner (234). Finally, apoptosis-antagonizing transcription factor (AATF), also termed Che- 1, that has been shown to bind Rb and promote cell cycle progression, enhances AR-mediated transactivation in a hormone- and dose- dependent manner and acts as cooperative coactivator for TSG101 (235).


m. Regulators of apoptosis

In addition to proteins that govern progression through the cell cycle, a few proteins with prominent roles in the execution of apoptosis act as AR coregulators. For example, the pro- apoptotic caspase-8 represses AR-dependent gene expression (236), but does this in a manner that is independently of its apoptotic protease activity. It does this by disrupting AR N/C interaction and inhibiting androgen- induced AR nuclear localization. Noteworthy, caspase-8 does not depend on its apoptotic protease activity to exert these functions. Caspase-8 interacts directly with the AR NTD FXXLF and WXXLF sequences, and mutations of these AR motifs prevent its repressive effect on AR-mediated transcription. In addition, knockdown of caspase-8 by RNA interference specifically affects the androgen-dependent expression of AR-targeting genes (236). Par-4, another pro- apoptotic protein, on the other hand acts as an AR coactivator (237). Par-4 physically interacts with the AR DBD, is recruited to the promoter of an AR driven gene in the presence of androgens, enhances association of the AR with DNA, and increases AR-dependent transcription. Androgen-induction of this gene is counteracted by a dominant-negative form of par-4 (237).


n. Viral oncoproteins

The human papillomavirus (HPV) E2 oncoprotein has been suggested to act as an AR coactivator by physical and functional interactions with the AR as well as the AR- associated coactivators SRC-2 and Zac1 (zinc- finger protein which regulates apoptosis and cell cycle arrest 1). SRC-2 and Zac1 are both able to act synergistically with HPV E2 proteins on the AR-dependent transcriptional activation systems (238,239). Similarly, HPV E6 and E7 are able to directly interact with the AR in the absence and presence of androgens (240). Depending on the cell type and the promoter context, these oncoproteins display AR coactivator or corepressor properties. Interestingly, also the hepatitis B virus (HBV) nonstructural protein x (HBx) can enhance AR activity (241,242). HBx dose-dependently increases androgen-stimulated AR-mediated transcription. HBx does not physically associate with ligand-bound AR in the nucleus, and it likely augments AR activity by increasing the phosphorylation of AR through HBx-mediated activation of the c-Src kinase signaling pathway (242).


o. Other, functionally diverse proteins.

o.1. Nuclear receptor co-regulators

A number of proteins that have been identified as AR coregulators cannot be readily classified into the categories listed above. Some of these have been reported to either positively or negatively regulate ligand-dependent transcription by nuclear receptors. General nuclear receptor coactivators that have been shown to enhance androgen dependent transcription by the AR include Asc-1 (activating signal cointegrator-1) (243), Asc-2 (244), components of the Trap/Mediator complex (63,245), CoCoA (coiled-coiled coactivator) (246), NRIP (nuclear receptor interaction protein) (247), PNRC (proline-rich nuclear receptor coregulatory protein) (248), TIF-1a (transcription intermediary factor 1a) (249), MRF1 (modulator recognition factor 1) (250), PDIP1 (PPARgamma-DBD-interacting protein 1) (251), Zac1 (252), GT198 (253) and ARA70 (254). Corepressors that negatively affect transactivation of the AR as well as other nuclear receptors comprise Alien (255), AES (aminoterminal enhancer of split) (256), components of the SMART and NCoR repressor complexes (17,257), RIP140 (receptor interacting protein 140 kDa) (258), PATZ (POZ-AT hook-zinc finger protein) (259), TGIF (5’TG3’ interacting factor) (260). This group of proteins also contains some coactivators (e.g. ART-27 (AR-trapped clone 27) (261) and ARA160 (262)) as well as corepressors (e.g. TIP110 (263), TZF (testicular zinc finger protein) (264-266) and ARR19 (AR corepressor 19kDa) (267)) that specifically interact with the AR. The coregulators listed here interact with the AR and affect its transcription activity via different regions of the receptor.


o.2. Kinases and phosphatases

Relatively few AR-coregulators are protein kinases and phosphatases. Some of these have been listed above. Others have been identified, including include male germ cell- associated kinase (MAK). MAK physically associates with the AR (AR NTD-DBD and DBD-LBD fragments) (268). MAK and AR are corecruited to promoters of AR target genes and MAK is able to enhance the AR transactivation potential in an androgen- and kinase-dependent manner. To this end, MAK acts in synergy with SRC-3 (268). In addition, the Ser/Thr protein kinase AR-interacting nuclear protein kinase (ANPK) interacts with the DBD-hinge region of the AR in a ligand- dependent manner (269). Overexpression of ANPK enhances AR-dependent transcription. AR does not appear to be a substrate for ANPK (269). Remarkably, also Dyrk1A, a dual specificity tyrosine phosphorykation regulated kinase which shares homology with ANPK, coactivates transactivation by the AR. This effect of Dyrk1A is mediated at least in part through physical and functional interaction with ARIP4, which is independent of its kinase activity (270). Similarly, extracellular signal-regulated kinase 8 (ERK8) negatively regulates transcriptional coactivation of the AR by ARA55 in a kinaseindependent manner (271).The serine/threonine protein kinase p90-kDa ribosomal S6 kinase (RSK), an important downstream effector of MAPK, also enhances the expression of AR target genes, an effect that was reversed by inhibiting RSK activity. This apparent coactivating activity of RSK involves both RSK kinase activity and its ability to associate with p300 (272).


Small carboxyl-terminal domain (CTD) phosphatase 2 (SCP2) interacts with the AR NTD (273). SCP2 and two other family members, SCP1 and SCP3, attenuate AR transcriptional activity, and are recruited in an androgen- and AR-dependent fashion onto the promoter of an AR-target gene. Silencing of SCP2 increases androgen-dependent transcription and augments AR loading to target gene promoter and enhancer. SCP2 is involved in promoter clearance during androgen-activated transcription (273). Finally, mediated by tumor antigen simian virus 40 small t antigen (ST), protein phosphatase 2A (PP2A) can be transferred onto the ligand-activated AR (274). Transfer by ST is strictly dependent on the agonist- activated conformation of AR, occurs within minutes of the addition of androgen to cells, and can occur in either the cytoplasm or the nucleus. ST rapidly dissociates from the complex upon PP2A binding to AR. PP2A is transferred onto the LBD of the AR, and the phosphatase activity is directed to five phosphoserines in the NTD AF-1, with a corresponding reduction in AR transactivation (274).


o.3. Diverse functions

- The remaining AR coregulators include the tumor suppressor genes LATS/KPM and PTEN. LATS2/KPM, which possesses kinase activity, interacts with the AR-LBD. This interaction is ligand-enhanced. LATS2 inhibits androgen-regulated gene expression by a mechanism that involves the inhibition of AR N-C terminal interaction. ChIP assays revealed the presence of LATS2 and AR at the promoter of AR target genes (275). PTEN also functions as an AR corepressor via a phosphatidylinositol-3-OH kinase/Akt- independent pathway (276-278). The direct interaction between the AR (amino acids 483651) and PTEN inhibits AR nuclear translocation and promotes AR protein degradation (278).


- Tob1 and the closely-related Tob2, members of an anti-proliferative protein family and negative regulators of osteoblast proliferation and differentiation both suppress AR- dependent transcriptional activity. Tob1 inhibits the nuclear foci formation of DHT- bound AR (279).


- The multifunctional DJ-1/PARK7 oncoprotein and neuromodulator was identified as an AR coactivator by its ability to interact with PIASx alpha/ARIP3 and to restore AR transcription activity by absorbing the corepressor PIASx alpha from the AR- PIASx alpha complex (280). DJ-1 interacts directly with the AR as well (281). DJ-1-binding protein, DJBP, binds the DBD of the AR in an androgen-dependent manner and colocalizes with DJ-1 or AR in the nucleus (282). DJBP represses androgen-dependent AR transactivation activity by recruiting a HDAC complex. DJ-1 partially restores AR’s activity by abrogating the DJBP-HDAC complex.


- Interestingly, apart from DJ-1, a second protein implicated in Parkinson’s disease possesses AR coregulator activity. Indeed, L- dopa-decarboxylase (DDC) interacts with the AR LBD and NTD, and enhances AR transactivation activity (283).


- Melanoma antigen gene protein MAGEA11 is an AR coactivator of particular interest (284). It specifically binds the AR N-terminal FXXLF motif, resulting in stabilization of the ligand-free AR and, in the presence of an agonist, increases exposure of AF2 to the recruitment and activation by the SRC/p160 coactivators. AR transactivation increases in response to MAGE-11 and the SRC/p160 coactivators through mechanisms that include but are not limited to the AF2 site. Thus, MAGEA11 functions as a unique AR coregulator that increases AR activity by modulating the AR interdomain interaction.


- Steroid receptor coactivator (SRA) was originally isolated as a coactivator for several nuclear receptor, including the AR, that acts as an RNA transcript (285). SRA transcripts exist in distinct ribonucleoprotein complexes that contain also SRC-1. SRA is translated as well, and 3 SRA isoforms (SRA1-3) enhance AR-mediated transcription (286).


3. Specific transcription factors

Further regulation of the transcriptional output by the AR is achieved by transcription factors, which bind to specific cognate DNA sequences. Over the last decade, multiple transcription factors have been shown to interact physically and functionally with the AR (287-324) (for an overview see Table 2). Regulation of AR-mediated transcription by these factors is mediated by different mechanisms. Some of these proteins interact directly with the AR (e.g. DAX-1 with the AR LBD (287)) and affect its ability to interact with AREs without binding directly to DNA themselves. Others (e.g. AP-1, (80)) compete with the AR for coregulators that are present in limited supply within the cell. Alternatively, some transcription factors (e.g. Foxa2, (288)) bind to DNA sequences that are interspersed in between or in close proximity to AREs, allowing cooperation in and co-regulation of transcription of the target genes. The importance of this latter mechanism of regulatory cross-talk between the AR and multiple DNA-binding transcription factors is underscored by the results of several recent ChiP-on-chip approaches aimed at mapping the genome-wide recruitment of the AR and characterizing the loci of AR enrichment (325-327). In these studies, a minority of the sites of AR occupancy meet the criteria that define a canonical ARE. Instead, most of the AR binding sites contain only an ARE half-site. A small fraction harbors no ARE-like motif at all. Strikingly, the sites of AR recruitment are selectively enriched in binding motifs for multiple specific transcription factors including Foxal, Oct1, GATA2, ETS1, AP-1, RAR, ZNF42, HNF-4α, EGR (325-327)... Occupancy at AR binding sites has been confirmed in a subset of these transcription factors (325). The presence of these transacting factors, in some cases observed in the absence of androgens, is critical for the recruitment of the AR and/or RNA polymerase II (325). Full and timely androgen induction of AR target gene expression requires normal expression of these factors and relies on the presence of intact consensus binding motifs for these factors (325). Moreover, silencing of one transacting factor can negatively affect the reciprocal occupancy of the AR binding sites by another (325). Overall, these studies offer a first glance at a hierarchical network of transcription factors that act at and regulate distinct steps in the generation of the AR transcriptional output. Moreover, they indicate that collaborating transcription factors can aid the AR in binding to sites other than canonical AREs. Such collaboration can offer insights into the manner by which the AR is able to exert finely-tuned and spatiotemporal regulation of target gene expression in a gene- and cell-specific manner, as composite response elements are likely to function as points of regulatory cross-talk. Recent array- based methods aimed at unravelling the AR interactome have isolated more DNA-binding transcription factors that are able to interact with the AR, including for instance AP-2, GATA-3, GATA-4 and E47 (328), suggesting the number of transacting factors able to stear AR recruitment and transcriptional activity in a similar manner may be substantial. It will be challenging but important to characterize the mechanism(s) of cross-talk between the AR and its associated transcription factors at loci of AR transcriptional activity and to ascertain the distribution and activity of AR coregulators at these sites.



IV. Implications and significance of the convergence of a multitude of diverse functions on the AR


a. Accurate transcriptional output by the AR requires the concerted action of numerous cellular pathways and processes

A glance at the overview of the AR coregulators reported to date and summarized in Table 1 reveals a daunting level of functional diversity among these proteins (see also Fig.2). Some of the functions listed in Table 1 can be anticipated. For instance, as the AR transcriptional complex needs to get access to the template genomic DNA to successfully execute target gene expression, it stands to reason that the AR enlists the help of regulatory proteins that can alleviate the constraints imposed by the chromatin structure. The recruitment of proteins that are able to unwind higher order chromatin structures or loosen DNA-histone interactions is a means to achieve this goal. Since the (de)acetylases and (de)methylases involved in this latter process can modify not only histones but also several components of the AR transcriptional complex as well, with severe consequences for the activity level and interplay between the individual components of the this complex, this allows for another level of tuning the transcriptional output (e.g. 63,79,81,84,87,91). The chromatin environment can also be altered by ubiquitination and sumoylation (54,55). Several AR coregulators possess the enzymatic activities required to induce these modifications and accordingly can modify the AR and several of its associated cofactors, again with severe consequences for the intrinsic activity, stability and the functional interactions of these proteins in the AR transcriptional complex (99-101, 115, 120, 123). Whether these events correspond with alterations in the local ubiquitination or sumoylation pattern in the chromatin environment of AR target genes has not yet been assessed. Similarly, from a mechanistic perspective, interaction of the AR as well as several of its associated coregulators with multiple components of the general transcription machinery makes sense. A number of proteins that are part of the spliceosome and/or function in RNA metabolism have been identified as coregulators for the AR. Traditionally, events involved in the maturation of (pre-)mRNAs have been designated to be post-transcriptional. An emerging view, however, is that all steps from transcription to translation are functionally and mechanistically coupled and that the different steps of the gene expression process should therefore be considered as co-transcriptional (138,139). Underlining the linkage between the transcriptional events leading to the production of a premature messenger RNA and its subsequent processing, splicing factors interact not only with the AR but also with RNA polymerase II subunits (138). Moreover, the nuclear compartments specifically enriched in splicing factors seem to be in close proximity of and potentially overlap with those where active transcription is taking place (138). Similarly, coupling between transcription and DNA repair allows the removal and repair of obstacles and DNA lesions (329-331). Thus, AR coregulators, which also function in DNA repair mechanisms may, therefore, not be completely unexpected. Noteworthy, transcription-DNA repair linkage typically involves chromatin remodeling activity as well as the action of the ubiquitin/proteasome pathway.


Some of the functions assigned to AR- associated coregulators may, at least at first sight, be harder to reconcile with active transcription taking place in the cell nucleus. For example, the identification of several AR coregulatory proteins with an active role in endocytosis, a process which originates at the plasma membrane and takes place predominantly in the cytosol, appears counterintuitive. It is important to keep in mind, however, that endocytic proteins are intrinsically connected with the actin cytoskeleton, and function as scaffolds that are able to link several functionally related or even seemingly unrelated cellular processes. As discussed above, components of the (actin) cytoskeleton as well as transducers and effectors of multiple intracellular signaling pathways have been assigned AR coregulator properties. Moreover, at least for some endocytic proteins, including HIP1, fractions of their cellular pool are able to undergo nucleocytoplasmic shuttling. About half of the cellular HIP1 translocates to the nucleus in an-androgen dependent manner where it is recruited to AREs, directly linking the endosomal compartment and the AR transcriptional complex (172). Finally, it is reasonable to consider that the endocytic compartment in AR-mediated transcription is related to receptor-mediated uptake. This concept has been proposed for some nuclear receptors, including the AR (332). The structural association of the cytoskeleton as well as scaffold proteins and signal transducers with endosomes does not preclude a role for these elements in AR-mediated transcription that is independent of endocytosis. The importance of the cytoskeleton in AR transactivation seems to be related mainly to the successful translocation of the AR to the nucleus; although both actin and actin-binding proteins are present in the nuclear AR transcriptional complex. Similarly, the importance of chaperone and cochaperones in AR-governed transcription is not merely related to their role in assuring the proper folding that enables the AR to bind its ligand - an event that is closely linked to proteasomal degradation. In addition, (co)chaperones are needed to ensure the conformation of the ligand-bound AR that allows its nuclear entry (14). Moreover, several of these proteins have been found to complex with the AR within the chromatin environment of target genes (156). Noteworthy, chaperones also fulfill important functions in the coating and/or uncoating of endosomes. Recent studies confirm the interaction of the AR with multiple components of the cellular pathways summarized here (167). It should be noted also that the classification we put forward does not preclude additional roles for AR- interacting regulatory proteins in the transcriptional process, such as, for instance, an architectural function in the assembly of nucleoprotein structures as has been proposed for SNURF/RNF4 (333).


b. Assembly of the AR transcriptional complex

The isolation of a multitude of proteins with AR coregulatory properties leads to speculation about the manner in which the formation of the AR transcriptional complex is orchestrated. Most of the work aimed at resolving this issue has been done using the gene encoding prostate specific antigen (PSA) as a model gene system. As is the case for most other AR target genes identified to date, androgen regulation of the PSA gene involves enhancer and promoter elements. Due to the presence of AREs (ARE I and II in the promoter region, ARE III in the enhancer region, respectively), the PSA enhancer and promoter region each display androgen responsiveness, but maximal androgen regulation requires the involvement of both regions (334,335). Thus far, a rather limited set of coregulators has been evaluated in the formation of the AR transcriptional complex at the PSA gene (64,336-338). This group consists mainly of regulatory proteins that are common, core components of the transcriptional complexes formed by many nuclear receptors and specific transcription factors, such as SRC-1, SRC-2, SRC-3, p300, p/CAF, BRG1, TRAP220 and CARM1. Upon androgen stimulation, the AR is recruited to both the enhancer and the promoter of the PSA gene, followed by the recruitment of these coactivators and RNA polymerase II. Contrary to RNA polymerase II, that displays an overall higher occupancy at the promoter region, the relative abundance of the AR and its associated coactivators is higher at the enhancer region. Of the coactivators studied, CARM-1 is the only exception, being recruited solely to the enhancer region. Overall, a combinatorial rather than a sequential recruitment of coactivators appears to be involved, with members of the p160 family of coactivators fulfilling a central role. The levels of proteins bound at the regulatory regions of the PSA gene rise gradually, reaching a maximum at 16 hours post-stimulation and then slowly decline. These kinetics of AR coactivator binding to the PSA regulatory regions correlate well with those of PSA gene transcription (337). Elegant molecular approaches have led to the proposal of an integrated chromosomal looping/RNA polymerase II tracking model resulting from the enhancer-promoter interaction. In this model, the recruitment of AR and its coactivators at both the enhancer and promoter regions creates a chromosomal loop which allows enhancer-bound AR and promoter- bound AR to share a common coactivator complex. At the same time it permits RNA polymerase II to track from the enhancer along the looped chromatin to the promoter region (337).


As indicated in Table 1, the presence of several other AR coregulators at regulatory regions of the PSA gene has been confirmed. Since these studies have focussed primarily on the recruitment of a single coregulator to a particular genomic AR binding site, they do not provide further information regarding the overall assembly of the AR transcriptional complex, the dynamics of its formation, or the relative importance of recruitment to the enhancer and the promoter... Resolving these issues will require concerted efforts of multiple research groups working in this field. Similarly, it will be vital to assess whether the manner in which the AR coregulator complex is assembled at the PSA gene can be generalized to other AR target genes. In this respect, the formation of the AR transcriptional complex at the KLK2 gene, a AR target gene with an enhancer/promoter organization that is similar to that of the PSA gene, appears to progress along the same lines (64). Very recently, several system approach studies aimed at evaluating the genome-wide binding of the AR suggest that many, if not most, of the target genes identified differ considerably in their genomic organization and in the localization, composition and clustering of AREs governing androgen-regulated transcription of these genes (325-327). Follow-up studies will therefore be critical to address the manner in which AR coregulators are recruited to these regulatory regions. Finally, the impact of cell-type specific context on these events will need to be explored.


c. Accomodation of coregulators by the AR

An intriguing question relates to the manner in which the AR can accommodate physical interaction with such a perplexing number of cofactors. Although some of these regulators are recruited to the AR transcriptional complex through an intermediary protein, the majority of them seem to be able to associate directly with the AR (see Table 1). Interaction of coregulators with different regions of the AR may offer some relief. Another means towards reducing the complexity of the AR- cofactor interaction could lay in the temporal recruitment and release of coregulators to and from the receptor. Definite insights into these matters will require concerted and extensive ChIP and Re-ChIP approaches and will most likely depend on collaborative efforts from several research groups working in this field. Some AR-associated coregulators have been claimed to display specificity for the AR. In most cases, these observations were performed at the time of isolation of the protein in question, with further characterization indicating a broader range of interaction and coregulation. For example, ARA70, the first AR coactivator to be identified by a yeast 2- hybrid approach, was initially put forward as AR-specific coregulator, but has since then been shown to modulate transcription by several other nuclear receptors (254,339). FHL2, on the other hand, does display specificity for the AR when compared with other nuclear receptors, but is known to act also as a cofactor for several specific transcription factors (340). Therefore, the number of truely AR-specific coregulators is likely to be very small. Based on the information available to date, ART-27 and ARR19 appear the only coregulators that can be qualified as AR-specific.


On the other hand, coregulators have been shown to display selectivity in the particular set of AR target genes with which they associate (58,341). Detailed exploration of this pattern of selectivity may require ChIP- on-chip approaches. Moreover, the tissue-and cell type-selective expression pattern observed for some coregulators and androgen-regulated changes in the expression of AR coactivators (341-344) may allow further modification of AR-coregulator association. Along the same lines, evidence for agonist- or antagonist-mediated induction of changes in the post-translational modification status of AR- associated coregulatory machinery is starting to emerge (345,257). Such changes may predispose coregulators to selectively interact with particular regulatory components of the AR transcriptional complex or lead to the release of cofactors from this complex.



V. Co-regulators in AR-dependent (patho)physiology


The dependency of the AR on its coregulators to form a productive transcriptional complex suggests an important role for these regulatory proteins in the development and maintenance of androgen-responsive tissues as well as in pathologies that are associated with aberrant AR function. In keeping with this notion, some AR-associated coregulators display selective or enriched expression in androgen- dependent tissues such as prostate and testis. A higher level of selectivity can be achieved by restricting the expression to specific cell types. For instance, FHL2 is selectively enriched in epithelial cells of the prostate whereas ARA55/Hic-5 is preferentially expressed in the prostatic stromal compartment (181, 346). Moreover, tissue selective AR coregulator splice variants have been described and, interestingly, such alternative splice forms can exhibit a change in activity from corepressor to coactivator or vice versa (266). The idea that androgen- responsive tissues rely on AR-coactivator expression for their functional and structural integrity is supported by knock-out mouse models. Some AR coregulator deficient mice demonstrate embryonic lethality, which suggests an important role early in development and/or an activity that is critical for overall physiology (347). In contrast, other models appear to be phenotypically normal, suggesting the function of the coregulator that is being targeted may overlap with and be rescued by the action of other cofactors (347). The actual involvement of the AR in these phenotypes, however, is hard to ascertain. Loss of other coregulators, such as those listed in Table 3 (98,153,154,348-351), gives rise to hormone-resistant phenotypes with varying degrees of severity ranging from compromised fertility, hypospadias, changes in the composition of prostate, seminal vesicles and testis.


Interestingly, AR-associated coregulators have been shown to be important for the development of pathologies that are linked to aberrant AR action. Prostate cancer (PCa) is a prominent example of such a condition. PCa cells depend on androgens for proliferation, a characteristic that is being exploited for therapeutic intervention. So-called androgen ablation therapy either targets the production of androgens or interferes with the activity of the AR (352). While androgen ablation therapy is initially successful in the majority of cases, eventually most tumors will find a way to circumvent this form of treatment and emerge as androgen depletion independent (ADI) cancers (353). Remarkably, despite the castrate levels of circulating androgens in these patients, the AR is still a critical determinant for ADI PCa cell proliferation (354,355). The unexpected “reactivation” of the AR in ADI PCa cells has been attributed to mechanisms of AR hypersensitivity (AR amplification and/or mutations), promiscuous activation of the AR (by adrenal androgens, non-androgenic steroids and even anti-androgens), outlaw AR pathways (AR activated by growth factors and cytokines, thereby bypassing the need for androgens) and local, intracrine production of androgens (independent of the low circulating serum androgens levels) (356,357). Over the last few years, the importance of the involvement of AR coactivator proteins in ADI AR activation is increasingly being recognized (358). In the progression of PCa a subset of AR coactivators has been shown to be overexpressed. Deregulated expression of AR coactivators tends to increase with tumor dedifferentiation and to correlate with aggressive disease and poor prognosis. Moreover, overexpression of these regulatory factors has been demonstrated to substantially contribute to the ADI mechanisms of AR activation described above. Therefore, overexpression of AR coactivators is considered to be a valuable target for therapeutic intervention (358). The molecular machinery underlying the aberrant expression of these critical cofactors in PCa disease progression is under intense investigation as a potential therapeutic target for the treatment of this severe disease (343,344). Similarly, efforts are being directed towards defining those regions within the AR that can mediate critical interactions with clinically relevant cofactors. Recent work from our lab led to the isolation of a discrete WXXLF motif in the AR NTD that is essential for ligandindependent AR activity that is resistant to antiandrogens such as bicalutamide (359,360). In addition to PCa, at least one case of complete androgen insensitivity syndrome, which is typically linked to an inactivating mutation of the AR (1,2), has been suggested to be causally related to a defect in or an absence of an AR-interacting regulatory protein, rather than to an aberration of the receptor itself (361). Very recently, an intriguing interplay between HBx, a nonstructural HBV protein acting as a coactivator for the AR, and the AR of host hepatocytes has been proposed to underly the male-predominance of hepatocellular carcinoma (242). As other viral oncoproteins have been suggested to be able to regulate the transcriptional activity of the AR (239-240), and since males are known to be more vulnerable to microbial infections, this mechanism may be more generally applicable. Finally, coactivators of the AR may also play a role in relatively mild conditions such as androgenic alopecia (362).



VI. Conclusions and future directions


As illustrated by this overview, a multitude of coregulators with diverse functions have the potential to converge on the AR and regulate its transcriptional output. A considerable part of this information has been derived from in vitro studies and the use of promoter-reporter constructs that do not fully recapitulate the activity of the AR in a chromatin environment (363). It will, therefore, be vital to monitor the construction of the AR transcriptional complex and the transcriptional output it generates in model systems that express an endogenous AR. Such studies will also help to determine the importance of tissue-selectivity in the recruitment of coregulators to the AR and the selective involvement of particular cellular pathways in these events.


Although an impressive number of different functions have already been ascribed to the regulatory proteins involved in the formation of a productive AR transcriptional complex, additional properties allowing these proteins to modulate transactivation by the AR cannot be ruled out. For instance, a role for SRC-1 in promoting binding of the AR to chromatin has recently been suggested (363). In this respect, a thorough characterization of the kinetics in which coregulators associate with the AR, not necessarily restricted to the DNA-bound state of the receptor, could prove to be very informative.


Typically, investigation into the functional interactions between the AR and its coregulators remains limited to the impact of coregulators on AR activity. The AR may, however, have a reciprocal effect on the intrinsic activity of its coregulators. This possibility is supported by the observation that the AR, independently of its DNA binding ability, dramatically increases the intrinsic transcriptional activity of SRC-2, CBP, and p300 that are tethered to DNA (364). This kind of “triggering” phenomenon might be important also in the non-genomic actions of the AR (which have not been addressed in the current manuscript). Moreover, mounting evidence suggests that the AR feeds back to regulate the composition and activity of its coregulator complex by modulating the expression of its cofactors (341-344). Similarly, it has been suggested that coactivators such as SRC-3 undergo post-translational modifications upon androgen treatment, resulting in specific modification “codes” that govern preferences in its interaction with other regulatory proteins (345).


An emerging view is that coregulators also function as signal integrators, relaying information from the cell surface to the nucleus (365). It will be important to determine whether this holds true also for the AR, and to identify the signalling mechanisms involved.


We apologize to authors whose work was inadvertently overlooked or could not be cited due to space constraints.


Legends


Table 1. Overview of AR-coregulators identified to date. Abbreviations : coA/coR, coactivator/corepressor ; direct/indirect, direct or indirect association with the AR ; ND, not determined. For those coregulators for which interaction between endogenously expressed coregulator and AR has been described, the method by which this information was obtained is noted as ColP (co- immunoprecipitation), MS (mass spectrometry) or ChIP (chromatin immunoprecipitation). Similarly, coregulators for which the function has been confirmed by assessing the expression of AR target genes have been marked as si (confirmation of function on endogenously expressed AR target genes obtained by siRNA-mediated knock-down of coregulator expression), AS (confirmation obtained via antisense oligos), DN (verification of coregulator function by dominant-negative isoforms), def (coregulator deficient cells were used to verify function) or over (overexpression experiments were performed).


Table 2. An overview of transcription factors that modulate AR activity. Abbreviations : A/R, the transcription factor activates or represses AR function ; direct/indirect, the transcription factor interacts directly or indirectly with the AR ; ND, not determined.


Figure 1. Covalent histone modifications associated with androgen action.


Figure 2. Schematic overview of the cellular pathways and processes converging on the AR. Abbreviations : AR, androgen receptor ; ARE, androgen response element ; chrom remod, chromatin remodeling ; HAT HDAC, histone acetyltransferase and histone deacetylase ; HMT HDM, histone methyltransferase and histone demethylase ; UB, components of the ubiquitination/proteasome pathway ; SUMO, components of the SUMOylation pathway, PIC, preinitiation complex.


EU5nHuwEGiwhlepxLrPt_PdfeQ_PyrqKtQ.png


Figure 1.


LvFlln1Tx_YRUo8UoxeKfmwHcTk_gmWWNA.png


Figure 2.


Table 1. Overview of AR coregulators identified to date.

j9XERj-ZcFN3IFbm93vX1k6QHJCb4mt9ow.png


YWn24oU703Wx7ioFrFRZ7PSxRg5kj8uyBg.png


WTvGg9_kYVdKvHXAXXZfPa-DQe7HcPMuvg.png


9x6eCN3Dlam8_kxzyzeKER-ze0nuCVfF5A.png


Cqk1W2U4f0TcsWNFfVulyrwu3ky2dwYPXg.png


6scLJ9QdvFs_UbPJ8Xp7vz3n1CaaV6nLQw.png



Table 2(a). An overview of transcription factors that modulate AR activity.

YBX1S3adkgtXL-HYjkgmkhJMAadtloGZ3Q.png



Table 2(b). Selection of coregulator deficient mouse models showing varying degrees of androgen resistance.

qZYuO-B-YN40JPQIpWSHHvjAzJigEPa9ig.png





References


Quigley CA, De Bellis A, Marschke KB, el-Awady MK, Wilson EM, French FS 1995 Androgen receptor defects: historical, clinical, and molecular perspectives. Endocr Rev 16:271-321 

Brinkmann AO 2001 Molecular basis of androgen insensitivity. Mol Cell Endocrinol 179:105-109 

Grossmann ME, Huang H, Tindall DJ 2001 Androgen receptor signaling in androgen-refractory prostate cancer. J Natl Cancer Inst 93:1687-1697 

Dehm SM, Tindall DJ 2005 Regulation of androgen receptor signaling in prostate cancer. Expert Rev Anticancer Ther 5:63-74 

De Maria N, Manno M, Villa E 2002 Sex hormones and liver cancer. Mol Cell Endocrinol 193:59-63 

Zouboulis CC, Chen WC, Thornton MJ, Qin K, Rosenfield R 2007 Sexual hormones in human skin. Horm Metab Res 39:85-95 

Davison SL, Bell R 2006 Androgen physiology. Semin Reprod Med 24:71-77 

Rainey WE, Carr BR, Sasano H, Suzuki T, Mason JI 2002 Dissecting human adrenal androgen production. Trends Endocrinol Metab 13:234-239 

Burger HG 2002 Androgen production in women. Fertil Steril 77:S3-5 

Davison SL, Davis SR 2003 Androgens in women. J Steroid Biochem Mol Biol 85:363-366 

Russell DW, Wilson JD 1994 Steroid 5 alpha-reductase: two genes/two enzymes. Annu Rev Biochem 63:25-61 

Deslypere JP, Young M, Wilson JD, McPhaul MJ 1992 Testosterone and 5 alpha-dihydrotestosterone interact differently with the androgen receptor to enhance transcription of the MMTV-CAT reporter gene. Mol Cell Endocrinol 88:15-22 

Wilson EM, French FS 1976 Binding properties of androgen receptors. Evidence for identical receptors in rat testis, epididymis, and prostate. J Biol Chem 251:5620-5629 

Prescott J, Coetzee GA 2006 Molecular chaperones throughout the life cycle of the androgen receptor. Cancer Lett 231:12-19 

Lee DK, Chang C 2003 Molecular communication between androgen receptor and general transcription machinery. J Steroid Biochem Mol Biol 84:41-49 

Heinlein CA, Chang C 2002 Androgen receptor (AR) coregulators: an overview. Endocr Rev 23: 175-200 

Wang L, Hsu CL, Chang C 2005 Androgen receptor corepressors : An overview. Prostate 63:117-130 

Tsai MJ, O'Malley BW 1994 Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem 63:451-486  

White R, Parker MG 1998 Molecular mechanisms of steroid hormone action. Endocrine- Related Cancer 5: 1-14 

Lavery DN, McEwan IJ 2006 The human androgen receptor AF1 transactivation domain: interactions with transcription factor IIF and molten-globule-like structural characteristics. Biochem Soc Trans 34:1054-1057 

He B, Lee LW, Minges JT, Wilson EM 2002 Dependence of selective gene activation on the androgen receptor NH2- and COOH-terminal interaction. J Biol Chem 277:25631-25639 

Jenster G, van der Korput HA, Trapman J, Brinkmann AO 1995 Identification of two transcription activation units in the N-terminal domain of the human androgen receptor. J Biol Chem 270:7341-7346 

Lumbroso S, Lobaccaro JM, Vial C, Sassolas G, Ollagnon B, Belon C, Pouget J, Sultan C 1997 Molecular analysis of the androgen receptor gene in Kennedy's disease. Report of two families and review of the literature. Horm Res 47: 23-29 

La Spada AR, Wilson EM, Lubahn DB, Harding AE, Fischbeck KH 1991 Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 352:77-79 

Giovannucci E, Stampfer MJ, Krithivas K, Brown M, Dahl D, Brufsky A, Talcott J, Hennekens CH, Kantoff PW 1997 The CAG repeat within the androgen receptor gene and its relationship to prostate cancer. Proc Natl Acad Sci USA 94: 3320-3323 

Shaffer PL, Jivan A, Dollins DE, Claessens F, Gewirth DT 2004 Structural basis of androgen receptor binding to selective androgen response elements. Proc Natl Acad Sci U S A 101:4758-4763 

Verrijdt G, Tanner T, Moehren U, Callewaert L, Haelens A, Claessens F 2006 The androgen receptor DNA-binding domain determines androgen selectivity of transcriptional response. Biochem Soc Trans 34:1089-1094 

Reid KJ, Hendy SC, Saito J, Sorensen P, Nelson CC 2001 Two classes of androgen receptor elements mediate cooperativity through allosteric interactions. J Biol Chem 276:2943-2952 

Wang Q, Lu J, Yong EL 2001 Ligand- and coactivator-mediated transactivation function (AF2) of the androgen receptor ligand-binding domain is inhibited by the cognate hinge region. J Biol Chem 276:7493-7499 

Haelens A, Tanner T, Denayer S, Callewaert L, Claessens F 2007 The hinge region regulates DNA binding, nuclear translocation, and transactivation of the androgen receptor. Cancer Res 67:4514-4523 

Jenster G, Trapman J, Brinkmann AO 1993 Nuclear import of the human androgen receptor. Biochem J 293:761-768 

Zhou ZX, Sar M, Simental JA, Lane MV, Wilson EM 1994 A ligand-dependent bipartite nuclear targeting signal in the human androgen receptor. Requirement for the DNA-binding domain and modulation by NH2-terminal and carboxyl-terminal sequences. J Biol Chem 269: 13115-13123 

Parker MG, White R 1996 Nuclear receptors spring into action. Nat Struct Biol 3:113-115 

Moras D, Gronemeyer H 1998 The nuclear receptor ligand-binding domain: structure and function. Curr Opin Cell Biol 10:384-391 

Matias PM, Donner P, Coelho R, Thomaz M, Peixoto C, Macedo S, Otto N, Joschko S, Scholz P, Wegg A, Basler S, Schafer M, Egner U, Carrondo MA 2000 Structural evidence for ligand specificity in the binding domain of the human androgen receptor. Implications for pathogenic gene mutations. J Biol Chem 275:26164-26171 

Sack JS, Kish KF, Wang C, Attar RM, Kiefer SE, An Y, Wu GY, Scheffler JE, Salvati ME, Krystek SR Jr, Weinmann R, Einspahr HM 2001 Crystallographic structures of the ligand-binding domains of the androgen receptor and its T877A mutant complexed with the natural agonist dihydrotestosterone. Proc Natl Acad Sci USA 98:4904-4909 

Heery DM, Kalkhoven E, Hoare S, Parker MG 1997 A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature 387:733-736 

He B, Kemppainen JA, Wilson EM 2000 FXXLF and WXXLF sequences mediate the NH2- terminal interaction with the ligand binding domain of the androgen receptor. J Biol Chem 275:22986-22994 

Chang CY, McDonnell DP 2002 Evaluation of ligand-dependent changes in AR structure using peptide probes. Mol Endocrinol 16:647-660 

Hsu CL, Chen YL, Yeh S, Ting HJ, Hu YC, Lin H, Wang X, Chang C 2003 The use of phage display technique for the isolation of androgen receptor interacting peptides with (F/W)XXL(F/W) and FXXLY new signature motifs. J Biol Chem 278:23691-23698 

He B, Wilson EM 2003 Electrostatic modulation in steroid receptor recruitment of LXXLL and FXXLF motifs. Mol Cell Biol 23:2135-2150 

Chang CY, McDonnell DP 2005 Androgen receptor-cofactor interactions as targets for new drug discovery. Trends Pharmacol Sci 26:225-228 

van Royen ME, Cunha SM, Brink MC, Mattern KA, Nigg AL, Dubbink HJ, Verschure PJ, Trapman J, Houtsmuller AB 2007 Compartmentalization of androgen receptor protein-protein interactions in living cells. J Cell Biol 177:63-72 

He B, Minges JT, Lee LW, Wilson EM 2002 The FXXLF motif mediates androgen receptor- specific interactions with coregulators. J Biol Chem 277:10226-10235 

Roeder RG 1996 The role of general initiation factors in transcription by RNA polymerase II. Trends Biochem Sci 21:327-335 

McEwan IJ, Gustafsson J 1997 Interaction of the human androgen receptor transactivation function with the general transcription factor TFIIF. Proc Natl Acad Sci USA 94:8485-8490 

Reid J, Murray I, Watt K, Betney R, McEwan IJ 2002 The androgen receptor interacts with multiple regions of the large subunit of general transcription factor TFIIF. J Biol Chem 277:4124741253 

Kumar R, Betney R, Li J, Thompson EB, McEwan IJ 2004 Induced alpha-helix structure in AF1 of the androgen receptor upon binding transcription factor TFIIF. Biochemistry 43:3008-30113 

Choudhry MA, Ball A, McEwan IJ 2006 The role of the general transcription factor IIF in androgen receptor-dependent transcription. Mol Endocrinol 20:2052-2061 

Lee DK, Duan HO, Chang C 2000 From androgen receptor to the general transcription factor TFIIH. Identification of cdk activating kinase (CAK) as an androgen receptor NH(2)-terminal associated coactivator. J Biol Chem 275:9308-9313 

Lee DK, Duan HO, Chang C 2001 Androgen receptor interacts with the positive elongation factor P-TEFb and enhances the efficiency of transcriptional elongation. J Biol Chem 276:9978-9984 

Lee DK, Li M, Chang C 2003 The second largest subunit of RNA polymerase II interacts with and enhances transactivation of androgen receptor. Biochem Biophys Res Commun 302:162-169 

Onate SA, Tsai SY, Tsai MJ, O'Malley BW 1995 Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 270:1354-1357 

Mellor J 2006 Dynamic nucleosomes and gene transcription. Trends Genet 22:320-329 

Li B, Carey M, Workman JL 2007 The role of chromatin during transcription. Cell 128:707-719 

Rouleau N, Domans'kyi A, Reeben M, Moilanen AM, Havas K, Kang Z, Owen-Hughes T, Palvimo JJ, Janne OA 2002 Novel ATPase of SNF2-like protein family interacts with androgen receptor and modulates androgen-dependent transcription. Mol Biol Cell 13:2106-2119 

Domanskyi A, Virtanen KT, Palvimo JJ, Janne OA 2006 Biochemical characterization of androgen receptor-interacting protein 4. Biochem J 393:789-795 

Marshall TW, Link KA, Petre-Draviam CE, Knudsen KE 2003 Differential requirement of SWI/SNF for androgen receptor activity. J Biol Chem 278:30605-30613 

Link KA, Burd CJ, Williams E, Marshall T, Rosson G, Henry E, Weissman B, Knudsen KE 2005 BAF57 governs androgen receptor action and androgen-dependent proliferation through SWI/SNF. Mol Cell Biol 25:2200-2215 

Hong CY, Suh JH, Kim K, Gong EY, Jeon SH, Ko M, Seong RH, Kwon HB, Lee K 2005 Modulation of androgen receptor transactivation by the SWI3-related gene product (SRG3) in multiple ways. Mol Cell Biol 25:4841-4852 

Monroy MA, Schott NM, Cox L, Chen JD, Ruh M, Chrivia JC 2003 SNF2-related CBP activator protein (SRCAP) functions as a coactivator of steroid receptor-mediated transcription through synergistic interactions with CARM-1 and GRIP-1. Mol Endocrinol 17:2519-2528 

Inoue H, Furukawa T, Giannakopoulos S, Zhou S, King DS, Tanese N 2002 Largest subunits of the human SWI/SNF chromatin-remodeling complex promote transcriptional activation by steroid hormone receptors. J Biol Chem 277:41674-41685 

Huang ZQ, Li J, Sachs LM, Cole PA, Wong J 2003 A role for cofactor-cofactor and cofactor- histone interactions in targeting p300, SWI/SNF and Mediator for transcription. EMBO J 22:21462155 

Kang Z, Janne OA, Palvimo JJ 2004 Coregulator recruitment and histone modifications in transcriptional regulation by the androgen receptor. Mol Endocrinol 18:2633-2648 

Metzger E, Wissmann M, Yin N, Müller JM, Schneider R, Peters AH, Günther T, Buettner R, Schüle R 2005 LSD1 demethylates repressive histone marks to promote androgen-receptor- dependent transcription. Nature 437:436-439 

Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y 

2006 JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell 125:483-495 

Wissmann M, Yin N, Müller JM, Greschik H, Fodor BD, Jenuwein T, Vogler C, Schneider R, Günther T, Buettner R, Metzger E, Schüle R 2007 Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat Cell Biol 9:347-353 

Kim J, Jia L, Tilley WD, Coetzee GA 2003 Dynamic methylation of histone H3 at lysine 4 in transcriptional regulation by the androgen receptor. Nucleic Acids Res 31:6741-6747 

Hong H, Kohli K, Trivedi A, Johnson DL, Stallcup MR 1996 GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proc Natl Acad Sci USA 93:4948-4952 

Ding XF, Anderson CM, Ma H, Hong H, Uht RM, Kushner PJ, Stallcup MR 1998 Nuclear receptor-binding sites of coactivators glucocorticoid receptor interacting protein 1 (GRIP1) and steroid receptor coactivator 1 (SRC-1): multiple motifs with different binding specificities. Mol Endocrinol 12:302-313 

Berrevoets CA, Doesburg P, Steketee K, Trapman J, Brinkmann AO 1998 Functional interactions of the AF-2 activation domain core region of the human androgen receptor with the amino-terminal domain and with the transcriptional coactivator TIF2 (transcriptional intermediary factor2). Mol Endocrinol 12:1172-1183 

Yeh S, Miyamoto H, Shima H, Chang C 1998 From estrogen to androgen receptor: a new pathway for sex hormones in prostate. Proc Natl Acad Sci USA 95:5527-5532 

Ma H, Hong H, Huang SM, Irvine RA, Webb P, Kushner PJ, Coetzee GA, Stallcup MR 1999 Multiple signal input and output domains of the 160-kilodalton nuclear receptor coactivator proteins. Mol Cell Biol 19:6164-6173 

74. Bevan CL, Hoare S, Claessens F, Heery DM, Parker MG 1999 The AF1 and AF2 domains of the androgen receptor interact with distinct regions of SRC1. Mol Cell Biol 19:8383-8392 

Alen P, Claessens F, Verhoeven G, Rombauts W, Peeters B 1999 The androgen receptor amino-terminal domain plays a key role in p160 coactivator-stimulated gene transcription. Mol Cell Biol 19:6085-6097 

Tan JA, Hall SH, Petrusz P, French FS 2000 Thyroid receptor activator molecule, TRAM-1, is an androgen receptor coactivator. Endocrinology 141:3440-3450 

Hong H, Kohli K, Garabedian MJ, Stallcup MR 1997 GRIP1, a transcriptional coactivator for the AF-2 transactivation domain of steroid, thyroid, retinoid, and vitamin D receptors. Mol Cell Biol 17:2735-2744 

Xu J, Li Q 2003 Review of the in vivo functions of the p160 steroid receptor coactivator family. Mol Endocrinol 17:1681-1692 

Fu M, Wang C, Reutens AT, Wang J, Angeletti RH, Siconolfi-Baez L, Ogryzko V, Avantaggiati ML, Pestell RG 2000 p300 and p300/cAMP-response element-binding protein- associated factor acetylate the androgen receptor at sites governing hormone-dependent transactivation. J Biol Chem 275:20853-20860 

Aarnisalo P, Palvimo JJ, Jänne OA 1998 CREB-binding protein in androgen receptor-mediated signaling. Proc Natl Acad Sci USA 95:2122-2127 

Fu M, Rao M, Wang C, Sakamaki T, Wang J, Di Vizio D, Zhang X, Albanese C, Balk S, Chang C, Fan S, Rosen E, Palvimo JJ, Jänne OA, Muratoglu S, Avantaggiati ML, Pestell RG 

2003 Acetylation of androgen receptor enhances coactivator binding and promotes prostate cancer cell growth. Mol Cell Biol 23:8563-8575 

Brady ME, Ozanne DM, Gaughan L, Waite I, Cook S, Neal DE, Robson CN 1999 Tip60 is a nuclear hormone receptor coactivator. J Biol Chem 274:17599-17604 

Gaughan L, Brady ME, Cook S, Neal DE, Robson CN 2001 Tip60 is a co-activator specific for class I nuclear hormone receptors. J Biol Chem 276:46841-46848 

Gaughan L, Logan IR, Cook S, Neal DE, Robson CN 2002 Tip60 and histone deacetylase 1 regulate androgen receptor activity through changes to the acetylation status of the receptor. J Biol Chem 277:25904-25913 

Sharma M, Zarnegar M, Li X, Lim B, Sun Z 2000 Androgen receptor interacts with a novel MYST protein, HBO1. J Biol Chem 275:35200-35208 

Contzler R, Regamey A, Favre B, Roger T, Hohl D, Huber M 2006 Histone acetyltransferase HBO1 inhibits NF-kappaB activity by coactivator sequestration. Biochem Biophys Res Commun 350:208-213 

Fu M, Liu M, Sauve AA, Jiao X, Zhang X, Wu X, Powell MJ, Yang T, Gu W, Avantaggiati ML, Pattabiraman N, Pestell TG, Wang F, Quong AA, Wang C, Pestell RG 2006 Hormonal control of androgen receptor function through SIRT1. Mol Cell Biol 26:8122-8135 

Karvonen U, Jänne OA, Palvimo JJ 2006 Androgen receptor regulates nuclear trafficking and nuclear domain residency of corepressor HDAC7 in a ligand-dependent fashion. Exp Cell Res 312:3165-3183 

Chen D, Ma H, Hong H, Koh SS, Huang SM, Schurter BT, Aswad DW, Stallcup MR 1999 Regulation of transcription by a protein methyltransferase. Science 284:2174-2177 

Majumder S, Liu Y, Ford OH, Mohler JL, Whang YE 2006 Involvement of arginine methyltransferase CARM1 in androgen receptor function and prostate cancer cell viability. Prostate 66:1292-1301 

Hosohata K, Li P, Hosohata Y, Qin J, Roeder RG, Wang Z 2003 Purification and identification of a novel complex which is involved in androgen receptor-dependent transcription. Mol Cell Biol 23:7019-7029 

Wang H, Huang ZQ, Xia L, Feng Q, Erdjument-Bromage H, Strahl BD, Briggs SD, Allis CD, Wong J, Tempst P, Zhang Y 2001 Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor. Science 293:853-857 

Lee DY, Northrop JP, Kuo MH, Stallcup MR 2006 Histone H3 lysine 9 methyltransferase G9a is a transcriptional coactivator for nuclear receptors. J Biol Chem 281:8476-8485 

Rayasam GV, Wendling O, Angrand PO, Mark M, Niederreither K, Song L, Lerouge T, Hager GL, Chambon P, Losson R 2003 NSD1 is essential for early post-implantation development and has a catalytically active SET domain. EMBO J 22:3153-3163 

Wang X, Yeh S, Wu G, Hsu CL, Wang L, Chiang T, Yang Y, Guo Y, Chang C 2001 Identification and characterization of a novel androgen receptor coregulator ARA267-alpha in prostate cancer cells. J Biol Chem 276:40417-40423 

Mukhopadhyay D, Riezman H 2007 Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science 315:201-205 

Kodadek T, Sikder D, Nalley K 2006 Keeping transcriptional activators under control. Cell 127:261-264 

Khan OY, Fu G, Ismail A, Srinivasan S, Cao X, Tu Y, Lu S, Nawaz Z 2006 Multifunction steroid receptor coactivator, E6-associated protein, is involved in development of the prostate gland. Mol Endocrinol 20:544-559 

Lin HK, Wang L, Hu YC, Altuwaijri S, Chang C 2002 Phosphorylation-dependent ubiquitylation and degradation of androgen receptor by Akt require Mdm2 E3 ligase. EMBO J 21:4037-4048 

Gaughan L, Logan IR, Neal DE, Robson CN 2005 Regulation of androgen receptor and histone deacetylase 1 by Mdm2-mediated ubiquitylation. Nucleic Acids Res 33:13-26 

Logan IR, Gaughan L, McCracken SR, Sapountzi V, Leung HY, Robson CN 2006 Human PIRH2 enhances androgen receptor signaling through inhibition of histone deacetylase 1 and is overexpressed in prostate cancer. Mol Cell Biol 26:6502-6510 

Moilanen AM, Poukka H, Karvonen U, Hakli M, Janne OA, Palvimo JJ 1998 Identification of a novel RING finger protein as a coregulator in steroid receptor-mediated gene transcription. Mol Cell Biol 18:5128-5139 

Poukka H, Aarnisalo P, Santti H, Janne OA, Palvimo JJ 2000 Coregulator small nuclear RING finger protein (SNURF) enhances Sp1- and steroid receptor-mediated transcription by different mechanisms. J Biol Chem 275:571-579 

Poukka H, Karvonen U, Yoshikawa N, Tanaka H, Palvimo JJ, Janne OA 2000 The RING finger protein SNURF modulates nuclear trafficking of the androgen receptor. J Cell Sci 113:29913001 

Rees I, Lee S, Kim H, Tsai FT 2006 The E3 ubiquitin ligase CHIP binds the androgen receptor in a phosphorylation-dependent manner. Biochim Biophys Acta 1764:1073-1079 

He B, Bai S, Hnat AT, Kalman RI, Minges JT, Patterson C, Wilson EM 2004 An androgen receptor NH2-terminal conserved motif interacts with the COOH terminus of the Hsp70-interacting protein (CHIP). J Biol Chem 279:30643-30653 

Cardozo CP, Michaud C, Ost MC, Fliss AE, Yang E, Patterson C, Hall SJ, Caplan AJ 2003 C-terminal Hsp-interacting protein slows androgen receptor synthesis and reduces its rate of degradation. Arch Biochem Biophys 410:134-140 

Beitel LK, Elhaji YA, Lumbroso R, Wing SS, Panet-Raymond V, Gottlieb B, Pinsky L, Trifiro MA 2002 Cloning and characterization of an androgen receptor N-terminal-interacting protein with ubiquitin-protein ligase activity. J Mol Endocrinol 29:41-60 

Kang HY, Yeh S, Fujimoto N, Chang C 1999 Cloning and characterization of human prostate coactivator ARA54, a novel protein that associates with the androgen receptor. J Biol Chem 274:8570-8576 

Ito K, Adachi S, Iwakami R, Yasuda H, Muto Y, Seki N, Okano Y 2001 N-Terminally extended human ubiquitin-conjugating enzymes (E2s) mediate the ubiquitination of RING-finger proteins, ARA54 and RNF8. Eur J Biochem 268:2725-2732 

Omwancha J, Zhou XF, Chen SY, Baslan T, Fisher CJ, Zheng Z, Cai C, Shemshedini L 2006 Makorin RING finger protein 1 (MKRN1) has negative and positive effects on RNA polymerase II-dependent transcription. Endocrine 29:363-373 

Faus H, Meyer HA, Huber M, Bahr I, Haendler B 2005 The ubiquitin-specific protease USP10 modulates androgen receptor function. Mol Cell Endocrinol 245:138-146 

Verma S, Ismail A, Gao X, Fu G, Li X, O'Malley BW, Nawaz Z 2004 The ubiquitin- conjugating enzyme UBCH7 acts as a coactivator for steroid hormone receptors. Mol Cell Biol 24:8716-8726 

Sun Z, Pan J, Hope WX, Cohen SN, Balk SP 1999 Tumor susceptibility gene 101 protein represses androgen receptor transactivation and interacts with p300. Cancer 86:689-696 

Burgdorf S, Leister P, Scheidtmann KH 2004 TSG101 interacts with apoptosis-antagonizing transcription factor and enhances androgen receptor-mediated transcription by promoting its monoubiquitination. J Biol Chem 279:17524-17534 

Jaworski T 2006 Degradation and beyond: control of androgen receptor activity by the proteasome system. Cell Mol Biol Lett 11:109-131 

Lin HK, Altuwaijri S, Lin WJ, Kan PY, Collins LL, Chang C 2002 Proteasome activity is required for androgen receptor transcriptional activity via regulation of androgen receptor nuclear translocation and interaction with coregulators in prostate cancer cells. J Biol Chem 277:36570-36576 

Kang Z, Pirskanen A, Janne OA, Palvimo JJ 2002 Involvement of proteasome in the dynamic assembly of the androgen receptor transcription complex. J Biol Chem 277:48366-48371 

Bossis G, Melchior F 2006 SUMO: regulating the regulator. Cell Div 1:13-20 

Poukka H, Karvonen U, Janne OA, Palvimo JJ 2000 Covalent modification of the androgen receptor by small ubiquitin-like modifier 1 (SUMO-1). Proc Natl Acad Sci USA 97:14145-14150 

Zheng Z, Cai C, Omwancha J, Chen SY, Baslan T, Shemshedini L 2006 SUMO-3 enhances androgen receptor transcriptional activity through a sumoylation-independent mechanism in prostate cancer cells. J Biol Chem 281:4002-4012 

Poukka H, Aarnisalo P, Karvonen U, Palvimo JJ, Janne OA 1999 Ubc9 interacts with the androgen receptor and activates receptor-dependent transcription. J Biol Chem 274:19441-19446 

Chang YL, Huang CJ, Chan JY, Liu PY, Chang HP, Huang SM 2007 Regulation of nuclear receptor and coactivator functions by the carboxyl terminus of ubiquitin-conjugating enzyme 9. Int J Biochem Cell Biol 39:1035-1046 

Junicho A, Matsuda T, Yamamoto T, Kishi H, Korkmaz K, Saatcioglu F, Fuse H, Muraguchi A 2000 Protein inhibitor of activated STAT3 regulates androgen receptor signaling in prostate carcinoma cells. Biochem Biophys Res Commun 278:9-13 

Matsuda T, Junicho A, Yamamoto T, Kishi H, Korkmaz K, Saatcioglu F, Fuse H, Muraguchi A 2001 Cross-talk between signal transducer and activator of transcription 3 and androgen receptor signaling in prostate carcinoma cells. Biochem Biophys Res Commun 283:179-187 

Tan J, Hall SH, Hamil KG, Grossman G, Petrusz P, Liao J, Shuai K, French FS 2000 Protein inhibitor of activated STAT-1 (signal transducer and activator of transcription-1) is a nuclear receptor coregulator expressed in human testis. Mol Endocrinol 14:14-26 

Kotaja N, Aittomaki S, Silvennoinen O, Palvimo JJ, Janne OA 2000 ARIP3 (androgen receptor-interacting protein 3) and other PIAS (protein inhibitor of activated STAT) proteins differ in their ability to modulate steroid receptor-dependent transcriptional activation. Mol Endocrinol 14:1986-2000 

Gross M, Liu B, Tan J, French FS, Carey M, Shuai K 2001 Distinct effects of PIAS proteins on androgen-mediated gene activation in prostate cancer cells. Oncogene 20:3880-3887 

Kotaja N, Vihinen M, Palvimo JJ, Janne OA 2002 Androgen receptor-interacting protein 3 and other PIAS proteins cooperate with glucocorticoid receptor-interacting protein 1 in steroid receptor-dependent signaling. J Biol Chem 277:17781-17788 

Nishida T, Yasuda H 2002 PIAS1 and PIASxalpha function as SUMO-E3 ligases toward androgen receptor and repress androgen receptor-dependent transcription. J Biol Chem 277:4131141317 

Kotaja N, Karvonen U, Janne OA, Palvimo JJ 2002 PIAS proteins modulate transcription factors by functioning as SUMO-1 ligases. Mol Cell Biol 22:5222-5234 

Gross M, Yang R, Top I, Gasper C, Shuai K 2004 PIASy-mediated repression of the androgen receptor is independent of sumoylation. Oncogene 23:3059-3066 

Sharma M, Li X, Wang Y, Zarnegar M, Huang CY, Palvimo JJ, Lim B, Sun Z 2003 hZimp10 is an androgen receptor co-activator and forms a complex with SUMO-1 at replication foci. EMBO J 22:6101-6114 

Huang CY, Beliakoff J, Li X, Lee J, Li X, Sharma M, Lim B, Sun Z 2005 hZimp7, a novel PIAS-like protein, enhances androgen receptor-mediated transcription and interacts with SWI/SNF- like BAF complexes. Mol Endocrinol 19:2915-2929 

Beliakoff J, Sun Z 2006 Zimp7 and Zimp10, two novel PIAS-like proteins, function as androgen receptor coregulators. Nucl Recept Signal 4:e017 

Cheng J, Wang D, Wang Z, Yeh ET 2004 SENP1 enhances androgen receptor-dependent transcription through desumoylation of histone deacetylase 1. Mol Cell Biol 24:6021-6028 

Fan M, Long X, Bailey JA, Reed CA, Osborne E, Gize EA, Kirk EA, Bigsby RM, Nephew 

KP 2002 The activating enzyme of NEDD8 inhibits steroid receptor function. Mol Endocrinol 16:315-330 

Auboeuf D, Dowhan DH, Dutertre M, Martin N, Berget SM, O'Malley BW 2005 A subset of nuclear receptor coregulators act as coupling proteins during synthesis and maturation of RNA transcripts. Mol Cell Biol 25:5307-5316 

Auboeuf D, Batsché E, Dutertre M, Muchardt C, O'Malley BW 2007 Coregulators: transducing signal from transcription to alternative splicing. Trends Endocrinol Metab 18:122-129 

Ishitani K, Yoshida T, Kitagawa H, Ohta H, Nozawa S, Kato S 2003 p54nrb acts as a transcriptional coactivator for activation function 1 of the human androgen receptor. Biochem Biophys Res Commun 306:660-665 

Zhao Y, Goto K, Saitoh M, Yanase T, Nomura M, Okabe T, Takayanagi R, Nawata H 2002 Activation function-1 domain of androgen receptor contributes to the interaction between subnuclear splicing factor compartment and nuclear receptor compartment. Identification of the p102 U5 small nuclear ribonucleoprotein particle-binding protein as a coactivator for the receptor. J Biol Chem 277:30031-30039 

Fan S, Goto K, Chen G, Morinaga H, Nomura M, Okabe T, Nawata H, Yanase T 2006 Identification of the functional domains of ANT-1, a novel coactivator of the androgen receptor. Biochem Biophys Res Commun 341:192-201 

Yang Z, Chang YJ, Miyamoto H, Yeh S, Yao JL, di Sant'Agnese PA, Tsai MY, Chang C 2007 Suppression of androgen receptor transactivation and prostate cancer cell growth by heterogeneous nuclear ribonucleoprotein A1 via interaction with androgen receptor coregulator ARA54. Endocrinology 148:1340-1349 

Mayeur GL, Kung WJ, Martinez A, Izumiya C, Chen DJ, Kung HJ 2005 Ku is a novel transcriptional recycling coactivator of the androgen receptor in prostate cancer cells. J Biol Chem 280:10827-10833 

Wang L, Hsu CL, Ni J, Wang PH, Yeh S, Keng P, Chang C 2004 Human checkpoint protein hRad9 functions as a negative coregulator to repress androgen receptor transactivation in prostate cancer cells. Mol Cell Biol 24:2202-2213 

Park JJ, Irvine RA, Buchanan G, Koh SS, Park JM, Tilley WD, Stallcup MR, Press MF, Coetzee GA 2000 Breast cancer susceptibility gene 1 (BRCAI) is a coactivator of the androgen receptor. Cancer Res 60:5946-5949 

Yeh S, Hu YC, Rahman M, Lin HK, Hsu CL, Ting HJ, Kang HY, Chang C 2000 Increase of androgen-induced cell death and androgen receptor transactivation by BRCA1 in prostate cancer cells. Proc Natl Acad Sci USA 97:11256-11261 

Shin S, Verma IM 2003 BRCA2 cooperates with histone acetyltransferases in androgen receptor-mediated transcription. Proc Natl Acad Sci USA 100:7201-7216 

Fliss AE, Rao J, Melville MW, Cheetham ME, Caplan AJ 1999 Domain requirements of DnaJ-like (Hsp40) molecular chaperones in the activation of a steroid hormone receptor. 

J Biol Chem 274:34045-34052 

Fan CY, Ren HY, Lee P, Caplan AJ, Cyr DM 2005 The type I Hsp40 zinc finger-like region is required for Hsp70 to capture non-native polypeptides from Ydj1. J Biol Chem 280:695-702 

Terada K, Yomogida K, Imai T, Kiyonari H, Takeda N, Kadomatsu T, Yano M, Aizawa S, Mori M 2005 A type I DnaJ homolog, DjA1, regulates androgen receptor signaling and spermatogenesis. EMBO J 24:611-622 

Rao J, Lee P, Benzeno S, Cardozo C, Albertus J, Robins DM, Caplan AJ 2001 Functional interaction of human Cdc37 with the androgen receptor but not with the glucocorticoid receptor. J Biol Chem 276:5814-5820 

Cheung-Flynn J, Prapapanich V, Cox MB, Riggs DL, Suarez-Quian C, Smith DF 2005 Physiological role for the cochaperone FKBP52 in androgen receptor signaling. Mol Endocrinol 19:1654-1666 

Yong W, Yang Z, Periyasamy S, Chen H, Yucel S, Li W, Lin LY, Wolf IM, Cohn MJ, Baskin LS, Sanchez ER, Shou W 2007 Essential role for Co-chaperone Fkbp52 but not Fkbp51 in androgen receptor-mediated signaling and physiology. J Biol Chem 282:5026-5036 

Febbo PG, Lowenberg M, Thorner AR, Brown M, Loda M, Golub TR 2005 Androgen mediated regulation and functional implications of fkbp51 expression in prostate cancer. J Urol 173:1772-1777 

Shatkina L, Mink S, Rogatsch H, Klocker H, Langer G, Nestl A, Cato AC 2003 The cochaperone Bag-1L enhances androgen receptor action via interaction with the NH2-terminal region of the receptor. Mol Cell Biol 23:7189-7197 

Froesch BA, Takayama S, Reed JC 1998 BAG-1L protein enhances androgen receptor function. J Biol Chem 273:11660-11666 

Alberti S, Esser C, Hohfeld J 2003 BAG-1--a nucleotide exchange factor of Hsc70 with multiple cellular functions. Cell Stress Chaperones 8:225-231 

Miralles F, Visa N 2006 Actin in transcription and transcription regulation. Curr Opin Cell Biol 18:261-266 

Ting HJ, Yeh S, Nishimura K, Chang C 2002 Supervillin associates with androgen receptor and modulates its transcriptional activity. Proc Natl Acad Sci USA 99:661-666 

Ting HJ, Hu YC, Chang C 2004 Actin monomer enhances supervillin-modulated androgen receptor transactivation. Biochem Biophys Res Commun 319:393-396 

Nishimura K, Ting HJ, Harada Y, Tokizane T, Nonomura N, Kang HY, Chang HC, Yeh S, Miyamoto H, Shin M, Aozasa K, Okuyama A, Chang C 2003 Modulation of androgen receptor transactivation by gelsolin: a newly identified androgen receptor coregulator. Cancer Res 63:4888-4894 

Ozanne DM, Brady ME, Cook S, Gaughan L, Neal DE, Robson CN 2000 Androgen receptor nuclear translocation is facilitated by the f-actin cross-linking protein filamin. Mol Endocrinol 14:1618-1626 

Loy CJ, Sim KS, Yong EL 2003 Filamin-A fragment localizes to the nucleus to regulate androgen receptor and coactivator functions. Proc Natl Acad Sci USA 100:4562-4567 

Wang Y, Kreisberg JI, Bedolla RG, Mikhailova M, Devere White RW, Ghosh PM 2007 A 90 kDa fragment of filamin A promotes Casodex-induced growth inhibition in Casodex-resistant androgen receptor positive C4-2 prostate cancer cells. Oncogene [Epub ahead of print] 

Huang SM, Huang CJ, Wang WM, Kang JC, Hsu WC 2004 The enhancement of nuclear receptor transcriptional activation by a mouse actin-binding protein, alpha actinin 2. J Mol Endocrinol 32:481-496 

Jasavala R, Martinez H, Thumar J, Andaya A, Gingras AC, Eng JK, Aebersold R, Han DK, Wright ME 2007 Identification of putative androgen receptor interaction protein modules: cytoskeleton and endosomes modulate androgen receptor signaling in prostate cancer cells. Mol Cell Proteomics 6:252-271 

Yang Z, Chang YJ, Miyamoto H, Ni J, Niu Y, Chen Z, Chen YL, Yao JL, di Sant'Agnese PA, Chang C 2007 Transgelin functions as a suppressor via inhibition of ARA54-enhanced androgen receptor transactivation and prostate cancer cell growth. 

Mol Endocrinol 21:343-358 

Zhang Y, Yang Y, Yeh S, Chang C 2004 ARA67/PAT1 functions as a repressor to suppress androgen receptor transactivation. Mol Cell Biol 24:1044-1057 

Maldonado-Baez L, Wendland B 2006 Endocytic adaptors: recruiters, coordinators and regulators. Trends Cell Biol 16:505-513 

Cheng ZJ, Singh RD, Marks DL, Pagano RE 2006 Membrane microdomains, caveolae, and caveolar endocytosis of sphingolipids. Mol Membr Biol 23:101-110 

Mills IG, Gaughan L, Robson C, Ross T, McCracken S, Kelly J, Neal DE 2005 Huntingtin interacting protein 1 modulates the transcriptional activity of nuclear hormone receptors. J Cell Biol 170:191-200 

Yang L, Lin HK, Altuwaijri S, Xie S, Wang L, Chang C 2003 APPL suppresses androgen receptor transactivation via potentiating Akt activity. J Biol Chem 278:16820-16827 

Takeshita Y, Fujinaga R, Zhao C, Yanai A, Shinoda K 2006 Huntingtin-associated protein 1 (HAP1) interacts with androgen receptor (AR) and suppresses SBMA-mutant-AR-induced apoptosis. Hum Mol Genet 15:2298-2312 

Ray MR, Wafa LA, Cheng H, Snoek R, Fazli L, Gleave M, Rennie PS 2006 Cyclin G- associated kinase: a novel androgen receptor-interacting transcriptional coactivator that is overexpressed in hormone refractory prostate cancer. Int J Cancer 118:1108-19 

Lu ML, Schneider MC, Zheng Y, Zhang X, Richie JP 2001 Caveolin-1 interacts with androgen receptor. A positive modulator of androgen receptor mediated transactivation. J Biol Chem 276:13442-13451 

Li L, Ren CH, Tahir SA, Ren C, Thompson TC 2003 Caveolin-1 maintains activated Akt in prostate cancer cells through scaffolding domain binding site interactions with and inhibition of serine/threonine protein phosphatases PP1 and PP2A. Mol Cell Biol 23:9389-9404 

Fujimoto N, Yeh S, Kang HY, Inui S, Chang HC, Mizokami A, Chang C 1999 Cloning and characterization of androgen receptor coactivator, ARA55, in human prostate. J Biol Chem 274:83168321 

Wang X, Yang Y, Guo X, Sampson ER, Hsu CL, Tsai MY, Yeh S, Wu G, Guo Y, Chang C 

Suppression of androgen receptor transactivation by Pyk2 via interaction and phosphorylation of the ARA55 coregulator. J Biol Chem 277:15426-15431 

Kasai M, Guerrero-Santoro J, Friedman R, Leman ES, Getzenberg RH, DeFranco DB 

The Group 3 LIM domain protein paxillin potentiates androgen receptor transactivation in prostate cancer cell lines. Cancer Res 63:4927-4935 

Müller JM, Isele U, Metzger E, Rempel A, Moser M, Pscherer A, Breyer T, Holubarsch C, Buettner R, Schüle R 2000 FHL2, a novel tissue-specific coactivator of the androgen receptor. EMBO J 19:359-369 

Nair SS, Guo Z, Mueller JM, Koochekpour S, Qiu Y, Tekmal RR, Schüle R, Kung HJ, Kumar R, Vadlamudi RK 2007 Proline-, glutamic acid-, and leucine-rich protein-1/modulator of nongenomic activity of estrogen receptor enhances androgen receptor functions through LIM-only coactivator, four-and-a-half LIM-only protein 2. Mol Endocrinol 21:613-624 

Tujague M, Thomsen JS, Mizuki K, Sadek CM, Gustafsson JA 2004 The focal adhesion protein vinexin alpha regulates the phosphorylation and activity of estrogen receptor alpha. 

J Biol Chem 279:9255-9263 

Lyons LS, Burnstein KL 2006 Vav3, a Rho GTPase guanine nucleotide exchange factor, increases during progression to androgen independence in prostate cancer cells and potentiates androgen receptor transcriptional activity. Mol Endocrinol 20:1061-1072 

Dong Z, Liu Y, Lu S, Wang A, Lee K, Wang LH, Revelo M, Lu S 2006 Vav3 oncogene is overexpressed and regulates cell growth and androgen receptor activity in human prostate cancer. Mol Endocrinol 20:2315-2325 

Su LF, Knoblauch R, Garabedian MJ 2001 Rho GTPases as modulators of the estrogen receptor transcriptional response. J Biol Chem 276:3231-3237 

Mahajan NP, Liu Y, Majumder S, Warren MR, Parker CE, Mohler JL, Earp HS, Whang 

YE 2007 Activated Cdc42-associated kinase Ack1 promotes prostate cancer progression via androgen receptor tyrosine phosphorylation. Proc Natl Acad Sci USA 104:8438-8443 

Metzger E, Müller JM, Ferrari S, Buettner R, Schüle R 2003 A novel inducible transactivation domain in the androgen receptor: implications for PRK in prostate cancer. EMBO J 22:270-280 

Rao MA, Cheng H, Quayle AN, Nishitani H, Nelson CC, Rennie PS 2002 RanBPM, a nuclear protein that interacts with and regulates transcriptional activity of androgen receptor and glucocorticoid receptor. J Biol Chem 277:48020-48027 

Hsiao PW, Lin DL, Nakao R, Chang C 1999 The linkage of Kennedy's neuron disease to ARA24, the first identified androgen receptor polyglutamine region-associated coactivator. J Biol Chem 274:20229-20234 

Yang F, Li X, Sharma M, Zarnegar M, Lim B, Sun Z 2001 Androgen receptor specifically interacts with a novel p21-activated kinase, PAK6. J Biol Chem 276:15345-15353 

Lee SR, Ramos SM, Ko A, Masiello D, Swanson KD, Lu ML, Balk SP 2002 AR and ER interaction with a p21-activated kinase (PAK6). Mol Endocrinol 16:85-99 

Schrantz N, da Silva Correia J, Fowler B, Ge Q, Sun Z, Bokoch GM 2004 Mechanism of p21-activated kinase 6-mediated inhibition of androgen receptor signaling. J Biol Chem 279:1922-1931 

Rigas AC, Ozanne DM, Neal DE, Robson CN 2003 The scaffolding protein RACK1 interacts with androgen receptor and promotes cross-talk through a protein kinase C signaling pathway. J Biol Chem 278:46087-46093 

Kraus S, Gioeli D, Vomastek T, Gordon V, Weber MJ 2006 Receptor for activated C kinase 1 (RACK1) and Src regulate the tyrosine phosphorylation and function of the androgen receptor. Cancer Res 66:11047-11054 

Guo Z, Dai B, Jiang T, Xu K, Xie Y, Kim O, Nesheiwat I, Kong X, Melamed J, Handratta VD, Njar VC, Brodie AM, Yu LR, Veenstra TD, Chen H, Qiu Y 2006 Regulation of androgen receptor activity by tyrosine phosphorylation. Cancer Cell 10:309-319 

De Miguel F, Lee SO, Onate SA, Gao AC 2003 Stat3 enhances transactivation of steroid hormone receptors. Nucl Recept 1:3 

Chen T, Wang LH, Farrar WL 2000 Interleukin 6 activates androgen receptor-mediated gene expression through a signal transducer and activator of transcription 3-dependent pathway in LNCaP prostate cancer cells. Cancer Res 60:2132-2135 

Yamamoto T, Sato N, Sekine Y, Yumioka T, Imoto S, Junicho A, Fuse H, Matsuda T 2003 Molecular interactions between STAT3 and protein inhibitor of activated STAT3, and androgen receptor. Biochem Biophys Res Commun. 306:610-615 

Kang HY, Huang KE, Chang SY, Ma WL, Lin WJ, Chang C 2002 Differential modulation of androgen receptor-mediated transactivation by Smad3 and tumor suppressor Smad4. J Biol Chem 277:43749-43756 

Hayes SA, Zarnegar M, Sharma M, Yang F, Peehl DM, ten Dijke P, Sun Z 2001 SMAD3 represses androgen receptor-mediated transcription. Cancer Res 61:2112-2118 

Kang HY, Lin HK, Hu YC, Yeh S, Huang KE, Chang C 2001 From transforming growth factor-beta signaling to androgen action: identification of Smad3 as an androgen receptor coregulator in prostate cancer cells. Proc Natl Acad Sci USA 98:3018-3023 

Chipuk JE, Cornelius SC, Pultz NJ, Jorgensen JS, Bonham MJ, Kim SJ, Danielpour D 

2002 The androgen receptor represses transforming growth factor-beta signaling through interaction with Smad3. J Biol Chem 277:1240-1248 

Zhang Y, Fondell JD, Wang Q, Xia X, Cheng A, Lu ML, Hamburger AW 2002 Repression of androgen receptor mediated transcription by the ErbB-3 binding protein, Ebp1. Oncogene 21:5609-56018 

Zhang Y, Hamburger AW 2005 Specificity and heregulin regulation of Ebp1 (ErbB3 binding protein 1) mediated repression of androgen receptor signalling. Br J Cancer 92:140-146 

Zhang Y, Wang XW, Jelovac D, Nakanishi T, Yu MH, Akinmade D, Goloubeva O, Ross DD, Brodie A, Hamburger AW 2005 The ErbB3-binding protein Ebp1 suppresses androgen receptor-mediated gene transcription and tumorigenesis of prostate cancer cells. Proc Natl Acad Sci USA 102:9890-9895 

Zhang Y, Akinmade D, Hamburger AW 2005 The ErbB3 binding protein Ebp1 interacts with Sin3A to repress E2F1 and AR-mediated transcription. Nucleic Acids Res 33:6024-6033 

Belandia B, Powell SM, Garcia-Pedrero JM, Walker MM, Bevan CL, Parker MG 2005 Hey1, a mediator of notch signaling, is an androgen receptor corepressor. Mol Cell Biol 25:1425-1436 

Bettoun DJ, Scafonas A, Rutledge SJ, Hodor P, Chen O, Gambone C, Vogel R, McElwee- Witmer S, Bai C, Freedman L, Schmidt A 2005 Interaction between the androgen receptor and RNase L mediates a cross-talk between the interferon and androgen signaling pathways. J Biol Chem 280:38898-38901 

Truica CI, Byers S, Gelmann EP 2000 Beta-catenin affects androgen receptor transcriptional activity and ligand specificity. Cancer Res 60:4709-4713 

Mulholland DJ, Cheng H, Reid K, Rennie PS, Nelson CC 2002 The androgen receptor can promote beta-catenin nuclear translocation independently of adenomatous polyposis coli. J Biol Chem 277:17933-17943 

Yang F, Li X, Sharma M, Sasaki CY, Longo DL, Lim B, Sun Z 2002 Linking beta-catenin to androgen-signaling pathway. J Biol Chem 277:11336-11344 

Song LN, Herrell R, Byers S, Shah S, Wilson EM, Gelmann EP 2003 Beta-catenin binds to the activation function 2 region of the androgen receptor and modulates the effects of the N-terminal domain and TIF2 on ligand-dependent transcription. Mol Cell Biol 23:1674-1687 

Pawlowski JE, Ertel JR, Allen MP, Xu M, Butler C, Wilson EM, Wierman ME 2002 Liganded androgen receptor interaction with beta-catenin: nuclear co-localization and modulation of transcriptional activity in neuronal cells. J Biol Chem 277:20702-20710 

Wang L, Lin HK, Hu YC, Xie S, Yang L, Chang C 2004 Suppression of androgen receptor- mediated transactivation and cell growth by the glycogen synthase kinase 3 beta in prostate cells. J Biol Chem 279:32444-32452 

Salas TR, Kim J, Vakar-Lopez F, Sabichi AL, Troncoso P, Jenster G, Kikuchi A, Chen SY, Shemshedini L, Suraokar M, Logothetis CJ, DiGiovanni J, Lippman SM, Menter DG 2004 Glycogen synthase kinase-3 beta is involved in the phosphorylation and suppression of androgen receptor activity. J Biol Chem 279:19191-19200 

Liao X, Thrasher JB, Holzbeierlein J, Stanley S, Li B 2004 Glycogen synthase kinase-3beta activity is required for androgen-stimulated gene expression in prostate cancer. Endocrinology 145:2941-2949 

Amir AL, Barua M, McKnight NC, Cheng S, Yuan X, Balk SP 2003 A direct beta-catenin- independent interaction between androgen receptor and T cell factor 4. J Biol Chem 278:30828-30834 

Yamamoto A, Hashimoto Y, Kohri K, Ogata E, Kato S, Ikeda K, Nakanishi M 2000 Cyclin E as a coactivator of the androgen receptor. J Cell Biol 150:873-880 

Ma ZQ, Liu Z, Ngan ES, Tsai SY 2001 Cdc25B functions as a novel coactivator for the steroid receptors. Mol Cell Biol 21:8056-8067 

Ngan ES, Hashimoto Y, Ma ZQ, Tsai MJ, Tsai SY 2003 Overexpression of Cdc25B, an androgen receptor coactivator, in prostate cancer. Oncogene 22:734-739 

Chua SS, Ma Z, Ngan E, Tsai SY 2004 Cdc25B as a steroid receptor coactivator. Vitam Horm 68:231-256 

Lim JT, Mansukhani M, Weinstein IB 2005 Cyclin-dependent kinase 6 associates with the androgen receptor and enhances its transcriptional activity in prostate cancer cells. Proc Natl Acad Sci USA 102:5156-51561 

Knudsen KE, Cavenee WK, Arden KC 1999 D-type cyclins complex with the androgen receptor and inhibit its transcriptional transactivation ability. Cancer Res 59:2297-2301 

Reutens AT, Fu M, Wang C, Albanese C, McPhaul MJ, Sun Z, Balk SP, Jänne OA, Palvimo JJ, Pestell RG 2001 Cyclin D1 binds the androgen receptor and regulates hormone- dependent signaling in a p300/CBP-associated factor (P/CAF)-dependent manner. Mol Endocrinol 15:797-811 

Petre-Draviam CE, Cook SL, Burd CJ, Marshall TW, Wetherill YB, Knudsen KE 2003 Specificity of cyclin D1 for androgen receptor regulation. Cancer Res 63:4903-4913 

Burd CJ, Petre CE, Moghadam H, Wilson EM, Knudsen KE 2005 Cyclin D1 binding to the androgen receptor (AR) NH2-terminal domain inhibits activation function 2 association and reveals dual roles for AR corepression. Mol Endocrinol 19:607-620 

Petre-Draviam CE, Williams EB, Burd CJ, Gladden A, Moghadam H, Meller J, Diehl JA, Knudsen KE 2005 A central domain of cyclin D1 mediates nuclear receptor corepressor activity. Oncogene 24:431-444 

Petre CE, Wetherill YB, Danielsen M, Knudsen KE 2002 Cyclin D1: mechanism and consequence of androgen receptor co-repressor activity. J Biol Chem 277:2207-2215 

Burd CJ, Petre CE, Morey LM, Wang Y, Revelo MP, Haiman CA, Lu S, Fenoglio-Preiser CM, Li J, Knudsen ES, Wong J, Knudsen KE 2006 Cyclin D1b variant influences prostate cancer growth through aberrant androgen receptor regulation. Proc Natl Acad Sci USA 103:2190-2195 

Yeh S, Miyamoto H, Nishimura K, Kang H, Ludlow J, Hsiao P, Wang C, Su C, Chang C 1998 Retinoblastoma, a tumor suppressor, is a coactivator for the androgen receptor in human prostate cancer DU145 cells. Biochem Biophys Res Commun 248:361-367 

Lu J, Danielsen M 1998 Differential regulation of androgen and glucocorticoid receptors by retinoblastoma protein. J Biol Chem 273:31528-31533 

Adegbola O, Pasternack GR 2005 A pp32-retinoblastoma protein complex modulates androgen receptor-mediated transcription and associates with components of the splicing machinery. Biochem Biophys Res Commun 334:702-708 

Hofman K, Swinnen JV, Claessens F, Verhoeven G, Heyns W 2003 The retinoblastoma protein-associated transcription repressor RBaK interacts with the androgen receptor and enhances its transcriptional activity. J Mol Endocrinol 31:583-596 

Burgdorf S, Leister P, Scheidtmann KH 2004 TSG101 interacts with apoptosis-antagonizing transcription factor and enhances androgen receptor-mediated transcription by promoting its monoubiquitination. J Biol Chem 279:17524-17534 

Qi W, Wu H, Yang L, Boyd DD, Wang Z 2007 A novel function of caspase-8 in the regulation of androgen-receptor-driven gene expression. EMBO J 26:65-75 

Gao S, Wang H, Lee P, Melamed J, Li CX, Zhang F, Wu H, Zhou L, Wang Z 2006 Androgen receptor and prostate apoptosis response factor-4 target the c-FLIP gene to determine survival and apoptosis in the prostate gland. J Mol Endocrinol 36:463-483 

Wu MH, Chan JY, Liu PY, Liu ST, Huang SM 2007 Human papillomavirus E2 protein associates with nuclear receptors to stimulate nuclear receptor- and E2-dependent transcriptional activations in human cervical carcinoma cells. Int J Biochem Cell Biol 39:413-425 

Wu MH, Huang CJ, Liu ST, Liu PY, Ho CL, Huang SM 2007 Physical and functional interactions of human papillomavirus E2 protein with nuclear receptor coactivators. Biochem Biophys Res Commun 356:523-528 

Wang WM, Chung MH, Huang SM 2003 Regulation of nuclear receptor activities by two human papillomavirus type 18 oncoproteins, E6 and E7. Biochem Biophys Res Commun 303:932939 

Zheng Y, Chen WL, Ma WL, Chang C, Ou JH 2007 Enhancement of gene transactivation activity of androgen receptor by hepatitis B virus X protein. Virology 363:454-461 

Chiu CM, Yeh SH, Chen PJ, Kuo TJ, Chang CJ, Chen PJ, Yang WJ, Chen DS 2007 Hepatitis B virus X protein enhances androgen receptor-responsive gene expression depending on androgen level. Proc Natl Acad Sci USA 104:2571-2578 

Lee YS, Kim HJ, Lee HJ, Lee JW, Chun SY, Ko SK, Lee K 2002 Activating signal cointegrator 1 is highly expressed in murine testicular Leydig cells and enhances the ligand-dependent transactivation of androgen receptor. Biol Reprod 67:1580-1587 

Goo YH, Na SY, Zhang H, Xu J, Hong S, Cheong J, Lee SK, Lee JW 2004 Interactions between activating signal cointegrator-2 and the tumor suppressor retinoblastoma in androgen receptor transactivation. J Biol Chem 279:7131-7135 

Wang Q, Sharma D, Ren Y, Fondell JD 2002 A coregulatory role for the TRAP-mediator complex in androgen receptor-mediated gene expression. J Biol Chem 277:42852-42858 

Yang CK, Kim JH, Li H, Stallcup MR 2006 Differential use of functional domains by coiled- coil coactivator in its synergistic coactivator function with beta-catenin or GRIP1. J Biol Chem 281:3389-3397 

Tsai TC, Lee YL, Hsiao WC, Tsao YP, Chen SL 2005 NRIP, a novel nuclear receptor interaction protein, enhances the transcriptional activity of nuclear receptors. J Biol Chem 280:20000-20009 

Zhou D, Quach KM, Yang C, Lee SY, Pohajdak B, Chen S 2000 PNRC: a proline-rich nuclear receptor coregulatory protein that modulates transcriptional activation of multiple nuclear receptors including orphan receptors SF1 (steroidogenic factor 1) and ERRalpha1 (estrogen related receptor alpha-1). Mol Endocrinol 14:986-998 

Teyssier C, Ou CY, Khetchoumian K, Losson R, Stallcup MR 2006 Transcriptional intermediary factor 1alpha mediates physical interaction and functional synergy between the coactivator-associated arginine methyltransferase 1 and glucocorticoid receptor-interacting protein 1 nuclear receptor coactivators. Mol Endocrinol 20:1276-1286 

Georgescu SP, Li JH, Lu Q, Karas RH, Brown M, Mendelsohn ME 2005 Modulator recognition factor 1, an AT-rich interaction domain family member, is a novel corepressor for estrogen receptor alpha. Mol Endocrinol 19:2491-2501 

Tomaru T, Satoh T, Yoshino S, Ishizuka T, Hashimoto K, Monden T, Yamada M, Mori M 2006 Isolation and characterization of a transcriptional cofactor and its novel isoform that bind the deoxyribonucleic acid-binding domain of peroxisome proliferator-activated receptor-gamma. Endocrinology 147:377-388 

Huang SM, Stallcup MR 2000 Mouse Zac1, a transcriptional coactivator and repressor for nuclear receptors. Mol Cell Biol 20:1855-1867 

Ko L, Cardona GR, Henrion-Caude A, Chin WW 2002 Identification and characterization of a tissue-specific coactivator, GT198, that interacts with the DNA-binding domains of nuclear receptors. Mol Cell Biol 22:357-369 

Yeh S, Chang C 1996 Cloning and characterization of a specific coactivator, ARA70, for the androgen receptor in human prostate cells. Proc Natl Acad Sci USA 93:5517-5521 

Moehren U, Papaioannou M, Reeb CA, Hong W, Baniahmad A 2007 Alien interacts with the human androgen receptor and inhibits prostate cancer cell growth. Mol Endocrinol 21:1039-1048 

Yu X, Li P, Roeder RG, Wang Z 2001 Inhibition of androgen receptor-mediated transcription by amino-terminal enhancer of split. Mol Cell Biol 21:4614-4625 

Zhu P, Baek SH, Bourk EM, Ohgi KA, Garcia-Bassets I, Sanjo H, Akira S, Kotol PF, Glass CK, Rosenfeld MG, Rose DW 2006 Macrophage/cancer cell interactions mediate hormone resistance by a nuclear receptor derepression pathway. Cell 124:615-629 

Carascossa S, Gobinet J, Georget V, Lucas A, Badia E, Castet A, White R, Nicolas JC, Cavaillès V, Jalaguier S 2006 Receptor-interacting protein 140 is a repressor of the androgen receptor activity. Mol Endocrinol 20:1506-1518 

Pero R, Lembo F, Palmieri EA, Vitiello C, Fedele M, Fusco A, Bruni CB, Chiariotti L 2002 PATZ attenuates the RNF4-mediated enhancement of androgen receptor-dependent transcription. J Biol Chem 277:3280-3285 

Sharma M, Sun Z 2001 5'TG3' interacting factor interacts with Sin3A and represses AR- mediated transcription. Mol Endocrinol 15:1918-1928 

Markus SM, Taneja SS, Logan SK, Li W, Ha S, Hittelman AB, Rogatsky I, Garabedian MJ 

2002 Identification and characterization of ART-27, a novel coactivator for the androgen receptor N terminus. Mol Biol Cell 13:670-682 

Hsiao PW, Chang C 1999 Isolation and characterization of ARA160 as the first androgen receptor N-terminal-associated coactivator in human prostate cells. J Biol Chem 274:22373-22379 

Liu Y, Kim BO, Kao C, Jung C, Dalton JT, He JJ 2004 Tip110, the human immunodeficiency virus type 1 (HIV-1) Tat-interacting protein of 110 kDa as a negative regulator of androgen receptor (AR) transcriptional activation. J Biol Chem 279:21766-21773 

Ishizuka M, Kawate H, Takayanagi R, Ohshima H, Tao RH, Hagiwara H 2005 A zinc finger protein TZF is a novel corepressor of androgen receptor. Biochem Biophys Res Commun 331:1025-1031 

Tao RH, Kawate H, Wu Y, Ohnaka K, Ishizuka M, Inoue A, Hagiwara H, Takayanagi R 

2006 Testicular zinc finger protein recruits histone deacetylase 2 and suppresses the transactivation function and intranuclear foci formation of agonist-bound androgen receptor competitively with TIF2. Mol Cell Endocrinol 247:150-165 

Tao RH, Kawate H, Ohnaka K, Ishizuka M, Hagiwara H, Takayanagi R 2006 Opposite effects of alternative TZF spliced variants on androgen receptor. Biochem Biophys Res Commun 341:515-521 

Jeong BC, Hong CY, Chattopadhyay S, Park JH, Gong EY, Kim HJ, Chun SY, Lee K 2004 Androgen receptor corepressor-19 kDa (ARR19), a leucine-rich protein that represses the transcriptional activity of androgen receptor through recruitment of histone deacetylase. 

Mol Endocrinol 18:13-25 

Ma AH, Xia L, Desai SJ, Boucher DL, Guan Y, Shih HM, Shi XB, Devere White RW, Chen HW, Tepper CG, Kung HJ 2006 Male Germ Cell-Associated Kinase, a Male-Specific Kinase Regulated by Androgen, Is a Coactivator of Androgen Receptor in Prostate Cancer Cells. Cancer Res 66:8439-8447 

Moilanen AM, Karvonen U, Poukka H, Jänne OA, Palvimo JJ 1998 Activation of androgen receptor function by a novel nuclear protein kinase. Mol Biol Cell 9:2527-2543 

Sitz JH, Tigges M, Baumgärtel K, Khaspekov LG, Lutz B 2004 Dyrk1A potentiates steroid hormone-induced transcription via the chromatin remodeling factor Arip4. Mol Cell Biol 24:58215834 

Saelzler MP, Spackman CC, Liu Y, Martinez LC, Harris JP, Abe MK 2006 ERK8 down- regulates transactivation of the glucocorticoid receptor through Hic-5. J Biol Chem 281:16821-16832 

Clark DE, Errington TM, Smith JA, Frierson HF, Weber MJ, Lannigan DA 2005 The serine/threonine protein kinase, p90 ribosomal S6 kinase, is an important regulator of prostate cancer cell proliferation. Cancer Res 65:3108-3116 

Thompson J, Lepikhova T, Teixido-Travesa N, Whitehead MA, Palvimo JJ, Jänne OA 

Small carboxyl-terminal domain phosphatase 2 attenuates androgen-dependent transcription. EMBO J 25:2757-2767 

Yang CS, Vitto MJ, Busby SA, Garcia BA, Kesler CT, Gioeli D, Shabanowitz J, Hunt DF, 

Rundell K, Brautigan DL, Paschal BM 2005 Simian virus 40 small t antigen mediates conformation-dependent transfer of protein phosphatase 2A onto the androgen receptor. Mol Cell Biol 25:1298-1308 

Powzaniuk M, McElwee-Witmer S, Vogel RL, Hayami T, Rutledge SJ, Chen F, Harada S, Schmidt A, Rodan GA, Freedman LP, Bai C 2004 The LATS2/KPM tumor suppressor is a negative regulator of the androgen receptor. Mol Endocrinol 18:2011-2023 

Li P, Nicosia SV, Bai W 2001 Antagonism between PTEN/MMAC1/TEP-1 and androgen receptor in growth and apoptosis of prostatic cancer cells. J Biol Chem 276:20444-20450 

Nan B, Snabboon T, Unni E, X-J Yuan , Whang YE, Marcelli M 2003 The PTEN tumor suppressor is a negative modulator of androgen receptor transcriptional activity. J Mol Endocrinol 31:169-183 

Lin HK, Hu YC, Lee DK, Chang C 2004 Regulation of androgen receptor signaling by PTEN (phosphatase and tensin homolog deleted on chromosome 10) tumor suppressor through distinct mechanisms in prostate cancer cells. Mol Endocrinol 18:2409-2423 

Kawate H, Wu Y, Ohnaka K, Nawata H, Takayanagi R 2005 Tob proteins suppress steroid hormone receptor-mediated transcriptional activation. Mol Cell Endocrinol 230:77-86 

Takahashi K, Taira T, Niki T, Seino C, Iguchi-Ariga SM, Ariga H 2001 DJ-1 positively regulates the androgen receptor by impairing the binding of PIASx alpha to the receptor. J Biol Chem 276:37556-37563 

Tillman JE, Yuan J, Gu G, Fazli L, Ghosh R, Flynt AS, Gleave M, Rennie PS, Kasper S 

DJ-1 binds androgen receptor directly and mediates its activity in hormonally treated prostate cancer cells. Cancer Res 67:4630-4637 

Niki T, Takahashi-Niki K, Taira T, Iguchi-Ariga SM, Ariga H 2003 DJBP: a novel DJ-1- binding protein, negatively regulates the androgen receptor by recruiting histone deacetylase complex, and DJ-1 antagonizes this inhibition by abrogation of this complex. Mol Cancer Res 1:247-261 

Wafa LA, Cheng H, Rao MA, Nelson CC, Cox M, Hirst M, Sadowski I, Rennie PS 2003 Isolation and identification of L-dopa decarboxylase as a protein that binds to and enhances transcriptional activity of the androgen receptor using the repressed transactivator yeast two-hybrid system. Biochem J 375:373-383 

Bai S, He B, Wilson EM 2005 Melanoma antigen gene protein MAGE-11 regulates androgen receptor function by modulating the interdomain interaction. Mol Cell Biol 25:1238-1257 

Lanz RB, McKenna NJ, Onate SA, Albrecht U, Wong J, Tsai SY, Tsai MJ, O'Malley BW 

1999 A steroid receptor coactivator, SRA, functions as an RNA and is present in an SRC-1 complex. Cell 97:17-27 

Kurisu T, Tanaka T, Ishii J, Matsumura K, Sugimura K, Nakatani T, Kawashima H 2006 Expression and function of human steroid receptor RNA activator in prostate cancer cells: role of endogenous hSRA protein in androgen receptor-mediated transcription. Prostate Cancer Prostatic Dis 9:173-178 

Holter E, Kotaja N, Makela S, Strauss L, Kietz S, Janne OA, Gustafsson JA, Palvimo JJ, Treuter E 2002 Inhibition of androgen receptor (AR) function by the reproductive orphan nuclear receptor DAX-1. Mol Endocrinol 16:515-528 

Yu X, Suzuki K, Wang Y, Gupta A, Jin R, Orgebin-Crist MC, Matusik R 2006 The role of forkhead box A2 to restrict androgen-regulated gene expression of lipocalin 5 in the mouse epididymis. Mol Endocrinol 20:2418-2431 

Ning YM, Robins DM 1999 AML3/CBFalpha1 is required for androgen-specific activation of the enhancer of the mouse sex-limited protein (Slp) gene. J Biol Chem 274:30624-30630 

Lobaccaro JM, Poujol N, Térouanne B, Georget V, Fabre S, Lumbroso S, Sultan C 1999 Transcriptional interferences between normal or mutant androgen receptors and the activator protein 1--dissection of the androgen receptor functional domains. Endocrinology 140:350-357 

Sato N, Sadar MD, Bruchovsky N, Saatcioglu F, Rennie PS, Sato S, Lange PH, Gleave ME 

1997 Androgenic induction of prostate-specific antigen gene is repressed by protein-protein interaction between the androgen receptor and AP-1/c-Jun in the human prostate cancer cell line LNCaP. J Biol Chem. 272:17485-17494 

Jorgensen JS, Nilson JH 2001 AR suppresses transcription of the alpha glycoprotein hormone subunit gene through protein-protein interactions with cJun and activation transcription factor 2. Mol Endocrinol 15:1496-1504 

Gonzalez MI, Tovaglieri A, Robins DM 2002 Androgen receptor interactions with Oct-1 and Brn-1 are physically and functionally distinct. Mol Cell Endocrinol 190:39-49 

Bubulya A, Wise SC, Shen XQ, Burmeister LA, Shemshedini L 1996 c-Jun can mediate androgen receptor-induced transactivation. J Biol Chem 271:24583-24589 

Chen SY, Cai C, Fisher CJ, Zheng Z, Omwancha J, Hsieh CL, Shemshedini L 2006 c-Jun enhancement of androgen receptor transactivation is associated with prostate cancer cell proliferation. Oncogene 25:7212-7223 

Mukhopadhyay NK, Ferdinand AS, Mukhopadhyay L, Cinar B, Lutchman M, Richie JP, Freeman MR, Liu BC 2006 Unraveling androgen receptor interactomes by an array-based method: discovery of proto-oncoprotein c-Rel as a negative regulator of androgen receptor. Exp Cell Res 312:3782-3795 

Chattopadhyay S, Gong EY, Hwang M, Park E, Lee HJ, Hong CY, Choi HS, Cheong JH, Kwon HB, Lee K 2006 The CCAAT enhancer-binding protein-alpha negatively regulates the transactivation of androgen receptor in prostate cancer cells. Mol Endocrinol 20:984-995 

Agoulnik IU, Krause WC, Bingman WE 3rd, Rahman HT, Amrikachi M, Ayala GE, Weigel NL 2003 Repressors of androgen and progesterone receptor action. J Biol Chem 278:3113631148 

Yang SZ, Abdulkadir SA 2003 Early growth response gene 1 modulates androgen receptor signaling in prostate carcinoma cells. J Biol Chem 278:39906-399911 

Panet-Raymond V, Gottlieb B, Beitel LK, Pinsky L, Trifiro MA 2000 Interactions between androgen and estrogen receptors and the effects on their transactivational properties. Mol Cell Endocrinol 167:139-150 

Li P, Lee H, Guo S, Unterman TG, Jenster G, Bai W 2003 AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR. Mol Cell Biol 23:104-118 

Dong XY, Chen C, Sun X, Guo P, Vessella RL, Wang RX, Chung LW, Zhou W, Dong JT 2006 FOXO1A is a candidate for the 13q14 tumor suppressor gene inhibiting androgen receptor signaling in prostate cancer. Cancer Res 66:6998-7006 

Gao N, Zhang J, Rao MA, Case TC, Mirosevich J, Wang Y, Jin R, Gupta A, Rennie PS, Matusik RJ 2003 The role of hepatocyte nuclear factor-3 alpha (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol Endocrinol 17:1484-1507 

Mirosevich J, Gao N, Gupta A, Shappell SB, Jove R, Matusik RJ 2006 Expression and role of Foxa proteins in prostate cancer. Prostate 66:1013-1028 

Chen G, Nomura M, Morinaga H, Matsubara E, Okabe T, Goto K, Yanase T, Zheng H, Lu J, Nawata H 2005 Modulation of androgen receptor transactivation by FoxH1. A newly identified androgen receptor corepressor. J Biol Chem 280:36355-36363 

Perez-Stable CM, Pozas A, Roos BA 2000 A role for GATA transcription factors in the androgen regulation of the prostate-specific antigen gene enhancer. Mol Cell Endocrinol 167:43-53 

Chen S, Wang J, Yu G, Liu W, Pearce D 1997 Androgen and glucocorticoid receptor heterodimer formation. A possible mechanism for mutual inhibition of transcriptional activity. J Biol Chem 272:14087-14092 

Jung C, Kim RS, Zhang HJ, Lee SJ, Jeng MH 2004 HOXB13 induces growth suppression of prostate cancer cells as a repressor of hormone-activated androgen receptor signaling. Cancer Res 64:9185-9192 

Darne CH, Morel L, Claessens F, Manin M, Fabre S, Veyssière G, Rombauts W, Jean CL 

1997 Ubiquitous transcription factors NF1 and Sp1 are involved in the androgen activation of the mouse vas deferens protein promoter. Mol Cell Endocrinol 132:13-23 

Préfontaine GG, Walther R, Giffin W, Lemieux ME, Pope L, Haché RJ 1999 Selective binding of steroid hormone receptors to octamer transcription factors determines transcriptional synergism at the mouse mammary tumor virus promoter. J Biol Chem 274:26713-26719 

Hong CY, Gong EY, Kim K, Suh JH, Ko HM, Lee HJ, Choi HS, Lee K 2005 Modulation of the expression and transactivation of androgen receptor by the basic helix-loop-helix transcription factor Pod-1 through recruitment of histone deacetylase 1. Mol Endocrinol 19:2245-2257 

Shenk JL, Fisher CJ, Chen SY, Zhou XF, Tillman K, Shemshedini L 2001 p53 represses androgen-induced transactivation of prostate-specific antigen by disrupting hAR amino- to carboxyl- terminal interaction. J Biol Chem 276:38472-38479 

Cronauer MV, Schulz WA, Burchardt T, Ackermann R, Burchardt M 2004 Inhibition of p53 function diminishes androgen receptor-mediated signaling in prostate cancer cell lines. Oncogene 23:3541-3549 

Oettgen P, Finger E, Sun Z, Akbarali Y, Thamrongsak U, Boltax J, Grall F, Dube A, Weiss A, Brown L, Quinn G, Kas K, Endress G, Kunsch C, Libermann TA 2000 PDEF, a novel prostate epithelium-specific ets transcription factor, interacts with the androgen receptor and activates prostate-specific antigen gene expression. J Biol Chem 275:1216-1225 

Palvimo JJ, Reinikainen P, Ikonen T, Kallio PJ, Moilanen A, Janne OA 1996 Mutual transcriptional interference between RelA and androgen receptor. J Biol Chem 271:24151-24156 

Chuang KH, Lee YF, Lin WJ, Chu CY, Altuwaijri S, Wan YJ, Chang C 2005 9-cis-retinoic acid inhibits androgen receptor activity through activation of retinoid X receptor. Mol Endocrinol 19:1200-1212 

Wang H, McKnight NC, Zhang T, Lu ML, Balk SP, Yuan X 2007 SOX9 is expressed in normal prostate basal cells and regulates androgen receptor expression in prostate cancer cells. Cancer Res 67:528-536 

Yuan X, Lu ML, Li T, Balk SP 2001 SRY interacts with and negatively regulates androgen receptor transcriptional activity. J Biol Chem 276:46647-46654 

Yuan H, Gong A, Young CY 2005 Involvement of transcription factor Sp1 in quercetin- mediated inhibitory effect on the androgen receptor in human prostate cancer cells. Carcinogenesis 26:793-801 

Gobinet J, Auzou G, Nicolas JC, Sultan C, Jalaguier S 2001 Characterization of the interaction between androgen receptor and a new transcriptional inhibitor, SHP. Biochemistry 40:15369-15377 

Jorgensen JS, Nilson JH 2001 AR suppresses transcription of the LHbeta subunit by interacting with steroidogenic factor-1. Mol Endocrinol 15:1505-1516 

Mu X, Chang C 2003 TR2 orphan receptor functions as negative modulator for androgen receptor in prostate cancer cells PC-3. Prostate 57:129-133 

Lee YF, Shyr CR, Thin TH, Lin WJ, Chang C 1999 Convergence of two repressors through heterodimer formation of androgen receptor and testicular orphan receptor-4: a unique signaling pathway in the steroid receptor superfamily. Proc Natl Acad Sci U S A 96:14724-14729 

Kivinen A, Patrikainen L, Kurkela R, Porvari K, Vihko P 2004 USF2 is connected to GAAAATATGATA element and associates with androgen receptor-dependent transcriptional regulation in prostate. Prostate 59:190-202 

Wang Q, Li W, Liu XS, Carroll JS, Jänne OA, Keeton EK, Chinnaiyan AM, Pienta KJ, Brown M 2007 A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol Cell 27:380-392 

Bolton EC, So AY, Chaivorapol C, Haqq CM, Li H, Yamamoto KR 2007 Cell- and gene- specific regulation of primary target genes by the androgen receptor. Genes Dev 21:2005-2017 

Massie CE, Adryan B, Barbosa-Morais NL, Lynch AG, Tran MG, Neal DE, Mills IG 2007 New androgen receptor genomic targets show an interaction with the ETS1 transcription factor. EMBO Rep 8:871-878 

Mukhopadhyay NK, Ferdinand AS, Mukhopadhyay L, Cinar B, Lutchman M, Richie JP, Freeman MR, Liu BC 2006 Unraveling androgen receptor interactomes by an array-based method: discovery of proto-oncoprotein c-Rel as a negative regulator of androgen receptor. Exp Cell Res 312:3782-3795 

Sarasin A, Stary A 2007 New insights for understanding the transcription-coupled repair pathway. DNA Repair (Amst) 6:265-269 

Reed SH, Gillette TG 2007 Nucleotide excision repair and the ubiquitin proteasome pathway- do all roads lead to Rome? DNA Repair (Amst) 6:149-156 

Loizou JI, Murr R, Finkbeiner MG, Sawan C, Wang ZQ, Herceg Z 2006 Epigenetic information in chromatin: the code of entry for DNA repair. Cell Cycle 5:696-701 

Hammes A, Andreassen TK, Spoelgen R, Raila J, Hubner N, Schulz H, Metzger J, Schweigert FJ, Luppa PB, Nykjaer A, Willnow TE 2005 Role of endocytosis in cellular uptake of sex steroids. Cell 122:751-762 

Häkli M, Karvonen U, Jänne OA, Palvimo JJ 2001 The RING finger protein SNURF is a bifunctional protein possessing DNA binding activity. J Biol Chem 276:23653-23660 

Cleutjens KB, van Eekelen CC, van der Korput HA, Brinkmann AO, Trapman J 1996 Two androgen response regions cooperate in steroid hormone regulated activity of the prostate-specific antigen promoter. J Biol Chem 271:6379-6388 

Cleutjens KB, van der Korput HA, van Eekelen CC, van Rooij HC, Faber PW, Trapman J 

1997 An androgen response element in a far upstream enhancer region is essential for high, androgenregulated activity of the prostate-specific antigen promoter. Mol Endocrinol 11:148-161 

Shang Y, Myers M, Brown M 2002 Formation of the androgen receptor transcription complex. Mol Cell 9:601-610 

Wang Q, Carroll JS, Brown M 2005 Spatial and temporal recruitment of androgen receptor and its coactivators involves chromosomal looping and polymerase tracking. Mol Cell 19:631-642 

Louie MC, Yang HQ, Ma AH, Xu W, Zou JX, Kung HJ, Chen HW 2003 Androgen-induced recruitment of RNA polymerase II to a nuclear receptor-p160 coactivator complex. Proc Natl Acad Sci U S A 100:2226-2230 

Alen P, Claessens F, Schoenmakers E, Swinnen JV, Verhoeven G, Rombauts W, Peeters B 1999 Interaction of the putative androgen receptor-specific coactivator ARA70/ELE1alpha with multiple steroid receptors and identification of an internally deleted ELElbeta isoform. Mol Endocrinol 13:117-128 

Johannessen M, M0ller S, Hansen T, Moens U, Van Ghelue M 2006 The multifunctional roles of the four-and-a-half-LIM only protein FHL2. Cell Mol Life Sci 63:268-284 

Agoulnik IU, Vaid A, Nakka M, Alvarado M, Bingman WE, Erdem H, Frolov A, Smith CL, Ayala GE, Ittmann MM, Weigel NL 2006 Androgens modulate expression of transcription intermediary factor 2, an androgen receptor coactivator whose expression level correlates with early biochemical recurrence in prostate cancer. Cancer Res 66:10594-10602 

Comuzzi B, Nemes C, Schmidt S, Jasarevic Z, Lodde M, Pycha A, Bartsch G, Offner F, Culig Z, Hobisch A 2004 The androgen receptor co-activator CBP is up-regulated following androgen withdrawal and is highly expressed in advanced prostate cancer. J Pathol 204:159-166 

Heemers HV, Sebo TJ, Debes JD, Regan KM, Raclaw KA, Murphy LM, Hobisch A, Culig Z, Tindall DJ 2007 Androgen deprivation increases p300 expression in prostate cancer cells. Cancer Res 67:3422-3430 

Heemers HV, Regan KM, Dehm SM, Tindall DJ 2007 Androgen induction of the androgen receptor co-activator FHL2 : evidence for a role for serum response factor in prostate cancer. Cancer Res In press 

Wu RC, Qin J, Yi P, Wong J, Tsai SY, Tsai MJ, O'Malley BW 2004 Selective phosphorylations of the SRC-3/AIB1 coactivator integrate genomic reponses to multiple cellular signaling pathways. Mol Cell 15:937-949 

Heitzer MD, DeFranco DB 2006 Hic-5/ARA55, a LIM domain-containing nuclear receptor coactivator expressed in prostate stromal cells. Cancer Res 66:7326-7333 

Smith CL, O'Malley BW 2004 Coregulator function: a key to understanding tissue specificity of selective receptor modulators. Endocr Rev 25:45-71 

Reyes JC, Barra J, Muchardt C, Camus A, Babinet C, Yaniv M 1998 Altered control of cellular proliferation in the absence of mammalian brahma (SNF2alpha). EMBO J 17:6979-6991 

Smith CL, DeVera DG, Lamb DJ, Nawaz Z, Jiang YH, Beaudet AL, O'Malley BW 2002 Genetic ablation of the steroid receptor coactivator-ubiquitin ligase, E6-AP, results in tissue-selective steroid hormone resistance and defects in reproduction. Mol Cell Biol 22:525-535 

Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai MJ, O'Malley BW 1998 Partial hormone resistance in mice with disruption of the steroid receptor coactivator-1 (SRC-1) gene. Science 279:1922-1925 

Gehin M, Mark M, Dennefeld C, Dierich A, Gronemeyer H, Chambon P 2002 The function of TIF2/GRIP1 in mouse reproduction is distinct from those of SRC-1 and p/CIP. Mol Cell Biol 22:5923-5937 

Miyamoto H, Messing EM, Chang C 2004 Androgen deprivation therapy for prostate cancer: current status and future prospects. Prostate 61:332-353 

Roy-Burman P, Tindall DJ, Robins DM, Greenberg NM, Hendrix MJ, Mohla S, Getzenberg RH, Isaacs JT, Pienta KJ 2005 Androgens and prostate cancer: are the descriptors valid? Cancer Biol Ther 4:4-5. 

Grossmann ME, Huang H, Tindall DJ 2001 Androgen receptor signaling in androgen- refractory prostate cancer. J Natl Cancer Inst 93:1687-1697 

Litvinov IV, De Marzo AM, Isaacs JT 2003 Is the Achilles' heel for prostate cancer therapy a gain of function in androgen receptor signaling? J Clin Endocrinol Metab 88:2972-2982 

Debes JD, Tindall DJ 2004 Mechanisms of androgen-refractory prostate cancer. N Engl J Med 351:1488-1490 

Titus MA, Schell MJ, Lih FB, Tomer KB, Mohler JL. 2005 Testosterone and dihydrotestosterone tissue levels in recurrent prostate cancer. Clin Cancer Res 11:4653-4657 

Heemers HV, Tindall DJ 2005 Androgen receptor coregulatory proteins as potential therapeutic targets in the treatment of prostate cancer. Curr Cancer Ther Rev 1:175-186 

Dehm SM, Tindall DJ 2006 Ligand-independent androgen receptor activity is activation function-2-independent and resistant to antiandrogens in androgen refractory prostate cancer cells. J Biol Chem 281:27882-27893 

Dehm SM, Regan KM, Schmidt, LJ, Tindall DJ 2007 Selective role of an NH2-terminal WxxLF motif for aberrant androgen receptor activation in androgen depletion-independent prostate cancer cells. Cancer Res In Press. 

Adachi M, Takayanagi R, Tomura A, Imasaki K, Kato S, Goto K, Yanase T, Ikuyama S, Nawata H 2000 Androgen-insensitivity syndrome as a possible coactivator disease. N Engl J Med 343:856-862 

Lee P, Zhu CC, Sadick NS, Diwan AH, Zhang PS, Liu JS, Prieto VG 2005 Expression of androgen receptor coactivator ARA70/ELE1 in androgenic alopecia. J Cutan Pathol 32:567-571 

Li J, Zhang D, Fu J, Huang Z, Wong J 2007 Structural and Functional Analysis of Androgen Receptor in Chromatin. Mol Endocrinol [Epub ahead of print] 

Slagsvold T, Kraus I, Fr0nsdal K, Saatcioglu F 2001 DNA binding-independent transcriptional activation by the androgen receptor through triggering of coactivators. 

J Biol Chem 276:31030-31036 

Rosenfeld MG, Lunyak VV, Glass CK 2006 Sensors and signals: acoactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response. Genes Dev 20:1405-1428 

Hu YC, Yeh S, Yeh SD, Sampson ER, Huang J, Li P, Hsu CL, Ting HJ, Lin HK, Wang L, Kim E, Ni J, Chang C 2004 Functional domain and motif analyses of androgen receptor coregulator ARA70 and its differential expression in prostate cancer. J Biol Chem 279:33438-33444