Review of the in Vivo Functions of the p160 Steroid Receptor Coactivator Family
Jianming Xu and
Qingtian Li
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
Address all correspondence and requests for reprints to: Jianming Xu, Ph.D., Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030. E-mail: jxu{at}bcm.tmc.edu.
 |
ABSTRACT
|
---|
The p160 steroid receptor coactivator (SRC) gene family contains three homologous members, which serve as transcriptional coactivators for nuclear receptors and certain other transcription factors. These coactivators interact with ligand-bound nuclear receptors to recruit histone acetyltransferases and methyltransferases to specific enhancer/promotor regions, which facilitates chromatin remodeling, assembly of general transcription factors, and transcription of target genes. This minireview summarizes our current knowledge about the molecular structures, molecular mechanisms, temporal and spatial expression patterns, and biological functions of the SRC family. In particular, this article highlights the roles of SRC-1 (NCoA-1), SRC-2 (GRIP1, TIF2, or NCoA-2) and SRC-3 (p/CIP, RAC3, ACTR, AIB1, or TRAM-1) in development, organ function, endocrine regulation, and nuclear receptor function, which are defined by characterization of the genetically manipulated animal models. Furthermore, this article also reviews our current understanding of the role of SRC-3 in breast cancer and discusses possible mechanisms for functional specificity and redundancy among SRC family members.
 |
INTRODUCTION
|
---|
STEROID HORMONES, THYROID hormones (THs), retinoids, vitamin D, prostaglandins, and bile acids play pivotal roles in the regulation of a variety of developmental events and physiological functions. The biological activities of these hormones and bioactive metabolites are mediated by their cognate nuclear receptors (NRs), which are ligand-dependent and DNA sequence-specific transcription factors. Most members of the NR superfamily contain a transcriptional activation function 1 in their N-terminal domains, a DNA binding function in their highly conserved central domains, and an activation function 2 in their C-terminal ligand binding domains. Upon hormonal binding, steroid receptors including estrogen receptors (ER
and ERß), progesterone receptors (PR-A and PR-B), androgen receptor (AR), glucocorticoid receptor (GR), and mineralocorticoid receptor change their conformations, form homodimers, bind to their cognate hormone response elements (HREs), recruit coactivators, and enhance their target gene transcription (reviewed in Refs. 1 and 2). On the other hand, NRs that form heterodimers with the retinoid X receptor, such as the thyroid hormone receptor (TR), the retinoic acid receptor (RAR), and the vitamin D receptor, bind their HREs in the absence of ligands, associate with corepressors, and inhibit transcription of their target genes. After binding to their ligands, these HRE-bound receptors release the repression function through dissociation of corepressors and gain transcriptional activation function through recruitment of coactivators (reviewed in Refs. 2 and 3).
The finding that activation of one overexpressed NR could inhibit another NRs transcriptional activity without any direct interaction or any overlapping DNA binding between these two NRs suggested that the NR transcriptional function might be mediated by limiting common coregulators (4). During the past several years, cloning methods based on protein-protein interactions between NRs and putative coregulators in the absence or presence of ligands identified a number of NR corepressors and coactivators (reviewed in Ref.2). A coactivator is usually defined according to its physical interaction with NRs in biochemical analysis and its ability to enhance NR-dependent transcription in transient transfection assays. Most individual coactivators that directly interact with NRs form distinct preexisting protein complexes with downstream intermediate factors for chromatin remodeling or for interaction with general transcription factors (Fig. 1
). These coactivator complexes can be efficiently recruited to specific promoters by ligand-activated and DNA-bound NRs or other classes of transcription factors. Since the concentration and function of each individual component in these coactivator complexes can be regulated through transcriptional control, various posttranslational modifications and degradation by multiple signaling pathways, the usage of these coactivator complexes by NRs may provide platforms for sophisticated transcriptional regulation. Among multiple coactivator complexes, the mammalian homolog of Drosophila switch defective/sucrose nonfermenter (SWI/SNF) complex containing brahma-related gene 1 and human brahma with ATPase activities can be recruited by several NRs for ATP-dependent chromatin remodeling through histone acetylation (Fig. 1
) (reviewed in Ref.5). The TR-associated protein (TRAP)/vitamin D receptor interacting protein (DRIP)/activator-recruited cofactor coactivator complex consists of more than a dozen proteins and interacts with NRs in a ligand-dependent manner via a single component referred to as PPAR-binding protein/TRAP220/DRIP205. The NR-recruited TRAP/DRIP/activator-recruited cofactor complex directly interacts with general transcription factors to coactivate target gene transcription (Fig. 1
) (reviewed in Ref.5). The activating signal cointegrator 2 complex contains a subset of trithorax group proteins, which can methylate histone H3 when recruited to specific chromatin regions by NRs (Fig. 1
) (6). The p160 steroid receptor coactivator (SRC) complex contains acetyltransferases including the cAMP response element binding protein (CREB)-binding protein (CBP), p300, and the p300/CBP-associated factor (p/CAF) and methyltransferases including coactivator-associated arginine methyltransferase 1 and protein arginine methyltransferase 1 (PRMT1). These chromatin-remodeling enzymes (acetyltransferases and methyltransferases) are recruited to promoters though interaction between NRs and p160 SRC coactivators in a ligand-dependent manner (Fig. 1
) (reviewed in Refs. 2 and 7). This minireview focuses on recent studies regarding the structural and functional characterization of the SRC family.

View larger version (36K):
[in this window]
[in a new window]
|
Fig. 1. NR-Dependent Transcription Is Mediated by Multiple Coactivator Complexes
Coactivator complexes including brahma-related gene/brahma, SRC, ASC-2, and TRAP/DRIP complexes can be recruited by ligand-bound NRs to specific chromatin regions for remodeling the chromatin, facilitating the assembly of general transcription factors (GTFs), and enhancing target gene transcription. H, Hormone; Ac, acetylation; Me, methylation.
|
|
 |
IDENTIFICATION OF THE SRC FAMILY
|
---|
Three homologous members of the SRC gene family have been identified in human and rodents. SRC-1 (full-length SRC-1 or nuclear receptor coactivator-1) was cloned through its ability to interact with the ligand-bound PR, ER, or TR (8, 9, 10). SRC-2 [glucocorticoid receptor-interacting protein 1, transcriptional intermediary factor 2 (TIF2), or nuclear receptor coactivator-2] was identified through its interactions with ligand binding domains of GR and ER (11, 12, 13). SRC-3 (p300/CBP-interacting protein, receptor-associated coactivator 3, acetyltransferase ACTR, amplified in breast cancer 1, or thyroid hormone receptor activator molecule 1) was initially identified in an amplified chromosomal region of a human breast cancer cell line and subsequently characterized as a nuclear receptor coactivator homologous to SRC-1 and SRC-2 (13, 14, 15, 16, 17, 18). All three members of the SRC family are able to interact with multiple NRs in a ligand-dependent manner and significantly enhance NR-dependent transcription (reviewed in Ref.7). In addition to NRs, members of the SRC family also interact and coactivate certain other transcription factors such as AP-1 (19), serum response factor (20), nuclear factor-
B (NF-
B) (21), and interferon-
and cAMP regulatory element-binding protein (CREB) (13). Therefore, when experimentally expressed in cultured cells, each member of the SRC family seems to serve as a general coactivator for multiple NRs and a limited number of other transcription factors. Conversely, the transcriptional activity of a specific NR can be mediated by any member of the SRC family. This functional relationship defined by in vitro experiments among various NRs and SRCs may provide a basic explanation for functional redundancy among members of the SRC family when they are coexpressed in vivo.
The amino acid sequences and biological functions of all known SRC-related proteins are evolutionarily related and relatively conserved among different species (Fig. 2
). The quail SRC-1 is 75% identical to mammalian SRC-1 proteins and highly expressed in steroid-sensitive brain regions (22). Except human and rodents, SRC-2 homologous proteins were also identified in zebrafish (zTIF2, GenBank NP_571852) and frog (xTIF2) (23). Overexpression of the NR- or CBP-binding domains of xTIF2 interfered with the biological function of xTIF2 and caused ectopic expression of Xenopus Brachyury and MyoD genes and severe developmental defects such as loss of head structures, shortened trunks, and open blastopores (23). These data suggest the existence of a NR pathway in frog that requires xTIF2 and CBP to regulate gene expression during development. SRC-3-related proteins were also identified in fruit fly and frog. In Drosophila, a coactivator for the ecdysone receptor, termed Taiman, was identified by a genetic approach and found sharing certain sequence homology to SRC-3. The highest level of amino acid sequence identity was found in the N-terminal basic helix-loop-helix (bHLH) domain of Taiman, which was 48% identical and 71% similar between amplified in breast cancer-1 and Taiman. Mutation of the Taiman gene caused defects in the migration of the border cells in the Drosophila ovary, and the mutant cells exhibited abnormal accumulation of E-cadherin, ß-catenin, and focal adhesion kinase. These findings suggest that Taiman may have a potential role in regulation of invasive cell behavior similar to the metastasis of cancer cells (24). The Xenopus SRC-3 (xSRC-3) is 72% identical to mammalian SRC-3. It was demonstrated that xSRC-3 can interact with and coactivate mammalian NRs such as retinoid X receptor, RAR, and TR (25), suggesting that the coactivator function of SRC family members in the frog is similar to that in mammals.

View larger version (30K):
[in this window]
[in a new window]
|
Fig. 2. A Homology Tree for SRC Family Members in Different Species
The homology tree was created with updated SRC-related sequences in the GenBank by using DNAMAN software (Lynnon Biosoft). According to the degrees of homology, SRC family members can be classified into four clusters. Taiman is the most distantly related member to all other SRC-related proteins. For reading the homology between any two groups or two genes, please align the vertical lines of the tree to the percentage scale bar. The first letter of each protein indicates species: h, human; m, mouse; r, rat; q, quail; x, Xenopus laevis; z, zebrafish; d, Drosophila.
|
|
 |
STRUCTURAL AND FUNCTIONAL DOMAINS OF THE SRC FAMILY
|
---|
Human and rodent proteins encoded by the SRC gene family are about 160 kDa in size and have an overall sequence similarity of 5055% and sequence identity of 4348% between the three members (Fig. 3
). Their most conserved N-terminal bHLH/Per/Ah receptor nuclear translocator (ARNT)/Sim domain was originally identified in Drosophila proteins (Per and Sim), where it is involved in DNA binding and heterodimerization between proteins containing these motifs (26). Although the bHLH/Per/ARNT/Sim domain in SRC-1 is not obligatory for coactivation of NRs in transfection assays (8), at least one study has shown that this domain is important for interaction of SRC-2 with myogenic factors including myogenin and MEF-2C (27). The relatively conserved central region of the SRC family members contains three LXXLL (L, leucine; X, any amino acid) motifs that are responsible for interaction with ligand-bound NRs (L1L3 in Fig. 3
) (13, 16, 28, 29). The LXXLL motif forms an amphipathic
-helix that binds a hydrophobic cleft formed in ligand-binding domains of NRs after binding ligands (30). Interestingly, distinct LXXLL motifs and contextual sequences exhibit differential binding affinity for different NRs, suggesting NRs have a preference for one LXXLL motif over another in the same coactivator or for one coactivator molecule over another. However, single mutation of any one of these three LXXLL motifs does not completely abolish the interaction of SRC members with NRs, suggesting that multiple LXXLL motifs are involved in the high-affinity binding of SRCs to NRs (reviewed in Ref.31).

View larger version (36K):
[in this window]
[in a new window]
|
Fig. 3. Structural and Functional Domains of the SRC Family Members
The similarity and identity of amino acid sequences for full-length human SRC proteins and their specific conserved regions are indicated above the bars. The letters within the bars indicate structural domains, and the lines under the bars indicate domains that interact with different factors or serve as transcriptional activation domains (AD) 1 and 2. PAS, Per/ARNT/Sim homologous domain; S/T, serine/threonine-rich regions; encircled L, typical LXXLL -helix motifs; boxed L, atypical LXXLL motifs (L4 is PDDLL in hSRC-2 or LDDLV in hSRC-3 and L6 is IDKLV in hSRC-1 or IPELV in hSRC-2 and hSRC-3) (31 ); Q, glutamine-rich regions; HAT, histone acetyltransferase domains identified in SRC-1 and SRC-3 (16 36 ).
|
|
There are two intrinsic transcriptional activation domains (AD1 and AD2) defined in transfection assays by tethering different regions of SRC proteins to the Gal4 DNA-binding domain. The AD1 region is responsible for interaction with the general transcriptional cointegrators, CBP and p300, but does not interact with NRs (Fig. 3
) (12, 15, 32). Interestingly, the AD1 domain also contains three LXXLL/LXXLL-like motifs (L4L6 in Fig. 3
); mutation of one or more of these motifs impairs the interaction of SRCs with the general transcriptional cointegrators, CBP and p300, as well as the activation function of SRCs, indicating that the major role of AD1 is to recruit acetyltransferases including CBP/p300 and p/CAF for chromatin remodeling (Fig. 1
) (29, 33). The second transcriptional activation domain (AD2) is located at the C terminus of SRC proteins (Fig. 3
). AD2 is responsible for interaction with histone methyltransferases, coactivator-associated arginine methyltransferase 1 and PRMT1 (34, 35). Recruitment of these histone methyltransferases to an enhancer/promoter by SRC coactivators may also be critical for NR- directed local chromatin remodeling and assembly of the transcriptional machinery around the promoter (Fig. 1
).
 |
ROLES FOR SRCs IN CHROMATIN REMODELING AND TRANSCRIPTIONAL ACTIVATION
|
---|
The C-terminal domains of SRC-1 and SRC-3 contain histone acetyltransferase (HAT) activities, raising the possibility that SRC coactivators may play a direct role in chromatin remodeling during the process of NR-directed initiation of transcription (16, 36). However, the SRC HAT activity is much weaker than those in CBP, p300, and p/CAF and inactivation of the HAT activity in SRC-1 by site-directed mutations does not significantly affect its coactivation function in a chromatin-based in vitro transcriptional assay system (36, 37). These data suggest that the intrinsic SRC HAT activity may not be essential for NR-directed initiation of transcription.
Instead, SRCs may play major roles in the chromatin remodeling and the assembly of general transcription factors through direct and indirect recruitments of other coactivators. Accumulated data support a sequential molecular mechanism for SRC function. First, the SRC preexisting complexes containing CBP, p300, p/CAF, CARM-1, and PRMT1 (specific components may be cell type dependent) are recruited to the chromatin environment through ligand binding-triggered direct interactions between NRs and SRCs, which results in site-specific acetylations and methylations of specific histones (10, 33, 34, 35, 38, 39, 40, 41, 42, 43). Second, the SWI/SNF chromatin-remodeling complex is recruited to the chromatin through direct or indirect interactions with CBP/p300, and the recruitment is stabilized by the CBP/p300-acetylated histone tails. The SWI/SNF complex contains ATPase and causes specific histone acetylations in an ATP-dependent manner, which results in changes of DNA topology (5, 44, 45). Third, the TRAP mediator complex can be recruited to the chromatin through interactions with SRC/CBP/p300 complex or direct interactions with NRs. Recent studies also showed that the recruitment of the TRAP complex by p300 is partially dependent on histone acetylation. The TRAP complex directly communicates with the basal transcriptional machinery and facilitates the initiation of gene expression (40, 45, 46, 47, 48, 49). Fourth, the entire process of NR-induced coactivator recruitment, assembly of transcription machinery, and initiation of transcription is dynamic and may happen in a cyclic fashion. For example, the ER
transcription complex appears to repeatedly cycle onto and off of target promoters in the presence of estrogen. The cycling is probably regulated by phosphorylation of the pol II C terminus, exchange of coactivators such as exchange of p300 for CBP, and covalent modifications of coregulators such as phosphorylation and acetylation of SRCs (40, 50, 51).
 |
BIOLOGICAL FUNCTIONS OF THE SRC FAMILY
|
---|
Our knowledge of the biological functions of SRC family members is based, in part, on expression patterns of mRNA and proteins and more specifically on phenotypes of genetically modified rodent models (Table 1
). For each SRC member, both aspects will be discussed.
SRC-1
The SRC-1 gene is located in chromosome 2 (p23) in humans and chromosome 12 (A23) in mice (52, 53). The SRC-1 gene is widely expressed in many tissues and cell types. SRC-1 mRNA has been detected by Northern blot in the brain, skeletal muscle, lung, kidney, pituitary, stomach, spleen, testis, heart, pancreas, liver, hypothalamus, adrenal gland, and thyroid gland (54, 55). SRC-1 protein has been identified in the testis, brain, lung, liver, kidney, and heart (13, 56). During mouse embryonic development, SRC-1 is widely expressed at embryonic d 8.5 (E8.5). At E11.5, SRC-1 is highly expressed in the neural tube. By E14.5 and E18.5, higher levels of SRC-1 expression were observed in the olfactory epithelium, brain, heart, anterior pituitary, tongue, and limb buds (57). In the adult mouse, SRC-1 remains highly expressed in certain brain regions including the olfactory bulb, hippocampus, piriform cortex, amygdala, hypothalamus, cerebellar Purkinje cells (PCs), and brainstem (58).
Despite these tissue-specific expression patterns in neuronal cells, no known human disease has been specifically linked to a genetic defect of the SRC-1 gene at this time. Likewise, mice lacking functional SRC-1 protein exhibited normal growth and fertility. However, genetic models provided important clues for partial resistance to steroid hormones due to the loss of SRC-1 function. For example, the estrogen-induced uterine growth and the estrogen- and progesterone-dependent uterine decidual response were decreased in ovariectomized female SRC-1-/- mice. Mammary gland ductal side branching and alveolar formation were also reduced in ovariectomized female SRC-1-/- mice treated with estrogen and progesterone. A similar partial response was observed from testosterone-stimulated prostate growth in the castrated male SRC-1-/- mice (56). These observations indicate that SRC-1 serves as one of the in vivo coactivators to mediate a part of the transcriptional activity for steroid receptors.
SRC-1 plays an important role in brain development and function. At birth, the gonads of male rats produce testosterone that is converted into dihydrotestosterone and estradiol in the brain. The dihydrotestosterone activates AR and results in behavioral masculinization (increase in male-typical behaviors). Estradiol activates ER and results in development of the sexually dimorphic nucleus (SDN) and behavioral defeminization (decrease in female-typical behaviors) in male rats. Infusion of testosterone into brains of female rats at the neonatal stage also results in a larger SDN and development of male sexual behavior in the adult. Interestingly, down-regulation of SRC-1 by infusion of antisense oligodeoxynucleotides (ODNs) into the androgen-treated hypothalamus of female rats at the neonatal stage significantly reduced the SDN volume and blocked behavioral defeminization. Accordingly, male and androgen-infused female rats treated with SRC-1 antisense at the neonatal stage displayed significantly higher levels of female sexual behaviors. In contrast, SRC-1 antisense treatment did not affect the masculinizing actions of neonatal testosterone treatment on male sexual behavior. These results suggest that SRC-1 plays an important role in ER-mediated SND development and behavioral defeminization but not in AR-mediated behavioral masculinization during sexual differentiation of the rat brain (59). Furthermore, infusion of antisense ODNs to SRC-1 and SRC-2 into the adult rodent brain inhibits ER-dependent PR synthesis in the ventromedial nucleus and blocks female reproductive behaviors. Intriguingly, SRC-1 knockout mice exhibit nearly normal female reproductive behaviors (60). The paradoxical observations on sexual behaviors between SRC-1 antisense-treated rodents and SRC-1 knockout mice could be due to the temporal differences in SRC-1 depletion. During development, the SRC-1 knockout mice may adapt a genetic compensatory mechanism such as up-regulation of other coactivators for maintenance of fundamental biological functions. Indeed, the level of SRC-2 mRNA is slightly elevated in the brain of SRC-1-/- mice (56). In contrast, when SRC-1 is down-regulated by acute administration of SRC-1 antisense, it could be possible that the genetic compensation has not yet been adapted before the effects of SRC-1 on sexual behaviors are observed.
In addition, SRC-1 is expressed at higher levels than other SRC family members in the cerebellar PCs, and mice lacking SRC-1 exhibit a delay in PC development during embryonic development and neonatal stages. Interestingly, the recovery of PC development by postnatal d 10 in SRC-1-/- mice correlates with an earlier and higher expression of SRC-2, suggesting a mechanism of genetic compensation from other SRC family members during development. The adult SRC-1 knockout mice also exhibit moderate motor learning dysfunction, probably due to the lack of SRC-1 in PCs or the delayed maturation of PCs (58). Since TRs and the retinoid-related orphan nuclear receptor play important roles in PC development, SRC-1 and SRC-2 may be required for normal function of retinoid-related orphan nuclear receptor and TRs in PC development and function.
Partial resistance to TH was also observed in SRC-1-/- mice as evidenced by elevated serum T3 and T4 THs and TSH. These studies demonstrated that SRC-1 is required for efficient down-regulation of TSH by T3, supporting the hypothesis that resistance to TH in humans can be caused by a defective TR coactivator such as SRC-1 (61). Interestingly, SRC-1 may be involved not only in transcriptional activation by liganded TRs, but also in the repression by liganded or unliganded TRs. For example, suppression of TSHß expression by T3 was attenuated in SRC-1 knockout mice (62). Intriguingly, multiple observations have suggested that the usage of SRC-1 by different TR isoforms is tissue specific. First, SRC-1 is important for both TR
- and TRß-mediated body growth since both TR
/SRC-1 and TRß/SRC-1 double-knockout mice exhibit more severe growth retardation than either TR
or TRß single-knockout mice. Second, SRC-1 partially mediates the TH effects on heart rates by TR
and TRß. Third, hypersensitivity to TH seen in TR
null mice, as demonstrated by overexpression of a TH-regulated gene, 5' deoiodinase, in the liver, is abolished in SRC-1-/- mice, suggesting that the hypersensitivity in TR
null mice is due to TRß function enhanced by SRC-1 (63). Fourth, SRC-1 is required for normal down-regulation of TSH by both TR
and TRß in the pituitary. In the absence of TR
, SRC-1 expression is elevated in the pituitary, suggesting that the excess amount of SRC-1 in TR
null mice may superactivate TRß and cause TH hypersensitivity for down-regulation of TSH (64).
The role of SRC-1 in peroxisomal proliferator-activated receptor-
(PPAR
) function also exhibits certain levels of tissue specificity, which is probably due to relative amounts of PPAR
, SRC family members, and other PPAR
coactivator such as PPAR
coactivator-1 (PGC-1). In the liver, SRC-1 seems not required for expression of PPAR
-regulated genes (65). In contrast, in the brown fat, activation of PPAR
triggers the recruitment of a coactivator complex containing PGC-1, SRC-1, and CBP/p300 (66). Inactivation of SRC-1 impairs the thermogenic activity of PGC-1 in the brown fat, decreases the energy expenditure, and results in obesity following a high-fat diet (67).
SRC-2
The SRC-2 gene is located in chromosome 8 (q21) in humans and chromosome 1 (A35) in mice (53, 68). SRC-2 mRNA has been detected in many tissues including placenta, testis, brain, heart, liver, pancreas, and uterus. The kidney, skeletal muscle, and mammary gland have little SRC-2 mRNA expression (12, 54, 56, 58). SRC-2 protein has also been identified in the testis, lung, brain, liver, and heart by Western blot (13). By immunohistochemistry, different levels of SRC-2 immunoreactivity have been found in epithelial cells of many tissues including gastrointestinal tract, pancreas, kidney, uterus, mammary gland, testis, prostate, lung, and adrenal gland. SRC-2 protein is also expressed in stromal cells of the colon, urinary bladder, uterus, and mammary gland and in smooth muscle cells of the gastrointestinal and urinary tracts, uterus, epididymis, prostate, and blood vessels. SRC-2 protein is undetectable in hepatocytes, thyroid gland, and striated muscle by immunohistochemistry (69). These studies indicate that SRC-2 is widely expressed in many organs and its expression amount varies between cell types and organs.
Similar to SRC-1-/- mice, SRC-2-/- mice exhibit nearly normal somatic growth. However, the fertility is significantly reduced in both male and female SRC-2 null mice. Male hypofertility is due to a decrease in sperm number, defective maturation of the spermatid acrosome, and age-dependent testicular degeneration. The male reproductive defect appears to be of Sertoli cell (SC) origin since SRC-2 is specifically expressed in SCs in the testis. The absence of SRC-2 in SCs results in lipid accumulation in SCs, germ cell apoptosis, and detachment of germ cells from SCs (70). However, it remains unclear what nuclear receptor function is impaired in the SCs without SRC-2. In agreement with the role of SRC-2 in mouse SCs, some men with oligospermic infertility possess an AR mutation from methionine to valine that disrupts the interaction between AR and SRC-2 (71). The hypofertility of female SRC-2 mutant mice is due to a placental hypoplasia caused by the absence of maternal SRC-2 in decidual stromal cells that face the developing placenta (70). In addition, although female SRC-2-/- mice exhibit normal sexual behavior, acute administration of antisense ODNs to either SRC-1 or SRC-2 into the hypothalamus efficiently blocks the sexual behavior of female wild-type rats. Although the mechanism for this discrepancy is unknown, a developmental adaptation such as alteration of other coactivator expression may have occurred in SRC-2-/- mice to support the female sexual behavior (60). These findings indicate that SRC-2 plays a critical role in reproductive behavior and functions.
Recent studies have shown that SRC-2 plays an important role in lipid metabolism and energy balance (67). In the white adipose tissue (WAT), SRC-2 serves as a coactivator for PPAR
. In SRC-2-/- mice, WAT expresses higher levels of leptin and lower levels of genes responsible for antilipolysis and fatty acid uptake and trapping, such as the perilipins, fatty acid binding protein aP2, lipoprotein lipase, and PPAR
, causing higher levels of lipolysis and a lower potential for fatty acid storage. In the brown adipose tissue (BAT), SRC-1 is a better coactivator than SRC-2 in the stabilization of the PPAR
and PGC-1 complex for transcriptional activation. The absence of SRC-2 in BAT facilitates the formation of PPAR
/PGC-1/SRC-1 complex for PPAR
-dependent transactivation. Thus, BAT lacking SRC-2 expresses higher levels of uncoupling protein 1, PGC-1, and acetyl coenzyme A oxidase, causing higher energy expenditure due to enhanced fatty acid oxidation and uncoupling of respiration. As a result, SRC-2 null mice exhibit higher body temperature under cold conditions, less fat accumulation, lower levels of fasting glycemia and triglyceride, and higher insulin sensitivity. Collectively, these mice are better able to protect themselves against obesity induced by high-fat diet or hyperphagia. Interestingly, a high-fat diet induces SRC-2 expression in both WAT and BAT and increases the ratio of SRC-2 to SRC-1, which may reflect a part of the molecular mechanisms for the enhancement of fat accumulation (67).
One study has shown that SRC-2 is expressed in proliferating myoblasts and postmitotic differentiated myotubes and potentiates skeletal muscle differentiation by acting as a critical coactivator for MEF-2- mediated transactivation (27). However, no defect in skeletal muscle development was observed in SRC-2 knockout mice (70), suggesting that SRC-2 is not essential for skeletal muscle differentiation and other SRC members may compensate for the loss of SRC-2 function in skeletal muscle development in vivo.
SRC-3
The SRC-3 gene is located in chromosome 20 (q12) in humans and chromosome 2 (H24) in mice (14, 53). Similar to other members of the family, SRC-3 is also widely expressed. Based on Northern blot analysis, SRC-3 mRNA was detected in the placenta, pancreas, lung, kidney, brain, liver, uterus, pituitary, mammary gland, and testis (16, 17, 18, 54, 56). Its protein was also found in the tissue extracts of testis, lung, liver, brain, heart, and mammary gland (13, 72). More notably, a knock-in mouse model harboring a ß-galactosidase reporter downstream of the endogenous SRC-3 promoter revealed that the mouse SRC-3 gene is mainly expressed in mammary gland epithelial cells, oocytes, vaginal epithelial layer, hepatocytes, and in the smooth muscle cells of many tissues such as blood vessels, intestines, and oviducts (72, 73). In the brain, SRC-3 expression in adult mice was mainly detected in the hippocampus and olfactory bulb (72). These results indicate that although widely expressed, SRC-3 is selectively higher in specific cell types.
Unlike SRC-1-/- and SRC-2-/- mice, SRC-3-/- mice displayed growth retardation and reduced adult body size, probably due to lower levels of IGF-I and partial tissue resistance to IGF-I (72, 74). Although the reproductive function of male SRC-3-/- mice was only slightly reduced, the development and function of the female reproductive system was abnormal (72). First, the estrogen levels were significantly lower in female SRC-3-/- mice at all ages examined, which in turn caused a delay in pubertal development evidenced by the postponed onset of vaginal opening and mammary gland growth (72). Second, mammary gland alveolar development in response to a combined stimulation of estrogen and progesterone was significantly decreased in adult SRC-3-/- females, suggesting that SRC-3 is involved in progesterone-stimulated cell proliferation and glandular differentiation during breast alveolar development. Third, the ovulation capacity of female SRC-3-/- mice after receiving pregnant mares serum gonadotropin and human chorionic gonadotropin treatments was significantly reduced (72). Because ovulated oocytes were efficiently fertilized when mating with fertile males, the reduction in ovulation capacity may be responsible for the dramatic decrease in total pups produced by the SRC-3-/- females when paired with fertile males for a period of 1 yr (75). The decrease in ovulation could be attributed to poor follicular development because of estrogen inefficiency or to a partial block of oocyte maturation because of the absence of SRC-3 in the oocytes. Collectively, these results indicate that the major physiological functions of SRC-3 are distinct from those of SRC-1 and SRC-2.
SRC-3 is coexpressed with ER
and ERß in the endothelial cells and vascular smooth muscle cells and may facilitate the estrogen-mediated vasoprotective effects, through inhibition of neointimal formation after vessel injury (73, 76). For example, the neointimal growth in ovariectomized wild-type mice was almost completely inhibited by estrogen treatment, but only partially inhibited in ovariectomized SRC-3-/- mice due to an insufficient suppression of vascular cell proliferation by estrogen (73).
Recently, SRC-3 was found to be associated with the I
B kinase (77). I
B kinase could phosphorylate SRC-3 and promote nuclear localization of SRC-3. In addition, SRC-3 was able to augment NF-
B-mediated gene expression while the expression of interferon-regulatory factor 1, a well known NF-
B target gene, was reduced in SRC-3-/- mice (77). These observations support a former discovery that SRC-3 is a NF-
B coactivator (78), suggesting that SRC-3 may play an important role in inflammatory and immune responses as well as cell survival mediated by NF-
B.
Functional Redundancy and Specificity Among SRC Family Members
Protein-protein interactions and in vitro or ex vivo transcriptional assays have made it clear that SRC family members possess many common features that permit interaction with and coactivation of NRs when they exist in excess amounts in the assay systems (8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 37). The viable phenotypes of knockout mice lacking functional SRC-1, SRC-2, or SRC-3 further support the notion that SRC family members may be able to partially compensate each others function in vivo (56, 70, 72).
Indeed, several observations clearly demonstrate that members of the SRC family can partially compensate each others biological functions, probably due to their similar structural and functional domains as well as their partially overlapping expression patterns in certain tissues. These observations include: expression of SRC-2 is elevated in several regions of the brain and some other tissues in SRC-1-/- mice (56, 58); developmental problems exist in the SRC-1-deficient cerebellar PCs where SRC-1 is expressed at high levels and SRC-2 at low levels in wild-type mice, but no developmental problem presents in the SRC-1-deficient hippocampus and olfactory bulb where all SCRs are expressed in wild type mice (58); high levels of SRC-2, low levels of SRC-1 and undetectable levels of SRC-3 are present in the testicular SCs and only SCs of SRC-2-/- mice are incapable of supporting spermatogenesis, probably due to low, noncompensatory amounts of SRC-1 and SRC-3 (70); the majority of SRC-1/SRC-2 double-knockout mice can not survive after birth, and the few survivors are much more resistant to TH as compared with single-mutant mice (75, 79); and down-regulation of both SRC-1 and SRC-2 is required to block estrogen-induced PR synthesis in the rat hypothalamus (60). These results indicate that some biological functions are dependent on the total amount of SRC family members.
However, several studies have demonstrated the existence of certain levels of specificities between different NRs and SRCs that might allow selective uses of distinct downstream coactivators and, therefore, selective activation of target genes for different NRs. For example, microinjection of expression plasmids for SRC-1 or SRC-2, but not SRC-3, was shown to rescue RAR-dependent transcription in SRC-1-immunodepleted cells, suggesting that SRC-3 can not compensate for the function of SRC-1 and SRC-2 in these assays (13). Another study showed that SRC-3 enhances ER
- and PR-dependent gene transcription, but stimulation of ERß-mediated transcription was not observed (18). More interestingly, a recent study demonstrated that PR interacts preferentially with SRC-1 in breast cancer cells, which recruits CBP and enhances acetylation of histone H4. In contrast, GR interacts preferentially with SRC-2, which recruits p/CAF and results in histone H3 acetylation. This study suggests that selective recruitment of SRCs by different NRs may determine the specific assembly of coactivator complexes to mediate specific transcription signals (42). In addition, variable tissue-specific expression patterns of SRC family members may be also responsible for their functional specificities. Although members of the SRC family are widely expressed, their expression levels are tissue and cell type dependent. The differences in temporal and spatial expression patterns of SRC members may explain, at least in part, the discrepancies between in vitro experiments where all SRC members enhance most NR-dependent transcriptions and in vivo experiments where SRC-1, SRC-2, and SRC-3 knockout mice exhibit different phenotypes (refer to the preceding sections).
SRC-3 and Breast Cancer
Since SRC-3 can enhance ER- and PR-dependent transcription, it was reasoned that altered SRC-3 expression might play a critical role in hormone-dependent cancers such as breast cancer (14, 80). Indeed, variable amplification frequencies (4.89.5%) of the SRC-3 gene have been reported in human breast tumors (14, 80). SRC-3 mRNA is also overexpressed in 3164% of breast tumors (14, 81). However, elevated levels of the SRC-3 protein may exist in only about 10% of breast tumors (82). Interestingly, overexpressed SRC-3 was detected in breast tumors positive and negative for ER and PR (80, 81). More importantly, SRC-3 overexpression in invasive breast tumors is correlated with high levels of human epidermal growth factor receptor 2 (HER2)/neu (81, 83). In tamoxifen-treated patients, high levels of SRC-3 expression are associated with tamoxifen resistance and worse survival rate. Patients with high levels of both SRC-3 and HER2 exhibit the worst responses to tamoxifen therapy (83). Since HER2 activates MAPK, which in turn phosphorylates ER and SRC-3 (84), the overexpression of both HER2 and SRC-3 may significantly enhance the agonist activity of tamoxifen and, therefore, reduce the antitumor activity of tamoxifen in patients with breast cancer (85).
In breast cancer cells, SRC-3 can be recruited to the estrogen-responsive cyclin D1 promoter to enhance cyclin D1 expression (86). Accordingly, depletion of SRC-3 in MCF-7 breast cancer cells significantly reduced the estrogen-mediated cell proliferation and inhibition of apoptosis. Down-regulation of SRC-3 in MCF-7 cells also reduced estrogen-dependent colony formation in soft agar and tumor growth in nude mice (87). Similar to the effect of SRC-3, depletion of SRC-1 or SRC-2 also inhibited the expression of estrogen-responsive genes and estrogen-dependent cell proliferation (88). More importantly, mammary gland tumorigenesis induced by expression of the mouse mammary tumor virus-v-Ha-ras transgene in the mammary epithelial cells can be significantly suppressed in SRC-3 knockout mice (75). These findings indicate that SRC-3, together with other coactivators, may play a permissive role in breast cancer initiation and progression. Selective inhibition of SRC-3 function in the mammary gland may be a useful approach for prevention and treatment of breast cancer.
 |
CONCLUDING REMARKS
|
---|
Although characterization of the genetically manipulated animal models and cell lines with altered gene expression has uncovered the basic biological functions and dysfunctions of the SRC family, a number of important issues remain to be addressed. First, the specific receptor preference for each SRC member needs to be further defined at natural concentrations of NRs and SRCs in cell types regulated by cognate hormones. Most former analyses were done under conditions with overexpression of NRs and coactivators in heterogenetic cell types, which made the experimental results difficult to extrapolate to physiological conditions. Based on the knowledge obtained from natural cellular conditions, physiological partnerships between specific NRs and SRC members should be further characterized. Second, the spatial expression levels of SRC members in different cell types need to be quantitatively measured because the relative amounts of SRC proteins in a cell type may contribute to their functional specificity and redundancy. Third, functional specificity and redundancy among SRC members need to be further studied by generation and characterization of combinatorial knockout mice for SRC-1, SRC-2, and SRC-3 and by other genetic approaches such as expression of chimeric SRC molecules and replacement of one SRC for another in mice. Fourth, the direct and indirect molecular mechanisms responsible for the phenotypes observed in SRC knockout mice need to be characterized to understand the network of gene regulation in which SRCs are involved. Fifth, more genetic screening should be performed to search for possible human diseases caused by SRC mutations. Except for the examination of SRC-3 levels in cancer patients, it may be important to check SRC-1 mutations in patients with partial hormone resistance, SRC-2 mutations in patients with hypofertility and obesity, and SRC-3 mutations in patients with lower IGF-I, growth retardation, lower estrogen level, and female hypofertility. Sixth, the role of SRC-3 in breast cancer initiation and progression and its responsible molecular mechanisms should be further studied before SRC-3 can be used as a target for breast cancer prevention and treatment. We believe that these questions and future studies will further advance our knowledge about the biological and pathological roles of the SRC family as well as the molecular mechanisms for SRC physiological functions and dysfunctions.
 |
ACKNOWLEDGMENTS
|
---|
We thank Dr. Bert W. OMalley, Dr. JoAnne Richards, Dr. Jeffrey Rosen, and Dr. Dennis Dowhan for discussions and corrections. We apologize to our colleagues whose contributions could not be cited due to page limitation.
 |
FOOTNOTES
|
---|
This work was partially supported by research grants from The National Institutes of Health, US Department of the Army, and the Andrew W. Mellon Foundation to J.X.
Abbreviations: AD, Activation domain; AR, androgen receptor; ARNT, Ah receptor nuclear translocator; BAT, brown adipose tissue; bHLH, basic helix-loop-helix; CBP, cAMP response element binding protein (CREB)-binding protein; DRIP, vitamin D receptor-interacting protein; ER, estrogen receptor; GR, glucocorticoid receptor; HAT, histone acetyltransferase; HER2, human epidermal growth factor receptor 2; HRE, hormone response element; NcoA, nuclear coactivator; NF-
B, nuclear factor-
B; NR, nuclear receptor; ODN, antisense oligodeoxynucleotide; PC, Purkinje cell; p/CAF, p300/CBP-associated factor; PGC-1, PPAR
coactivator-1; PPAR, peroxisomal proliferator-activated receptor; PR, progesterone receptor; PRMT1, protein arginine methyltransferase 1; RAR, retinoic acid receptor; SC, Sertoli cell; SDN, sexually dimorphic nucleus; SRC, steroid receptor coactivator; SWI/SNF, switch defective/sucrose nonfermenter; TH, thyroid hormone; TIF, transcriptional intermediary factor; TR, thyroid hormone receptor; TRAP, TR-associated protein; WAT, white adipose tissue.
Received for publication April 2, 2003.
Accepted for publication June 4, 2003.
 |
REFERENCES
|
---|
- Tsai MJ, OMalley BW 1994 Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem 63:451486[CrossRef][Medline]
- McKenna NJ, OMalley BW 2002 Combinatorial control of gene expression by nuclear receptors and coregulators. Cell 108:465474[Medline]
- Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM 1995 The nuclear receptor superfamily: the second decade. Cell 83:835839[Medline]
- Meyer ME, Gronemeyer H, Turcotte B, Bocquel MT, Tasset D, Chambon P 1989 Steroid hormone receptors compete for factors that mediate their enhancer function. Cell 57:433442[Medline]
- Rosenfeld MG, Glass CK 2001 Coregulator codes of transcriptional regulation by nuclear receptors. J Biol Chem 276:3686536868[Free Full Text]
- Goo YH, Sohn YC, Kim DH, Kim SW, Kang MJ, Jung DJ, Kwak E, Barlev NA, Berger SL, Chow VT, Roeder RG, Azorsa DO, Meltzer PS, Suh PG, Song EJ, Lee KJ, Lee YC, Lee JW 2003 Activating signal cointegrator 2 belongs to a novel steady-state complex that contains a subset of trithorax group proteins. Mol Cell Biol 23:140149[Abstract/Free Full Text]
- Xu J, OMalley BW 2002 Molecular mechanisms and cellular biology of the steroid receptor coactivator (SRC) family in steroid receptor function. Rev Endocr Metab Disord 3:185192[CrossRef][Medline]
- Onate SA, Tsai SY, Tsai MJ, OMalley BW 1995 Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 270:13541357[Abstract]
- Takeshita A, Yen PM, Misiti S, Cardona GR, Liu Y, Chin WW 1996 Molecular cloning and properties of a full-length putative thyroid hormone receptor coactivator. Endocrinology 137:35943597[Abstract]
- Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, Lin SC, Heyman RA, Rose DW, Glass CK, Rosenfeld MG 1996 A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors. Cell 85:403414[Medline]
- 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 93:49484952[Abstract/Free Full Text]
- Voegel JJ, Heine MJ, Zechel C, Chambon P, Gronemeyer H 1996 TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. EMBO J 15:36673675[Abstract]
- Torchia J, Rose DW, Inostroza J, Kamei Y, Westin S, Glass CK, Rosenfeld MG 1997 The transcriptional co-activator p/CIP binds CBP and mediates nuclear-receptor function. Nature 387:677684[CrossRef][Medline]
- Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan XY, Sauter G, Kallioniemi OP, Trent JM, Meltzer PS 1997 AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science 277:965968[Abstract/Free Full Text]
- Li H, Gomes PJ, Chen JD 1997 RAC3, a steroid/nuclear receptor-associated coactivator that is related to SRC-1 and TIF2. Proc Natl Acad Sci USA 94:84798484[Abstract/Free Full Text]
- Chen H, Lin RJ, Schiltz RL, Chakravarti D, Nash A, Nagy L, Privalsky ML, Nakatani Y, Evans RM 1997 Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300. Cell 90:569580[Medline]
- Takeshita A, Cardona GR, Koibuchi N, Suen CS, Chin WW 1997 TRAM-1, A novel 160-kDa thyroid hormone receptor activator molecule, exhibits distinct properties from steroid receptor coactivator-1. J Biol Chem 272:2762927634[Abstract/Free Full Text]
- Suen CS, Berrodin TJ, Mastroeni R, Cheskis BJ, Lyttle CR, Frail DE 1998 A transcriptional coactivator, steroid receptor coactivator-3, selectively augments steroid receptor transcriptional activity. J Biol Chem 273:2764527653[Abstract/Free Full Text]
- Lee SK, Kim HJ, Na SY, Kim TS, Choi HS, Im SY, Lee JW 1998 Steroid receptor coactivator-1 coactivates activating protein-1-mediated transactivations through interaction with the c-Jun and c-Fos subunits. J Biol Chem 273:1665116654[Abstract/Free Full Text]
- Kim HJ, Kim JH, Lee JW 1998 Steroid receptor coactivator-1 interacts with serum response factor and coactivates serum response element-mediated transactivations. J Biol Chem 273:2856428567[Abstract/Free Full Text]
- Na SY, Lee SK, Han SJ, Choi HS, Im SY, Lee JW 1998 Steroid receptor coactivator-1 interacts with the p50 subunit and coactivates nuclear factor
B-mediated transactivations. J Biol Chem 273:1083110834[Abstract/Free Full Text]
- Charlier TD, Lakaye B, Ball GF, Balthazart J 2002 Steroid receptor coactivator SRC-1 exhibits high expression in steroid-sensitive brain areas regulating reproductive behaviors in the quail brain. Neuroendocrinology 76:297315[CrossRef][Medline]
- de la Calle-Mustienes E, Gomez-Skarmeta JL 2000 XTIF2, a Xenopus homologue of the human transcription intermediary factor, is required for a nuclear receptor pathway that also interacts with CBP to suppress Brachyury and XMyoD. Mech Dev 91:119129[CrossRef][Medline]
- Bai J, Uehara Y, Montell DJ 2000 Regulation of invasive cell behavior by taiman, a Drosophila protein related to AIB1, a steroid receptor coactivator amplified in breast cancer. Cell 103:10471058[Medline]
- Kim HJ, Lee SK, Na SY, Choi HS, Lee JW 1998 Molecular cloning of xSRC-3, a novel transcription coactivator from Xenopus, that is related to AIB1, p/CIP, and TIF2. Mol Endocrinol 12:10381047[Abstract/Free Full Text]
- Huang ZJ, Edery I, Rosbash M 1993 PAS is a dimerization domain common to Drosophila period and several transcription factors. Nature 364:259262[CrossRef][Medline]
- Chen SL, Dowhan DH, Hosking BM, Muscat GE 2000 The steroid receptor coactivator, GRIP-1, is necessary for MEF-2C-dependent gene expression and skeletal muscle differentiation. Genes Dev 14:12091228[Abstract/Free Full Text]
- Heery DM, Kalkhoven E, Hoare S, Parker MG 1997 A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature 387:733736[CrossRef][Medline]
- Voegel JJ, Heine MJ, Tini M, Vivat V, Chambon P, Gronemeyer H 1998 The coactivator TIF2 contains three nuclear receptor-binding motifs and mediates transactivation through CBP binding-dependent and -independent pathways. EMBO J 17:507519[Abstract/Free Full Text]
- Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA, Greene GL 1998 The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 95:927937[Medline]
- Leo C, Chen JD 2000 The SRC family of nuclear receptor coactivators. Gene 245:111[CrossRef][Medline]
- Onate SA, Boonyaratanakornkit V, Spencer TE, Tsai SY, Tsai MJ, Edwards DP, OMalley BW 1998 The steroid receptor coactivator-1 contains multiple receptor interacting and activation domains that cooperatively enhance the activation function 1 (AF1) and AF2 domains of steroid receptors. J Biol Chem 273:1210112108[Abstract/Free Full Text]
- Li J, OMalley BW, Wong J 2000 p300 requires its histone acetyltransferase activity and SRC-1 interaction domain to facilitate thyroid hormone receptor activation in chromatin. Mol Cell Biol 20:20312042[Abstract/Free Full Text]
- 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 284:21742177[Abstract/Free Full Text]
- Koh SS, Chen D, Lee YH, Stallcup MR 2001 Synergistic enhancement of nuclear receptor function by p160 coactivators and two coactivators with protein methyltransferase activities. J Biol Chem 276:10891098[Abstract/Free Full Text]
- Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J, Mizzen CA, McKenna NJ, Onate SA, Tsai SY, Tsai MJ, OMalley BW 1997 Steroid receptor coactivator-1 is a histone acetyltransferase. Nature 389:194198[CrossRef][Medline]
- Liu Z, Wong J, Tsai SY, Tsai MJ, OMalley BW 2001 Sequential recruitment of steroid receptor coactivator-1 (SRC-1) and p300 enhances progesterone receptor- dependent initiation and reinitiation of transcription from chromatin. Proc Natl Acad Sci USA 98:1242612431[Abstract/Free Full Text]
- Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T, Schulman IG, Juguilon H, Montminy M, Evans RM 1996 Role of CBP/P300 in nuclear receptor signalling. Nature 383:99103[CrossRef][Medline]
- Sheppard HM, Harries JC, Hussain S, Bevan C, Heery DM 2001 Analysis of the steroid receptor coactivator 1 (SRC1)-CREB binding protein interaction interface and its importance for the function of SRC1. Mol Cell Biol 21:3950[Abstract/Free Full Text]
- Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M 2000 Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell 103:843852[Medline]
- Demarest SJ, Martinez-Yamout M, Chung J, Chen H, Xu W, Dyson HJ, Evans RM, Wright PE 2002 Mutual synergistic folding in recruitment of CBP/p300 by p160 nuclear receptor coactivators. Nature 415:549553[CrossRef][Medline]
- Li X, Wong J, Tsai SY, Tsai MJ, OMalley BW 2003 Progesterone and glucocorticoid receptors recruit distinct coactivator complexes and promote distinct patterns of local chromatin modification. Mol Cell Biol 23:37633773[Abstract/Free Full Text]
- Lee YH, Koh SS, Zhang X, Cheng X, Stallcup MR 2002 Synergy among nuclear receptor coactivators: selective requirement for protein methyltransferase and acetyltransferase activities. Mol Cell Biol 22:36213632[Abstract/Free Full Text]
- Dilworth FJ, Fromental-Ramain C, Yamamoto K, Chambon P 2000 ATP-driven chromatin remodeling activity and histone acetyltransferases act sequentially during transactivation by RAR/RXR in vitro. Mol Cell 6: 10491058
- 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 22:2146155[Abstract/Free Full Text]
- Fondell JD, Ge H, Roeder RG 1996 Ligand induction of a transcriptionally active thyroid hormone receptor coactivator complex. Proc Natl Acad Sci USA 93:83298333[Abstract/Free Full Text]
- Rachez C, Suldan Z, Ward J, Chang CP, Burakov D, Erdjument-Bromage H, Tempst P, Freedman LP 1998 A novel protein complex that interacts with the vitamin D3 receptor in a ligand-dependent manner and enhances VDR transactivation in a cell-free system. Genes Dev 12:17871800[Abstract/Free Full Text]
- Sharma D, Fondell JD 2002 Ordered recruitment of histone acetyltransferases and the TRAP/Mediator complex to thyroid hormone-responsive promoters in vivo. Proc Natl Acad Sci USA 99:79347939[Abstract/Free Full Text]
- Ito M, Roeder RG 2001 The TRAP/SMCC/Mediator complex and thyroid hormone receptor function. Trends Endocrinol Metab 12:127134[CrossRef][Medline]
- Chen H, Lin RJ, Xie W, Wilpitz D, Evans RM 1999 Regulation of hormone-induced histone hyperacetylation and gene activation via acetylation of an acetylase. Cell 98:675686[Medline]
- Rowan BG, Weigel NL, OMalley BW 2000 Phosphorylation of steroid receptor coactivator-1. Identification of the phosphorylation sites and phosphorylation through the mitogen-activated protein kinase pathway. J Biol Chem 275:44754483[Abstract/Free Full Text]
- Carapeti M, Aguiar RC, Chase A, Goldman JM, Cross NC 1998 Assignment of the steroid receptor coactivator-1 (SRC-1) gene to human chromosome band 2p23. Genomics 52:242244[CrossRef][Medline]
- Ning G, Jurecic V, Baldini A, Xu J 1999 Structure and chromosomal locations of mouse steroid receptor coactivator gene family. In Vitro Cell Dev Biol Anim 35:481486[Medline]
- Li H, Chen JD 1998 The receptor-associated coactivator 3 activates transcription through CREB-binding protein recruitment and autoregulation. J Biol Chem 273:59485954[Abstract/Free Full Text]
- Misiti S, Schomburg L, Yen PM, Chin WW 1998 Expression and hormonal regulation of coactivator and corepressor genes. Endocrinology 139:24932500[Abstract/Free Full Text]
- Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai MJ, OMalley BW 1998 Partial hormone resistance in mice with disruption of the steroid receptor coactivator-1 (SRC-1) gene. Science 279:19221925[Abstract/Free Full Text]
- Misiti S, Koibuchi N, Bei M, Farsetti A, Chin WW 1999 Expression of steroid receptor coactivator-1 mRNA in the developing mouse embryo: a possible role in olfactory epithelium development. Endocrinology 140:19571960[Abstract/Free Full Text]
- Nishihara E, Yoshida-Komiya H, Chan CS, Liao L, Davis RL, OMalley BW, Xu J 2003 SRC-1 null mice exhibit moderate motor dysfunction and delayed development of cerebellar Purkinje cells. J Neurosci 23:213222[Abstract/Free Full Text]
- Auger AP, Tetel MJ, McCarthy MM 2000 Steroid receptor coactivator-1 (SRC-1) mediates the development of sex-specific brain morphology and behavior. Proc Natl Acad Sci USA 97:75517555[Abstract/Free Full Text]
- Apostolakis EM, Ramamurphy M, Zhou D, Onate S, OMalley BW 2002 Acute disruption of select steroid receptor coactivators prevents reproductive behavior in rats and unmasks genetic adaptation in knockout mice. Mol Endocrinol 16:15111523[Abstract/Free Full Text]
- Weiss RE, Xu J, Ning G, Pohlenz J, OMalley BW, Refetoff S 1999 Mice deficient in the steroid receptor co-activator 1 (SRC-1) are resistant to thyroid hormone. EMBO J 18:19001904[Abstract/Free Full Text]
- Takeuchi Y, Murata Y, Sadow P, Hayashi Y, Seo H, Xu J, OMalley BW, Weiss RE, Refetoff S 2002 Steroid receptor coactivator-1 deficiency causes variable alterations in the modulation of T3-regulated transcription of genes in vivo. Endocrinology 143:13461352[Abstract/Free Full Text]
- Sadow PM, Chassande O, Gauthier K, Samarut J, Xu J, OMalley BW, Weiss RE 2003 Specificity of thyroid hormone receptor subtype and steroid receptor coactivator-1 on thyroid hormone action. Am J Physiol 284:E36E46
- Sadow PM, Koo E, Chassande O, Gauthier K, Samarut J, Xu J, OMalley BW, Seo H, Murata Y, Weiss RE 2003 Thyroid hormone receptor-specific interactions with steroid receptor coactivator (SRC) -1 in the pituitary. Mol Endocrinol 17:882894[Abstract/Free Full Text]
- Qi C, Zhu Y, Pan J, Yeldandi AV, Rao MS, Maeda N, Subbarao V, Pulikuri S, Hashimoto T, Reddy JK 1999 Mouse steroid receptor coactivator-1 is not essential for peroxisome proliferator-activated receptor
-regulated gene expression. Proc Natl Acad Sci USA 96:15851590[Abstract/Free Full Text]
- Puigserver P, Adelmant G, Wu Z, Fan M, Xu J, OMalley B, Spiegelman BM 1999 Activation of PPAR
coactivator-1 through transcription factor docking. Science 286:13681371[Abstract/Free Full Text]
- Picard F, Gehin M, Annicotte J, Rocchi S, Champy MF, OMalley BW, Chambon P, Auwerx J 2002 SRC-1 and TIF2 control energy balance between white and brown adipose tissues. Cell 111:931941[Medline]
- Kalkhoven E, Valentine JE, Heery DM, Parker MG 1998 Isoforms of steroid receptor co-activator 1 differ in their ability to potentiate transcription by the oestrogen receptor. EMBO J 17:232243[Abstract/Free Full Text]
- Puustinen R, Sarvilinna N, Manninen T, Tuohimaa P, Ylikomi T 2001 Localization of glucocorticoid receptor interacting protein 1 in murine tissues using two novel polyclonal antibodies. Eur J Endocrinol 145:323333[Medline]
- 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 22:59235937[Abstract/Free Full Text]
- Ghadessy FJ, Lim J, Abdullah AA, Panet-Raymond V, Choo CK, Lumbroso R, Tut TG, Gottlieb B, Pinsky L, Trifiro MA, Yong EL 1999 Oligospermic infertility associated with an androgen receptor mutation that disrupts interdomain and coactivator (TIF2) interactions. J Clin Invest 103:15171525[Abstract/Free Full Text]
- Xu J, Liao L, Ning G, Yoshida-Komiya H, Deng C, OMalley BW 2000 The steroid receptor coactivator SRC-3 (p/CIP/RAC3/AIB1/ACTR/TRAM-1) is required for normal growth, puberty, female reproductive function, and mammary gland development. Proc Natl Acad Sci USA 97:63796384[Abstract/Free Full Text]
- Yuan Y, Liao L, Tulis DA, Xu J 2002 Steroid receptor coactivator-3 is required for inhibition of neointima formation by estrogen. Circulation 105:26532659[Abstract/Free Full Text]
- Wang Z, Rose DW, Hermanson O, Liu F, Heiman T, Wu W, Szeto D, Glieberman A, Krones A, Pratt K, Rosenfeld R, Glass CK, Rosenfeld MG 2000 Regulation of somatic growth by the p160 coactivator p/CIP. Proc Natl Acad Sci USA 97:1354913554[Abstract/Free Full Text]
- Liao L, Kuang SQ, Yuan Y, Gonzalez SM, OMalley BW, Xu J 2002 Molecular structure and biological function of the cancer-amplified nuclear receptor coactivator SRC-3/AIB1. J Steroid Biochem Mol Biol 83:314[CrossRef][Medline]
- Mendelsohn ME, Karas RH 1999 The protective effects of estrogen on the cardiovascular system. N Engl J Med 340:18011811[Free Full Text]
- Wu RC, Qin J, Hashimoto Y, Wong J, Xu J, Tsai SY, Tsai MJ, OMalley BW 2002 Regulation of SRC-3 (pCIP/ACTR/AIB-1/RAC-3/TRAM-1) coactivator activity by I
B kinase. Mol Cell Biol 22:35493561[Abstract/Free Full Text]
- Werbajh S, Nojek I, Lanz R, Costas MA 2000 RAC-3 is a NF-
B coactivator. FEBS Lett 485:195199[CrossRef][Medline]
- Weiss RE, Gehin M, Xu J, Sadow PM, OMalley BW, Chambon P, Refetoff S 2002 Thyroid function in mice with compound heterozygous and homozygous disruptions of SRC-1 and TIF-2 coactivators: evidence for haploinsufficiency. Endocrinology 143:15541557[Abstract/Free Full Text]
- Bautista S, Valles H, Walker RL, Anzick S, Zeillinger R, Meltzer P, Theillet C 1998 In breast cancer, amplification of the steroid receptor coactivator gene AIB1 is correlated with estrogen and progesterone receptor positivity. Clin Cancer Res 4:29252929[Abstract]
- Bouras T, Southey MC, Venter DJ 2001 Overexpression of the steroid receptor coactivator AIB1 in breast cancer correlates with the absence of estrogen and progesterone receptors and positivity for p53 and HER2/neu. Cancer Res 61:903907[Abstract/Free Full Text]
- List HJ, Reiter R, Singh B, Wellstein A, Riegel AT 2001 Expression of the nuclear coactivator AIB1 in normal and malignant breast tissue. Breast Cancer Res Treat 68:2128[CrossRef][Medline]
- Osborne CK, Bardou V, Hopp TA, Chamness GC, Hilsenbeck SG, Fuqua SA, Wong J, Allred DC, Clark GM, Schiff R 2003 Role of the estrogen receptor coactivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in breast cancer. J Natl Cancer Inst 95:353361[Abstract/Free Full Text]
- Font de Mora J, Brown M 2000 AIB1 is a conduit for kinase-mediated growth factor signaling to the estrogen receptor. Mol Cell Biol 20:50415047[Abstract/Free Full Text]
- Smith CL, Nawaz Z, OMalley BW 1997 Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4-hydroxytamoxifen. Mol Endocrinol 11:657666[Abstract/Free Full Text]
- Planas-Silva MD, Shang Y, Donaher JL, Brown M, Weinberg RA 2001 AIB1 enhances estrogen-dependent induction of cyclin D1 expression. Cancer Res 61:38583862[Abstract/Free Full Text]
- List HJ, Lauritsen KJ, Reiter R, Powers C, Wellstein A, Riegel AT 2001 Ribozyme targeting demonstrates that the nuclear receptor coactivator AIB1 is a rate-limiting factor for estrogen-dependent growth of human MCF-7 breast cancer cells. J Biol Chem 276:2376323768[Abstract/Free Full Text]
- Cavarretta IT, Mukopadhyay R, Lonard DM, Cowsert LM, Bennett CF, OMalley BW, Smith CL 2002 Reduction of coactivator expression by antisense oligodeoxynucleotides inhibits ER
transcriptional activity and MCF-7 proliferation. Mol Endocrinol 16:253270[Abstract/Free Full Text]