minireview_ role of orphan nuclear receptors in cancer and potential as drug targets

Upload: franciscrick69

Post on 06-Jul-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    1/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 1/36

    Mol Endocrinol. 2014 Feb; 28(2): 157–172.

    Published online 2013 Dec 2. doi: 10.1210/me.2013-1291

    PMCID: PMC3896638

    Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    Stephen Safe, Un-Ho Jin, Erik Hedrick, Alexandra Reeder , and Syng-Ook Lee

    Department of Veterinary Physiology and Pharmacology (S.S., E.H., A.R.), Texas A&M University, College Station, Texas 77808; and Institute of Biosciences and Technology (S.S.,

    U.-H.J., S.-O.L.), Texas A&M Health Science Center, Houston, Texas 77030

    Corresponding author.

     Address all correspondence and requests for reprints to: Stephen Safe, Department of Veterinary Physiology and Pharmacology, 4466 Texas A&M University, College Station, TX

    77843-4466., E-mail: [email protected].

    Received 2013 Sep 17; Accepted 2013 Nov 21.

    Copyright © 2014 by The Endocrine Society

    Abstract

    The nuclear orphan receptors for which endogenous ligands have not been identified include nuclear receptor (NR)0B1 (adrenal hypoplasia

    congenita critical region on chromosome X gene), NR0B2 (small heterodimer partner), NR1D1/2 (Rev-Erbα/β), NR2C1 (testicular receptor 

    2), NR2C2 (testicular receptor 4), NR2E1 (tailless), NR2E3 (photoreceptor-specific NR [PNR]), NR2F1 chicken ovalbumin upstream

     promoter transcription factor 1 (COUP-TFI), NR2F2 (COUP-TFII), NR2F6 (v-erbA-related protein), NR4A1 (Nur77), NR4A2 (Nurr1),

     NR4A3 (Nor1), and NR6A1 (GCNF). These receptors play essential roles in development, cellular homeostasis, and disease including cancer 

    where over- or underexpression of some receptors has prognostic significance for patient survival. Results of receptor knockdown or 

    overexpression in vivo and in cancer cell lines demonstrate that orphan receptors exhibit tumor-specific pro-oncogenic or tumor suppressor-like

    activity. For example, COUP-TFII expression is both a positive (ovarian) and negative (prostate and breast) prognostic factor for cancer 

     patients; in contrast, the prognostic activity of adrenal hypoplasia congenita critical region on chromosome X gene for the same tumors is the

    inverse of COUP-TFII. Functional studies show that Nur77 is tumor suppressor like in acute leukemia, whereas silencing Nur77 in pancreatic,

    colon, lung, lymphoma, melanoma, cervical, ovarian, gastric, and some breast cancer cell lines induces one or more of several responses

    including growth inhibition and decreased survival, migration, and invasion. Although endogenous ligands for the orphan receptors have not been

    http://www.ncbi.nlm.nih.gov/pmc/about/copyright.htmlmailto:dev@nullhttp://www.ncbi.nlm.nih.gov/pmc/about/copyright.htmlmailto:dev@nullhttp://www.ncbi.nlm.nih.gov/pubmed/?term=Lee%20SO%5Bauth%5Dhttp://www.ncbi.nlm.nih.gov/pubmed/?term=Reeder%20A%5Bauth%5Dhttp://www.ncbi.nlm.nih.gov/pubmed/?term=Hedrick%20E%5Bauth%5Dhttp://www.ncbi.nlm.nih.gov/pubmed/?term=Jin%20UH%5Bauth%5Dhttp://www.ncbi.nlm.nih.gov/pubmed/?term=Safe%20S%5Bauth%5Dhttp://dx.doi.org/10.1210%2Fme.2013-1291

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    2/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 2/36

    identified, there is increasing evidence that different structural classes of compounds activate, inactivate, and directly bind several orphan

    receptors. Thus, the screening and development of selective orphan receptor modulators will have important clinical applications as novel

    mechanism-based agents for treating cancer patients overexpressing one or more orphan receptors and also for combined drug therapies.

    The nuclear receptor (NR) superfamily of ligand-activated receptors exhibit a common modular structure (Figure 1A) and play essential roles,

    not only in maintaining cellular homeostasis but also in various disease processes including cancer (1 – 5). The intricate interplay between the

    activation or deactivation of NRs by different structural classes of endogenous ligands, such as the steroid and thyroid hormones, lipids, vitamins

    and other biochemicals, is essential for their function. The 48 NR family members have been classified into subgroups based on the identification

    of endogenous ligands for each receptor (1

    ). The endocrine receptors include the steroid hormone receptors that bind steroid hormones and the

    heterodimeric receptors that partner with the retinoid X receptor and bind thyroid hormones, retinoids, and vitamin D. The identification of 

    specific endogenous ligands for the endocrine receptors has facilitated the design and development of selective receptor modulators (SRMs) that

    exhibit tissue-specific agonist or antagonist activities (6) and are used extensively for treatment of hormone-/hormone receptor-dependent

    diseases (7, 8). Tamoxifen is one of many selective estrogen receptor (ER) modulators used in endocrine therapies for treating ER-positive

     breast cancer patients (9, 10).

    The adopted orphan receptors are a second major subtype of NRs that are subdivided into 2 groups based, in part, on their ligands. The lipid

    sensor receptor subtypes and their ligands include retinoid X receptor (9- cis-retinoic acid), peroxisome proliferator-activated receptors

    (PPARs) (fatty acids), liver X receptor (oxysterols), farnesoid X receptor (bile acids), and pregnane X receptor, which binds cholesterol

    derivatives. The enigmatic orphan receptor subtype includes constitutive androstane receptor (androstane and many drugs/xenobiotics),

    hepatocyte nuclear factor-4, and steroidogenic factor-1/liver receptor homolog 1(LRH-1) (phospholipids), retinoid acid-related orphan

    receptor (cholesterol and retinoic acids), and estrogen-related receptor (estrogens). Although adopted orphan receptors bind endogenous

    ligands, it is not clear whether their functions in normal or diseased cells are regulated by these ligands. SRMs have been identified for many

    adopted orphan receptors (11 – 14), and compounds such as the insulin-sensitizing thiazolidinediones (PPARγ agonists) have had extensive

    clinical applications.

    The orphan receptors are the third class of NRs, and endogenous ligands for these receptors have not been identified. The crystal structure of 

    the ligand-binding domain of the orphan receptor Nurr1 (NR4A2) shows that several hydrophobic residues protrude into the ligand-binding

     pocket, and a typical coactivator-binding site is lacking (15), suggesting that some orphan receptors may not bind ligands (16, 17). Like other 

     NR classes, the orphan receptors play important roles in cellular homeostasis and diseases including cancer (18 – 24), and several recent reports

    document the expression and potential functions of orphan receptors in different tumors and cancer cell lines (21 – 24). Breast tumors are

    routinely classified as ER or ER , and expression of ER has prognostic significance that dictates selection of therapeutic regimens. However,+ −

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    3/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 3/36

    analysis of ER and ER tumors for expression (mRNA) of all 48 NRs also demonstrated the important prognostic significance of several

    orphan receptors (21) (Figure 1B). The NR4A (Nur77/TR3, Nurr1, and Nor1) and NR2F6 [v-erbA-related protein (EAR2)] receptors are

    uniquely overexpressed in all (ER and ER combined) tumors. Moreover, Nur77, EAR2, and chicken ovalbumin upstream promoter 

    transcription factor II (COUP-TFII) are among a limited group of NRs that are prognostic for breast cancer classification and histologic grade,

    and COUP-TFII expression was a positive prognostic factor for tamoxifen-treated ER breast cancer patients (21

    ). Examination of lung tumor 

    and nontumor tissue indicated highly variable NR expression; however, gene combinations and individual receptors, such as the orphan receptor 

    small heterodimer partner (SHP, NR0B2), predicted enhanced survival for early-stage lung cancer patients (22). Moreover, expression of  Nur77 in normal lung epithelium from patients was an indicator for good prognosis (23). NR profiling of the NCI60 cancer cell panel

    demonstrated that relative expression levels of some orphan receptors also correlated with drug sensitivity (24). For example, cancer cell

    sensitivity to microtubule-disrupting drugs was enhanced in cells expressing low levels of EAR2 and COUP-TFII, whereas high levels of the

    orphan receptor tailless (TLX, NR2E1) correlated with sensitivity to 9-fluoroprednisolone (24).

    It is clear that NRs are expressed in multiple cancers and contribute to the cancer cell phenotype, and ligands (agonists or antagonists) for these

    receptors are important chemotherapeutic agents (7–10, 12–14, 19, 20). In this review, the prognostic and functional roles of orphan receptors

    in cancer cells and tumors will be outlined, and the potential therapeutic applications of SRMs that target orphan receptors will be highlighted.

    Expression And Prognostic Significance Of Orphan Receptors in Cancer 

    NR0B receptors

    The NR0B orphan subfamily contains 2 unique receptors, namely dosage-sensitive sex reversal and adrenal hypoplasia congenita critical region

    on chromosome X gene (DAX-1/NR0B1) and SHP/NR0B2. Both receptors express the ligand-binding domain (LBD) typical of all NRs, but

    they do not contain a DNA-binding domain and therefore, their functions are dependent on receptor-protein, but not on receptor-DNA,

    interactions (25 – 27). DAX-1 is expressed primarily in the hypothalamic-pituitary-adrenal/gonadal axis (28), and DAX-1 mutations result in

    adrenal hypoplasia, hormone deficiencies, and gonadal dysfunction (hypogonadotropic hypogonadism) (29 – 31). Despite the limited expression

    of DAX-1 in normal tissues, this receptor is more highly expressed in mammary (32, 33), endometrial (34), ovarian (35), adrenocortical, and

     pituitary (36, 37), lung (38), and prostate (39) tumors, and Ewing's sarcoma (40 – 44). The prognostic significance of high or low expression of 

    DAX-1 has been reported only for a limited number of cancers. For example, in a cohort of node-negative breast cancer cases, low and high

    DAX-1 mRNA expression correlated with poor and enhanced survival, respectively (33) (Figure 1B). DAX-1 was also highly expressed in

    endometrial tumors compared with nontumor tissues and, although DAX-1 expression was inversely correlated with tumor grade, a correlation

     between DAX-1 levels and disease-free survival was not observed (34

    ). Nuclear DAX-1 was also highly expressed in 21/40 prostate tumors,

    and immunoreactivity for receptor expression was inversely correlated with Gleason score. In contrast, DAX-1 was expressed at a high

    + −

    + −

    +

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    4/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 4/36

    frequency in epithelial ovarian tumors, and levels correlated with several tumor parameters (eg, size, grade, Ki-67 labeling) including decreased

    disease-free survival (35) and, in lung cancer, DAX-1 expression was a negative prognostic factor associated with high rates of lymph node

    metastasis and decreased disease-free survival (38). SHP (NR0B2) also lacks the conserved DNA binding domain common to other NRs, and

    SHP is widely expressed in multiple tissues (45 – 47). Studies on the expression and prognostic significance of SHP in tumors are limited to

    reports of higher receptor levels in hepatocellular carcinomas compared with normal liver (48 – 50) and higher levels in intestinal metaplasia (a

     precancerous lesion) compared with gastric mucosa (51). Thus, the prognostic significance of high or low expression of DAX-1 and SHP is

    variable and tumor specific (Figure 1B), and the underlying mechanisms that trigger altered expression of these genes in specific tumors has not been reported.

    NRID receptors

    Rev-erbα (NRID1) and Rev-erbβ (NRID2) are widely expressed in tissues (52) and play an integral role in regulation of circadian rhythms and

    important mammalian molecular clock genes such as Bmal1 and Clock (53 – 55). Examination of more than 100 breast tumors showed Rev-erbα

    was the only NR receptor among the 66 amplified genes that correlated with poor clinical outcomes (56). Rev-erbα, in combination with

    SMARCE1/BIRC5 copy numbers, was a significant predictor of breast tumor recurrence (within 5 years) of women with ER/PR-positive or -

    negative tumors (57); however, these results were in contrast to a recent study showing lower expression of Rev-erbα in breast tumors (ER 

    and ER ) than in normal breast tissue (21).

    NR2C receptors

    The orphan nuclear testicular receptor 2 (TR2; NR2C1) and TR4 (NR2C2) are structurally related and exhibit comparable expression patterns

    in multiple tissues (58), and expression of both receptors are lower in ER and ER breast cancer compared with normal breast tissue (21).

    However, their importance as prognostic factors for breast or other tumors has not been reported. A recent study reported that the testicular 

    orphan NR4-associated protein 16 (TRA16) was overexpressed in approximately 90% of non-small cell-lung tumors compared with

    nondetectable levels in normal lung tissue and was a negative prognostic factor for patient survival (59). It was hypothesized that TRA16

    interacts with TR2, resulting in derepression of ERβ, which plays an inhibitory role in lung tumorigenesis.

    NR2E receptors

    The nuclear orphan receptor TLX (NR4E1) has been extensively characterized in mouse models as an important neurogenesis gene required for 

    maintaining neural stem cells (59 – 64), and the expression and prognostic significance of TLX have been extensively investigated in brain cancers

    (65 – 70). TLX is overexpressed in neurocytomas compared with expression in the ventricular zone and neuroendocrine-specific protein ( 65)

    and in proliferative and mesenchymal-like subtypes of astrocytomas compared with the proneural subtype, and this is also observed for other 

    +

    + −

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    5/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 5/36

    neural stem cell markers. TLX is overexpressed in gliomas (67, 68); high levels of TLX mRNA in tumors correlated with decreased patient

    survival (68), and TLX expression was higher in more aggressive (grades III and IV) compared with less aggressive (grade II) tumors (69) (

    Figure 1B).

    The photoreceptor-specific NR (PNR) (NR2E3) was identified in a search for TLX-related genes, and PNR expression is primarily observed in

    the retina and retinal cells (71, 72). A woman with paraneoplastic retinopathy and poorly differentiated lung carcinoma expressed antibodies

    against PNR (73), and serum PNR autoantibodies were higher in pancreatic cancer patients than in noncancer patients (74). There was also a

    correlation between expression of ERα (ESR1) and PNR in the NCI-60 cancer cell lines and in breast tumors from 2 different patient cohorts

    (75), and higher or lower expression of PNR correlated with good or poor clinical outcomes, respectively. Expression of PNR in breast tumors

    was also associated with enhanced recurrence-free survival of breast cancer patients treated with tamoxifen (75) (Figure 1B). PNR protein was

    also detected in ER-positive MCF-7 and T47D breast cancer cell lines, and knockdown of PNR significantly decreased ERα and estrogen-

    induced cell proliferation and estrogen-responsive genes. Although the expression and prognostic significance of TLX and PNR in multiple

    tumor types has not been extensively investigated, the prognostic significance of this subfamily of orphan receptors for decreased or enhanced

    clinical outcomes should be further investigated in other tumor types.

    NR2F receptors

    The 2 chicken ovalbumin upstream promoter transcription factors (COUP-TFs) are COUP-TF1 (NR2F1) and COUP-TFII (NR4F2), which

    are 97% and 99% homologous in their ligand- and DNA-binding domains, respectively, are widely expressed in multiple tissues and tumors

    (76 – 78). Prostate tumor samples exhibited higher expression of COUP-TFII protein in tumor vs nontumor samples; approximately 60% of all

    tumors exhibited intermediate to intense staining, whereas only 5% of the nontumor tissue stained positive for the receptor (79). Higher levels of 

    COUP-TFII expression predicted earlier recurrence of the disease. In another study, COUP-TFI was expressed in 21/28 prostate tumors;

    however, correlations between expression levels and disease parameters were not observed (80). Among 119 breast tumors, 59% stained

     positive for COUP-TFII, and higher expression of the receptor was a negative prognostic factor for several parameters including overall patient

    survival (81). Another report showed that COUP-TFII was expressed in both breast tumor and nontumor tissue; COUP-TFII expression was

    correlated with ERα expression, and high grade tumors tended to be COUP-TFII negative (82). Thus, at least for breast cancer, the prognostic

    significance of COUP-TFII varies between studies, and this may be due to different subsets of patients. COUP-TFII is differentially expressed

    in normal ovarian stroma (high) and epithelial (low) cells and in ovarian tumors, this expression pattern is reversed. Patients with tumors

    expressing high epithelial/stroma ratios of COUP-TFII exhibited decreased tumor recurrence times (83). Another study reported relatively low

    expression of COUP-TFII in ovarian tumors compared with nontumor tissue; however, COUP-TFII tissue compartmental ratios were not

    reported (84). COUP-TFII was observed in more than 57% of tumors from colon cancer patients, and there was minimal expression in normal

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    6/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 6/36

    colonic mucosa; expression of the receptor correlated with increased rates of disease-free survival (85).

    EAR2 (NR2F6) is the third and more distant member of the NR2F subfamily and is expressed in the human fetal liver and in mouse embryos

    (86, 87). EAR2 is more highly expressed in both ER and ER breast tumors compared with normal breast tissue (88), and results of NR 

     profiling of the NCI-60 cancer cell panel show that cells with lower levels of EAR2 exhibit enhanced sensitivity to anticancer drugs targeting

    microtubules (89). EAR2 is also more highly expressed in lymphomas (90) and colorectal tumors than in nontumor tissue. Among the nuclear 

    orphan receptors, NR2F subfamily members have been more extensively investigated for their expression and prognostic significance in multiple

    tumors. Patients with tumors expressing high levels of COUP-TFII exhibited decreased (breast and prostate) and increased (colon and ovarian)

    survival/decreased disease recurrence (Figure 1B) and, for breast and prostate cancer, the prognostic significance correlated with the pro-

    oncogenic functions of COUP-TFII in mouse models (79, 91).

    NR4A and NR6A receptors

     NR4A receptors (NR4A1 [NGFI-B, Nur77, TR3]; NR4A2 [Nurr1]; NR4A [Nor1]) were initially identified as immediate-early genes induced

     by nerve growth factors in PC12 cells, and these receptors are important in normal and tumor tissues (reviewed in References 92 and 93).

    Expression of Nur77 in tumors and cancer cell lines has been extensively investigated, and higher levels are expressed in colon, pancreatic, lung,

    and breast tumors compared with nontumor tissue (21, 94 – 96). Nur77 is highly expressed in both ER and ER breast tumors (21), and thereceptor tends to be more highly expressed in highly differentiated, lower grade tumors (97). Examination of publicly available array data for 

    lung tumors showed that high expression of Nur77 mRNA predicted increased patient survival (20), whereas in another cohort study, high levels

    of Nur77 protein were prognostic for decreased patient survival (94) (Figure 1B). Nur77 expression was detected in whole bone marrow,

    CD33 myeloid cells, and CD34 progenitors from healthy patients, whereas levels of Nur77 were decreased in acute myeloid leukemia

    (AML) patients (98). Nur77 mRNA was also part of a gene signature in tumors associated with metastasis (99). Nurr1 is more highly

    expressed in ER breast tumors compared with normal mammary tissue (21), and Nurr1 (cytosolic and nuclear) protein is more highly

    expressed in bladder tumors compared with surrounding tissue (100). Higher levels of cytoplasmic Nurr1 were a prognostic factor for high

    tumor grade, decreased survival, and increased distant metastasis in a cohort of bladder cancer patients (100). In breast cancer patients, Nurr1

    expression was higher in normal breast epithelium than breast tumors, and expression of this receptor was inversely correlated with lymph node

    metastasis but correlated with increased relapse-free survival (101) (Figure 1B). Nurr1 was more highly expressed in prostate tumors vs the

    normal prostate and was correlated with tumor classification and Gleason score as a negative prognostic factor (102). Recent studies also show

    that Nurr1 expression predicts poor survival and drug resistance in colon and gastric cancer patients (103, 104).

     Nor1 is more highly expressed in ER and ER breast tumors compared with normal breast tissue; however, Nor1 levels did not correlate with

    tumor characteristics (21). Expression of GCNF (NR6A1) is lower in both ER and ER breast tumors compared with normal mammary tissue

    + −

    − +

    + +

    +

    + −

    + −

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    7/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 7/36

    (24), and cancer cells expressing low levels of GCNF exhibit higher sensitivity to the anticancer drug ecteinascidin (24).

    Summary

    Reports on prognostic significance and levels of nuclear orphan receptors in tumors are incomplete; however, Figure 1 illustrates that expression

    of several orphan receptors in tumors predicts increased or decreased patient survival that is dependent on tumor type and, for some receptors,

    their prognostic significance for a specific tumor varied between studies. Tumor suppressor genes and oncogenes are characteristically up- or 

    down-regulated (or mutated) in tumors; however, altered expression of other genes such as orphan receptors is not well understood.Examination of the ENCODE database (db = hg19 and c = chr12 and g = wgEncodeUwhistone) for DAX-1 in A549 lung cancer cells vs

    normal human lung fibroblasts showed high expression of the H3K4me3 activation mark over the chromosome region of the DAX-1 gene only

    in the cancer cell line. It is possible that overexpression of some orphan NRs in tumors (eg, lung) may be due to epigenetic effects and this is

    currently being investigated.

    Functions and Pathways Regulated by Orphan Receptors in Cancer 

    Multiple genes and their corresponding proteins serve as prognostic factors for selected tumors, and oncogenes such as Ras and Myc and

    epidermal growth factor receptors that are activated or overexpressed in tumors also exhibit oncogenic functions. This section of the review will

    outline the functions and pathways regulated by orphan NRs and determine correlations (or lack thereof) with their corresponding prognostic

    significance. The functions of orphan receptors have primarily been investigated in knockout mouse models, by RNA interference (RNAi),

    antisense constructs, or overexpression in cells; however, correlations between prognostic and functional (oncogenic or tumor suppressor-like)

    activities of orphan receptors are limited.

    NR0B receptors

    DAX-1 expression is a negative (lung and ovarian) and positive (prostate and breast) prognostic factor for patient survival, and DAX-1 silencing

    in lung cancer cells decreases growth, survival, and invasion, demonstrating pro-oncogenic functions for this receptor. DAX-1 and the

    oncogenic fusion protein EWS (Ewing sarcoma)/FLI1 coordinately regulate multiple genes in Ewing' sarcoma, and EWS/FLI1 regulates DAX-1

    expression in these tumors (40 – 44). In mouse embryonic stem cells, DAX-1 and LRH-1 (NR4A2) cooperatively mediate the activation of the

    stem cell marker Oct4, and DAX-1/LRH-1 may play a role in stem cell and cancer stem cell function ( 105). DAX-1 interactions with EWS-

    FLI1 and other NRs is both dependent and independent of the LXXLL motif that facilitates interactions with some receptors ( 106 – 114), and

    this may competitively displace NR coactivators and other nuclear LXXLL-interacting nuclear factors. Although SHP expression varies in some

    tumors, the correlation with patient outcomes has not been determined; however, SHP mice develop hepatocellular tumors at 12–15 months

    of age, suggesting a possible tumor-suppressive function for this receptor in liver carcinogenesis, and this has been related to the inverse

    −/−

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    8/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 8/36

    expression of SHP and cyclin D1 (48). SHP expression is higher in intestinal metaplasia than in gastritis mucosa, and SHP plays a tumor-

     promoting role in this precancerous adenocarcinomal lesion by regulating expression of caudal-related homeobox gene (CDX1) that is involved

    in intestinal metaplasia (51).

    There is extensive literature on the mechanism of action of SHP in cancer and noncancer cell context, and a major function of this receptor 

    involves direct interactions with NRs, other transcription factors, and nuclear cofactors to inhibit gene expression. SHP interacts with 23 NRs,

    17 nuclear cofactors including coactivators and corepressors, and 12 transcription factors including oncogenes (c-jun/JunD), tumor suppressors

    (p53), and other factors such as NFκB that play a role in tumorigenesis (47, 115). Many SHP-interacting proteins have been identified instudies on the role of SHP in a noncancer cell context, and these interactions may also influence the tumorigenicity of transformed cell lines. SHP

    expression is lower in prostate cancer cells compared with human prostate RWPE-1 epithelial cells and inversely correlated with miR-141 levels

    that repress SHP (116). Overexpression of p53 in 293T cells enhanced proteasome-dependent degradation of SHP protein, and SHP

    overexpression enhanced the ubiquitin ligase activity of MDM2 which, in turn, decreased p53 stability (117). Nur77 also binds and inactivates

     p53, and Nur77 antagonists activate p53 and inhibit mammalian target of rapamycin (mTOR) signaling in lung cancer cells (94, 118), suggesting

    that SHP ligands could also potentially affect p53 function. SHP also interacts with and inhibits ERα- and ERβ-mediated transactivation;

    however, it is unclear whether these effects modify mammary carcinogenesis (119, 120). Thus, among the NR0B receptors, DAX-1 expression

    is a prognostic factor for decreased lung cancer survival and exhibits pro-oncogenic activity (Figure 2); more extensive studies are required todefine the role of DAX-1 and SHP in tumorigenesis.

    NRID and NR2C receptors

    The function of Rev-erbα/β receptors in tumors and cell lines has not been determined; however, these receptors activate or repress multiple

    genes including N-myc and deleted in breast cancer 1 (DBC1) through either direct DNA (promoter) binding or protein-protein interactions

    (52, 121 – 126). Increased expression of TR4 was observed in prostate cancer stem/progenitor cells expressing CD133 (PCaCD133 )

    compared with PCaCD133 cells, and knockdown of TR4 in PCaCD133 cells resulted in increased sensitivity to treatment with docetaxel

    and etoposide, suggesting a role for TR4 in chemoresistance (127). TR2 and TR4 suppress ER-mediated gene expression and proliferation of 

     breast cancer cells (128, 129) and also suppress androgen receptor function in prostate cancer cell lines (130), due to direct interactions

     between TR2/TR4 with other NRs or nuclear factors and with cis-promoter elements (59, 128 – 136).

    NR2E receptors

    TLX is highly expressed in brain tumors and is a prognostic factor for decreased patient survival (68), and in vitro and in vivo studies

    demonstrate oncogenic-like activity of this receptor. Knockout of TLX in mouse models confirmed the role of TLX in promoting brain tumor 

    +

    − +

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F2/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    9/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 9/36

    formation and neural stem cell-dependent tumors (69). Silencing TLX in IMR-32 and SH-SY5Y neuroblastoma cells decreased cell growth and

    anchorage-independent colony formation (70). Overexpression of TLX increased cell proliferation and foci formation in Ink4a/Arf 

    astrocytes, and tumorigenesis was also enhanced in mice bearing these astrocytes as xenografts (68). TLX overexpression resulted in

    development of gliomas and glioma-like lesions in mice (67) and, in U87MG glioma cells, TLX overexpression increased cell proliferation and

    formation of colonies in soft agar (68). The mechanisms of TLX-mediated pro-oncogenic activity are complex and may be linked to direct and

    indirect regulation of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-2α through binding to von Hippel-Lindau protein

    (70) and activation of cyclin D1 (68).

    Knockout of PNR in ER-positive breast cancer cell lines decreased ERα-mediated responses including gene expression (75), and mechanistic

    studies show that PNR in combination with protein inhibitor of signal transducer and activator of transcription (STAT)3 (protein inhibitor of 

    activated STAT 3), bind directly to the ERα gene promoter to regulate ERα gene expression (75). High-throughput genetic screens showed that

    PNR induced apoptosis and p53-responsive genes, and this was due to stabilization of p53 by interactions with PNR and p300-dependent

    acetylation of p53 (137). Regulation of ERα and p53 by PNR in cancer cells suggests the potential clinical importance of this gene as a drug

    target.

    NR2F receptors

    The functions of COUP-TFII in tumor development have been investigated in mouse models with tissue-specific loss of COUP-TFII. For 

    example, in tumor xenograft and mouse mammary tumor models, the loss of COUP-TFII resulted in decreased tumorigenesis due to decreased

    neoangiogenesis, and decreased tumor growth and metastasis were observed in the mammary tumor model (91). COUP-TFII knockdown (81)

    or overexpression (138) in breast cancer cells confirmed a role for this receptor in breast cancer cell growth, survival, motility, and invasion

    (138). These functional observations correlated with the prognosis of decreased survival of patients with tumors expressing high levels of 

    COUP-TFII (81). In contrast, the effects of COUP-TFII on drug resistance in breast cancer cells were dependent on the specific agent and cell

    line (138, 139). For example, overexpression of COUP-TFII in triple-negative MDA-MB-231 cells enhanced the growth-inhibitory effects of 

    tamoxifen (139), whereas cells expressing low levels of COUP-TFII exhibited increased sensitivity to microtubule disrupting drugs such as taxol

    (101). The loss of COUP-TFII in RIP-Tag2 mice resulted in inhibition of pancreatic islet tumorigenesis at multiple stages (140); however,

    studies in pancreatic cancer cell lines have not been reported. In transgenic models of prostate cancer, loss of COUP-TFII decreased prostate

    tumor progression and survival (79). The pro-oncogenic activity of COUP-TFII was further confirmed by overexpression of this receptor in

    wild-type or transgenic mice with decreased or loss of phosphatase and tensin analog expression. COUP-TFII overexpression alone did not

    initiate prostate tumorigenesis but clearly enhanced this process in mice with loss of phosphatase and tensin analog, thus confirming the tumor 

     promoter-like activity of COUP-TFII in prostate cancer (79). Low levels of COUP-TFII were observed in prostate cancer cell lines, and

    −/−

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    10/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 10/36

    overexpression of the receptor inhibited androgen-induced cell growth and gene expression (141); these results were in contrast to the in vivo

    studies. Overexpression of COUP-TFII in noninvasive A549 lung cancer cells increased invasiveness, and knockdown of the receptor in A460

    cells that express relatively high levels of the receptor decreased invasiveness (142). These functional studies were consistent with pro-

    oncogenic activity for COUP-TFII for breast, pancreatic, prostate, and lung cancer, whereas in ovarian cancer cells (ES-2 and TOV112D),

    knockdown of COUP-TFII increased proliferation of ES2 cells but increased apoptosis in both cell lines (83).

    COUP-TFII modulates gene expression through direct promoter DNA binding or through protein-protein interactions, and many of the genes

    regulated by COUP-TFII, including angiopoietin-1 (91) and VEGF/VEGFR2 (81, 140), are associated with angiogenic pathways. COUP-TFIIalso interacts with nucleolin in breast cancer, and nucleolin coactivates COUP-TF-mediated regulation of retinoic acid receptor β in breast

    cancer cells (82). COUP-TFII also modulates ligand-activated ERα signaling in breast cancer (139) and decreases androgen-dependent

    activation of prostate-specific antigen in prostate cancer cells through direct COUP-TFII-AR interactions (141). COUP-TFII overexpression in

    lung cancer cells enhanced invasiveness, and this was accompanied by activation of focal adhesion kinase, matrix metalloproteinase 2 and

    urokinase receptor (142). COUP-TFII also regulates expression of several genes and pathways in ovarian cancer cells but not angiogenic genes

    (83), indicating the importance of cancer cell context on the role of this orphan receptor. Knockdown of EAR2 by RNAi induces apoptosis in

    colon cancer, and this is correlated with decreased expression of the antiapoptotic factor X-linked inhibitor of apoptosis (143). Loss of EAR2 in

    RKO cells also decreases tumor growth in athymic mice bearing these cells as xenografts compared with growth of cells expressing wild-typeEAR2.

    NR4A and NR6A receptors

    Although Nur77 appears to be a tumor suppressor in the Nur77 /Nor1 mouse that spontaneously develops leukemia, knockdown or 

    overexpression studies in most cancer cell lines indicate that Nur77 is pro-oncogenic. Silencing of Nur77 in pancreatic, colon, lung, lymphoma,

    melanoma, cervical, ovarian, and gastric cancer cell lines inhibited cell growth, survival, migration, and/or invasion (94, 99, 144 – 149).

    Combined knockdown of histone deacetylase inhibitor-induced Nur77 and Nor1 increased growth and decreased apoptosis in AML cell lines

    (150), whereas in other leukemia cell lines (eg, Jurkat), we have observed pro-oncogenic activity for Nur77 (our unpublished observations), and

    studies on the tumor type-specific role of Nur77 in leukemic cells are ongoing. The oncogenic and tumor suppressor-like functions of Nur77 in

    lung and AML cell lines and knockout mouse models, respectively, correlated with the decreased and increased survival of lung and AML

    cancer patients expressing high levels of this receptor.

    The caged retinoid compound CD437, and several analogs inhibit growth and induce apoptosis in lung and other cancer cell lines (151 – 161),

    and these responses have been linked to Nur77 (162). Retinoid-induced proapoptotic responses are Nur77 dependent and are blocked by the

    nuclear export inhibitor leptomycin B, indicating that the effects of CD437 and other apoptosis inducers are due to the extranuclear effects of 

    −/− −/−

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    11/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 11/36

     Nur77 (163). The mechanisms of extranuclear Nur77-mediated apoptosis in some cell lines involves a novel pathway in which Nur77 is

    associated with mitochondria and forms a novel Nur77-bcl2 proapoptotic complex that induces release of cytochrome c and activation of the

    intrinsic apoptosis pathway (163, 164). A peptide mimic of Nur77 that binds bcl2 and paclitaxel, which acts as a Nur77 mimic, also induce

    apoptosis in cancer cells (165, 166). Platinum-resistant ovarian cancer cells have been linked to low Nur77 expression because platinum-

    induced apoptosis is also due to nuclear export of Nur77 (167). The increased survival of liver cancer cells is due to expression of 

    chromodomain helicase/adenosine triphosphatase (ATPase) DNA-binding protein 1-like (CHD1L), which inhibits nuclear export of Nur77

    (168). CD437 also induces nuclear translocation of SHP to form a complex with bcl-2; however, SHP, but not Nur77, binds CD437 and

    structurally related analogs (169).

    RNAi studies in cancer cell lines showed that Nurr1 was associated with bladder cancer cell invasion (170), melanoma cell migration (145),

     prostate cancer cell growth, migration and invasion (102), and breast cancer growth (101). Nurr1 is also associated with drug resistance in

    colon and gastric cancer cells (104). In contrast, it was also reported that gastrin induced Nurr1 expression in gastric cancer cells, and

    knockdown or overexpression of the receptor showed that Nurr1 suppressed gastrin-induced migration and invasion of gastric cancer cells

    (171). The differences between the 2 studies in gastric cancer cells may be due to gastrin-induced changes in intracellular cytosol-nuclear 

    shuttling of Nurr1, which favors cytosolic accumulation of the receptor (171). Thus, like Nur77, Nurr1 primarily exhibits oncogenic activity in

    cancer cells and the decreased survival of bladder, gastric, and prostate cancer patients that overexpress Nurr1 correlates with the oncogenicactivity of this receptor in their corresponding cancer cell lines.

    Combined knockdown of Nur77 and Nor1 in mice results in the rapid formation of AML in mice (98); however, the expression and role of 

     Nurr1 and GCNF in tumors is poorly defined and requires additional research. Although functional and prognostic studies on orphan receptors

    in different tumor types are limited, with the exception of leukemia, overexpression of these receptors in solid tumors predicted decreased

    survival, and this correlated with their oncogenic activities observed in corresponding cancer cell lines (Figure 2).

    Orphan NR Ligands

    The expression and pro-oncogenic or tumor suppressor-like functions of orphan receptors in tumors indicate that orphan receptor ligands actingas agonists or antagonists represent a potentially novel class of selective receptor modulators for cancer chemotherapy. Identification of ligands

    for TLX, EAR2, and GCNF has not been reported; however, ligand binding and/or ligand-dependent activation or inactivation of the remaining

    orphan NRs has been observed (Figure 3) and provides a basis for development of selected receptor modulators of orphan receptors.

    NR0B receptors

    The DAX-1 promoter has been used in a screening assay of 50 000 compounds in EWS/FLI1 in Ewing's sarcoma cells (172), and mithramycin

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F2/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    12/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 12/36

    was identified as the most effective agent. Mithramycin exhibits anticancer activity; however, inhibition of EWS/FLI1-mediated transactivation

     by mithramycin could also be due to other activities of this drug, which inhibits transactivation of GC-rich gene promoter sites (173 – 176).

    The adamantyl retinoid CD437 and related derivatives induce apoptosis in several cancer cell lines (151 – 157), and this compound specifically

     binds SHP in a competitive displacement assay (Figure 2) (158 – 161). CD437 and other adamantyl retinoids and hepatocyte nuclear factor 4α

    induce SHP in liver and liver cancer cells, and the receptor is equilibrated in nuclear, cytosolic, and mitochondrial fractions of the cell (169).

    SHP also colocalizes with bcl-2 in the outer mitochondrial membrane, resulting in release of cytochrome c and induction of apoptosis. The

    retinoid-induced nuclear translocation of SHP to mitochondria has also been reported for Nur77 (NR4A1), which also interacts with bcl-2(162) but does not bind CD437. The parallel functions of the adamantyl retinoids in inducing nuclear export of both SHP and Nur77 may also

     be complicated by the reported SHP-mediated repression of nuclear Nur77 (177). Although it is clear that the adamantyl retinoids effectively

    induce apoptosis in several different cancer cell lines, the interplay between SHP and Nur77 and their separate and combined roles in mediating

    apoptosis in cancer cells has not been resolved.

    NRID, NR2C, NR2E, and NR2F receptors

    Heme associates with the LBD of Rev-erbα/β receptors (Figure 3) and enhances recruitment of the corepressor, nuclear receptor corepressor,

    to repress target genes such as Bmal1 (178, 179). Synthetic Rev-erbα/β agonists and antagonists have been characterized and shown to befunctional (180 – 182), suggesting that these receptors may be druggable targets for specific tumors. The crystal structure of the LBD of TR4

    shows that, like COUP-TFII, the ligand-binding cavity and the coactivator-binding sites on the AF-2 helix are blocked (183). Nevertheless,

    TR4-dependent transactivation is enhanced by some coactivators, and both retinol and all-trans-retinoic acid bind the receptor (LBD) (183) (

    Figure 3). PNR agonists have also been identified in transactivation assays (137, 184); however, the direct binding or effects of these

    compounds have not been reported.

    The crystal structure of the COUP-TFII LBD shows that the receptor exists in an autorepressed condition with helix α 10 extending into the

    ligand-binding pocket, and the AF2 helix is folded into a position that blocks coactivator binding (88). Despite these constraints, 9-cis- and all-

    trans retinoic acid induce COUP-TFII-dependent transactivation and enhance coactivator binding (Figure 3). These results suggest thatCOUP-TFII may also be targeted by tissue-selective agonists or antagonists that can be developed as anticancer agents to either activate or 

    inhibit the function of this receptor.

    NR4A receptors

    At least 2 structurally different classes of ligands that activate nuclear Nur77 have been reported, and these include cytosporone B (CsnB) and

    related analogs (185, 186) and a series of 1,1-bis(3′-indolyl)-1-( p-substituted phenyl)methanes (C-DIMs) (95, 96, 117, 187 – 189) (Figure 3).

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F2/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    13/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 13/36

    CsnB and related compounds directly bind Nur77, activate the receptor in transactivation assays, and induce recruitment of coactivators;

    however, the proapoptotic activity of CsnB is due to both nuclear and extranuclear pathways. CsnB induces nuclear export of Nur77 to the

    mitochondria, and CsnB also decreases expression of the antiapoptotic gene brain and reproductive organ-expressed (BRE) via nuclear Nur77-

    dependent repression (185, 186). The C-DIM compounds are synthetic analogs derived from 1,1-bis(3′-indolyl)methane (DIM) which is a

    major metabolite of indole-3-carbinol, a cancer chemopreventive and chemotherapeutic phytochemical that is one of the active components of 

    cruciferous vegetables. C-DIMs contain a substituted phenyl (or other aromatic) group and may also have indole ring substituents (Figure 3). A

    subset of C-DIMs activate PPARγ in cancer cell lines (190, 191), and the p-methoxyphenyl analog (DIM-C-pPhOCH ) activates nuclear 

     Nur77-dependent transactivation in multiple cancer cell lines and, like CsnB, DIM-C-pPhOCH induces apoptosis and inhibits cancer cell and

    tumor growth (94, 96). In addition, the p-hydroxyphenyl (DIM-C-pPhOH), p-carboxymethylphenyl (DIM-C-pPhCOOMe), and other analogs

    have been identified as inactivators of Nur77 that also induce apoptosis and inhibit cancer cell and tumor growth ( 192). Studies on the direct

     binding of C-DIMs to Nur77 are currently underway; however, using a combination of Nur77 silencing (siNur77) and treatment with DIM-C-

     pPhOH, several Nur77-dependent genes/pathways associated with this receptor have been identified (94, 96, 192, 193). Nur77 activates

    survivin and bcl2 through interaction of Nur77 with p300 and the DNA-bound Sp1 transcription factor, and siNur77 or DIM-C-pPhOH

    decreases survivin and bcl2 expression in cancer cells (96). A second pathway is only observed in p53-positive cancer cell lines in which Nur77

     binds and inactivates p53, and this has been linked to activation of mTOR signaling (94). Treatment of cancer cells with siNur77 or DIM-C-

     pPhOH results in mTOR inhibition, and this is due to activation of p53 and p53-dependent induction of sestrin 2 which, in turn, activates themTOR inhibitor phospho-cAMP activated protein kinase-α (94). A third recently discovered pathway involves the regulation of the thioredoxin

    genes that maintain low oxidative stress levels in pancreatic cancer cells, and transfection with siNur77 or treatment with DIM-C-pPhOH

    induced oxidative and endoplasmic reticulum stress and apoptosis (193). These results not only demonstrate the effectiveness of agents that act

    as Nur77 antagonists but also reveal the pro-oncogenic pathways regulated by Nur77 and the importance of the receptor as a drug target.

    X-ray crystallographic studies of the LBD of Nurr1 showed that hydrophobic substituents fill the ligand-binding pocket, suggesting this may be a

    true orphan receptor devoid of endogenous or exogenous ligands (15). A library screen for Nurr1 activators identified only 6-mercaptopurine

    and a few structural analogs (Figure 3) that activated Nurr1 through the N-terminal and not the C-terminal LBD (194). However, substituted

     benzimidazole and pyridinone derivatives and C-DIMs have subsequently been identified as potential Nurr1 agonists (195, 196), and the p-

    chlorophenyl C-DIM analog (DIM-C-pPhCl) activates Nurr1 and is a potent inhibitor of bladder cancer cell and tumor growth (170). Nurr1 is

    a potential target for cancer chemotherapy; however, more extensive characterization of Nurr1 agonists and antagonists and their receptor 

     binding is essential. Nor1 expression and function in tumors has not been reported; however, like Nurr1, 6-mercaptopurine induced Nor1-

    dependent transactivation and prostaglandin A2-activated Nor1 via the LBD (197, 198) (Figure 3).

    Summary and Conclusions

    33

    7/3/2016 Mi i i R l Of O h N l R t i C d P t ti l D T t

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    14/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 14/36

    Figure 1 illustrates the tumor site-specific expression and prognostic significance of orphan receptors. The prognostic significance of one or more

    orphan receptors has been reported in multiple tumors, and orphan receptors such as TLX may be highly specific for brain tumors in which this

    receptor is a negative prognostic factor and functions as an oncogene (67 – 69). Other orphan receptors such as DAX-1, COUP-TFII, and

     Nur77 play a significant prognostic and/or functional role in multiple tumors (Figure 1B); however, for many tumors only one of the prognostic

    or functional parameters is known. Reports for some receptors are contradictory, and the prognostic and functional parameters for many orphan

    receptors vary with tumor type. For example, in most cancer cells and tumors, Nur77 exhibits pro-oncogenic activity, whereas Nur77

    /Nor1 double-knockout mice rapidly develop AML-like leukemia and Nur77 may act as a tumor suppressor (98). The prognostic activity

    of some receptors such as COUP-TFII and DAX-1 are also highly variable. High levels of DAX-1 expression are both favorable (breast,

     prostate) and unfavorable (lung, ovarian), whereas COUP-TFII expression is a favorable (ovarian, colon) and an unfavorable (breast, prostate)

     prognostic factor for a different set of cancer patients. Orphan NR expression in multiple cancers needs to be further investigated for its

     prognostic significance and function and as a potential target for SRMs that can be used alone or in combination therapies.

     Nur77 is among the most thoroughly investigated of the orphan NRs, and the prognostic and functional activity of this receptor is cancer cell and

    tumor context dependent and is also influenced by ligand-induced nuclear and extranuclear (nongenomic) pathways as illustrated in Figure 4.

    The extranuclear function of Nur77 is somewhat unique, and the nuclear-cytosolic ratios influence cell death and survival pathways as well as

    drug efficacy. The CHD1L gene product inhibits nuclear export of Nur77 and Nur77-mediated apoptosis in liver cancer cells ( 168), and a

    recent report suggests that cisplatin-induced apoptosis in ovarian cancer cells is dependent on nuclear export of this orphan receptor (167). The

    antineoplastic activity of Nur77 is associated with ligand-activated genomic and nongenomic pathways, and it is possible that these dual

     pathways may also be important for other orphan NRs. The expression of orphan receptors in tumors coupled with the ongoing identification of 

    receptor agonists and antagonists (Figure 3) indicate that selective orphan receptor modulators represent a novel and important class of 

    mechanism-based drugs that will have significant future clinical applications for cancer chemotherapy.

    Acknowledgments

    This work was supported by National Institutes of Health Grants (R01CA142697 and CA-124998), Texas AgriLife Research, and the Syd

    Kyle Chair.

    Disclosure Summary: The authors have nothing to disclose.

    Footnotes

     Abbreviations:

     AML acute myeloid leukemia

    −/

    − −/−

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F3/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F4/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/figure/F1/

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    15/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 15/36

    COUP-TF chicken ovalbumin upstream promoter transcription factor

    CsnB cytosporone B

    DAX-1 adrenal hypoplasia congenita critical region on chromosome X gene

    DIM 1,1-bis(3′-indolyl)methane

    EAR2 v-erbA-related protein

    ER estrogen receptor

    EWS Ewing sarcoma

    LBD ligand-binding domain

    LRH-1 liver receptor homolog 1

    mTOR mammalian target of rapamycin

    NR nuclear receptor

    PNR photoreceptor-specific NR

    PPAR peroxisome proliferator-activated receptor

    RNAi RNA interference

    SHP small heterodimeric partner

    SRM selective receptor modulator

    TR testicular receptor

    VEGF vascular endothelial growth factor.

    References1. Sonoda J, Pei L, Evans RM. Nuclear receptors: decoding metabolic disease. FEBS Lett. 2008;582:2–9 [PMCID: PMC2254310]

    [PubMed: 18023286]

    2. Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a

    hierarchical transcriptional network. Cell. 2006;126:789–799 [PubMed: 16923397]

    3. Sherman MH, Downes M, Evans RM. Nuclear receptors as modulators of the tumor microenvironment. Cancer Prev Res (Phila). 2012;5:3– 

    10 [PMCID: PMC3408218] [PubMed: 22135047]

    4. Evans RM. The nuclear receptor superfamily: a rosetta stone for physiology. Mol Endocrinol. 2005;19:1429–1438 [PubMed: 15914712]

    5. Gronemeyer H, Gustafsson JA, Laudet V. Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov. 2004;3:950– 

    964 [PubMed: 15520817]

    6. Katzenellenbogen JA, O'Malley BW, Katzenellenbogen BS. Tripartite steroid hormone receptor pharmacology: interaction with multiple

    effector sites as a basis for the cell- and promoter-specific action of these hormones. Mol Endocrinol. 1996;10:119–131 [PubMed: 8825552]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    16/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 16/36

    7. Jordan VC. SERMs: meeting the promise of multifunctional medicines. J Natl Cancer Inst. 2007;99:350–356 [PubMed: 17341726]

    8. Jordan VC, O'Malley BW. Selective estrogen-receptor modulators and antihormonal resistance in breast cancer. J Clin Oncol.

    2007;25:5815–5824 [PubMed: 17893378]

    9. Jordan VC. Antiestrogens and selective estrogen receptor modulators as multifunctional medicines. 1. Receptor interactions. J Med Chem.

    2003;46:883–908 [PubMed: 12620065]

    10. Jordan VC. Antiestrogens and selective estrogen receptor modulators as multifunctional medicines. 2. Clinical considerations and newagents. J Med Chem. 2003;46:1081–1111 [PubMed: 12646017]

    11. Timsit YE, Negishi M. CAR and PXR: the xenobiotic-sensing receptors. Steroids. 2007;72:231–246 [PMCID: PMC1950246]

    [PubMed: 17284330]

    12. Willson TM, Brown PJ, Sternbach DD, Henke BR. The PPARs: from orphan receptors to drug discovery. J Med Chem. 2000;43:527– 

    550 [PubMed: 10691680]

    13. Tobin JF, Freedman LP. Nuclear receptors as drug targets in metabolic diseases: new approaches to therapy. Trends Endocrinol Metab.

    2006;17:284–290 [PubMed: 16870465]

    14. Shi Y. Orphan nuclear receptors in drug discovery. Drug Discov Today. 2007;12:440–445 [PMCID: PMC2748783]

    [PubMed: 17532527]

    15. Wang Z, Benoit G, Liu J, et al. Structure and function of Nurr1 identifies a class of ligand-independent nuclear receptors. Nature.

    2003;423:555–560 [PubMed: 12774125]

    16. Nagy L, Schwabe JW. Mechanism of the nuclear receptor molecular switch. Trends Biochem Sci. 2004;29:317–324

    [PubMed: 15276186]

    17. Li Y, Lambert MH, Xu HE. Activation of nuclear receptors: a perspective from structural genomics. Structure. 2003;11:741–746

    [PubMed: 12842037]

    18. Riggins RB, Mazzotta MM, Maniya OZ, Clarke R. Orphan nuclear receptors in breast cancer pathogenesis and therapeutic response.

    Endocr Relat Cancer. 2010;17:R213–231 [PMCID: PMC3518023] [PubMed: 20576803]

    19. Lee SO, Li X, Khan S, Safe S. Targeting NR4A1 (TR3) in cancer cells and tumors. Expert Opin Ther Targets. 2011;15:195–206

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    17/36

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 17/36

    [PMCID: PMC4407471] [PubMed: 21204731]

    20. Safe S, Kim K, Li X, Lee SO. NR4A orphan receptors and cancer. Nucl Recept Signal Atlas (NURSA). 2011;9:e002

    21. Muscat GE, Eriksson NA, Byth K, et al. Research resource: nuclear receptors as transcriptome: discriminant and prognostic value in breast

    cancer. Mol Endocrinol. 2013;27:350–365 [PubMed: 23292282]

    22. Jeong Y, Xie Y, Lee W, et al. Research resource: Diagnostic and therapeutic potential of nuclear receptor expression in lung cancer. Mol

    Endocrinol. 2012;26:1443–1454 [PMCID: PMC3404298] [PubMed: 22700587]

    23. Jeong Y, Xie Y, Xiao G, et al. Nuclear receptor expression defines a set of prognostic biomarkers for lung cancer. PLoS Med.

    2010;7:e1000378. [PMCID: PMC3001894] [PubMed: 21179495]

    24. Holbeck S, Chang J, Best AM, Bookout AL, Mangelsdorf DJ, Martinez ED. Expression profiling of nuclear receptors in the NCI60 cancer 

    cell panel reveals receptor-drug and receptor-gene interactions. Mol Endocrinol. 2010;24:1287–1296 [PMCID: PMC2875807]

    [PubMed: 20375240]

    25. Lalli E, Sassone-Corsi P. DAX-1, an unusual orphan receptor at the crossroads of steroidogenic function and sexual differentiation. Mol

    Endocrinol. 2003;17:1445–1453 [PubMed: 12775766]

    26. Lalli E, Alonso J. Targeting DAX-1 in embryonic stem cells and cancer. Expert Opin Ther Targets. 2010;14:169–177

    [PubMed: 20055716]

    27. Ehrlund A, Treuter E. Ligand-independent actions of the orphan receptors/corepressors DAX-1 and SHP in metabolism, reproduction and

    disease. J Steroid Biochem Mol Biol. 2012;130:169–179 [PubMed: 21550402]

    28. Guo W, Burris TP, McCabe ER. Expression of DAX-1, the gene responsible for X-linked adrenal hypoplasia congenita and

    hypogonadotropic hypogonadism, in the hypothalamic-pituitary-adrenal/gonadal axis. Biochem Mol Med. 1995;56:8–13 [PubMed: 8593542]

    29. Bardoni B, Zanaria E, Guioli S, et al. A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet.

    1994;7:497–501 [PubMed: 7951319]

    30. Zanaria E, Muscatelli F, Bardoni B, et al. An unusual member of the nuclear hormone receptor superfamily responsible for X-linked adrenal

    hypoplasia congenita. Nature. 1994;372:635–641 [PubMed: 7990953]

    31. Muscatelli F, Strom TM, Walker AP, et al. Mutations in the DAX-1 gene give rise to both X-linked adrenal hypoplasia congenita and

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    18/36

    p p g g

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 18/36

    hypogonadotropic hypogonadism. Nature. 1994;372:672–676 [PubMed: 7990958]

    32. Conde I, Alfaro JM, Fraile B, Ruíz A, Paniagua R, Arenas MI. DAX-1 expression in human breast cancer: comparison with estrogen

    receptors ER-α, ER-β and androgen receptor status. Breast Cancer Res. 2004;6:R140–148 [PMCID: PMC400665] [PubMed: 15084237]

    33. Zhang H, Slewa A, Janssen E, et al. The prognostic value of the orphan nuclear receptor DAX-1 (NROB1) in node-negative breast cancer.

    Anticancer Res. 2011;31:443–449 [PubMed: 21378322]

    34. Saito S, Ito K, Suzuki T, et al. Orphan nuclear receptor DAX-1 in human endometrium and its disorders. Cancer Sci. 2005;96:645–652[PubMed: 16232195]

    35. Abd-Elaziz M, Akahira J, Moriya T, Suzuki T, Yaegashi N, Sasano H. Nuclear receptor DAX-1 in human common epithelial ovarian

    carcinoma: an independent prognostic factor of clinical outcome. Cancer Sci. 2003;94:980–985 [PubMed: 14611675]

    36. Kaneko T, Kojima Y, Umemoto Y, Sasaki S, Hayashi Y, Kohri K. Usefulness of transcription factors Ad4BP/SF-1 and DAX-1 as

    immunohistologic markers for diagnosis of advanced adrenocortical carcinoma. Horm Res. 2008;70:294–299 [PubMed: 18824868]

    37. Ikuyama S, Mu YM, Ohe K, et al. Expression of an orphan nuclear receptor DAX-1 in human pituitary adenomas. Clin Endocrinol (Oxf).

    1998;48:647–654 [PubMed: 9666878]

    38. Oda T, Tian T, Inoue M, et al. Tumorigenic role of orphan nuclear receptor NR0B1 in lung adenocarcinoma. Am J Pathol.

    2009;175:1235–1245 [PMCID: PMC2731142] [PubMed: 19644015]

    39. Nakamura Y, Suzuki T, Arai Y, Sasano H. Nuclear receptor DAX1 in human prostate cancer: a novel independent biological modulator.

    Endocr J. 2009;56:39–44 [PubMed: 18827407]

    40. García-Aragoncillo E, Carrillo J, Lalli E, et al. DAX1, a direct target of EWS/FLI1 oncoprotein, is a principal regulator of cell-cycle

     progression in Ewing's tumor cells. Oncogene. 2008;27:6034–6043 [PubMed: 18591936]

    41. Kinsey M, Smith R, Iyer AK, McCabe ER, Lessnick SL. EWS/FLI and its downstream target NR0B1 interact directly to modulate

    transcription and oncogenesis in Ewing's sarcoma. Cancer Res. 2009;69:9047–9055 [PMCID: PMC2789197] [PubMed: 19920188]

    42. Kinsey M, Smith R, Lessnick SL. NR0B1 is required for the oncogenic phenotype mediated by EWS/FLI in Ewing's sarcoma. Mol Cancer 

    Res. 2006;4:851–859 [PubMed: 17114343]

    43. Mendiola M, Carrillo J, García E, et al. The orphan nuclear receptor DAX1 is up-regulated by the EWS/FLI1 oncoprotein and is highly

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    19/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 19/36

    expressed in Ewing tumors. Int J Cancer. 2006;118:1381–1389 [PubMed: 16206264]

    44. Gangwal K, Sankar S, Hollenhorst PC, et al. Microsatellites as EWS/FLI response elements in Ewing's sarcoma. Proc Natl Acad Sci

    USA. 2008;105:10149–10154 [PMCID: PMC2481306] [PubMed: 18626011]

    45. Seol W, Choi HS, Moore DD. An orphan nuclear hormone receptor that lacks a DNA binding domain and heterodimerizes with other 

    receptors. Science. 1996;272:1336–1339 [PubMed: 8650544]

    46. Lee HK, Lee YK, Park SH, et al. Structure and expression of the orphan nuclear receptor SHP gene. J Biol Chem. 1998;273:14398– 14402 [PubMed: 9603951]

    47. Zhang Y, Hagedorn CH, Wang L. Role of nuclear receptor SHP in metabolism and cancer. Biochim Biophys Acta. 2011;1812:893–908

    [PMCID: PMC3043166] [PubMed: 20970497]

    48. Zhang Y, Xu P, Park K, Choi Y, Moore DD, Wang L. Orphan receptor small heterodimer partner suppresses tumorigenesis by modulating

    cyclin D1 expression and cellular proliferation. Hepatology. 2008;48:289–298 [PMCID: PMC3800167] [PubMed: 18537191]

    49. He N, Park K, Zhang Y, Huang J, Lu S, Wang L. Epigenetic inhibition of nuclear receptor small heterodimer partner is associated with and

    regulates hepatocellular carcinoma growth. Gastroenterology. 2008;134:793–802 [PubMed: 18325392]

    50. Wilczek E, Szparecki G, Lukasik D, et al. Loss of the orphan nuclear receptor SHP is more pronounced in fibrolamellar carcinoma than in

    typical hepatocellular carcinoma. PLoS One. 2012;7:e30944. [PMCID: PMC3264646] [PubMed: 22292081]

    51. Park MJ, Kim KH, Kim HY, Kim K, Cheong J. Bile acid induces expression of COX-2 through the homeodomain transcription factor 

    CDX1 and orphan nuclear receptor SHP in human gastric cancer cells. Carcinogenesis. 2008;29:2385–2393 [PubMed: 18775915]

    52. Giguère V. Orphan nuclear receptors: from gene to function. Endocr Rev. 1999;20:689–725 [PubMed: 10529899]

    53. Yin L, Lazar MA. The orphan nuclear receptor Rev-erbα recruits the N-CoR/histone deacetylase 3 corepressor to regulate the circadian

    Bmal1 gene. Mol Endocrinol. 2005;19:1452–1459 [PubMed: 15761026]

    54. Crumbley C, Burris TP. Direct regulation of CLOCK expression by REV-ERB. PLoS One. 2011;6:e17290. [PMCID: PMC3066191]

    [PubMed: 21479263]

    55. Preitner N, Damiola F, Lopez-Molina L, et al. The orphan nuclear receptor REV-ERBα controls circadian transcription within the positive

    limb of the mammalian circadian oscillator. Cell. 2002;110:251–260 [PubMed: 12150932]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    20/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 20/36

    56. Chin K, DeVries S, Fridlyand J, et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell.

    2006;10:529–541 [PubMed: 17157792]

    57. Davis LM, Harris C, Tang L, et al. Amplification patterns of three genomic regions predict distant recurrence in breast carcinoma. J Mol

    Diagn. 2007;9:327–336 [PMCID: PMC1899419] [PubMed: 17591932]

    58. Lee YF, Lee HJ, Chang C. Recent advances in the TR2 and TR4 orphan receptors of the nuclear receptor superfamily. J Steroid Biochem

    Mol Biol. 2002;81:291–308 [PubMed: 12361719]

    59. Fang F, Zheng Q, Zhang J, et al. Testicular orphan nuclear receptor 4-associated protein 16 promotes non-small cell lung carcinoma by

    activating estrogen receptor β and blocking testicular orphan nuclear receptor 2. Oncol Rep. 2013;29:297–305 [PMCID: PMC3583481]

    [PubMed: 23129017]

    60. Monaghan AP, Grau E, Bock D, Schütz G. The mouse homolog of the orphan nuclear receptor tailless is expressed in the developing

    forebrain. Development. 1995;121:839–853 [PubMed: 7720587]

    61. Zhang CL, Zou Y, Yu RT, Gage FH, Evans RM. Nuclear receptor TLX prevents retinal dystrophy and recruits the corepressor atrophin1.

    Genes Dev. 2006;20:1308–1320 [PMCID: PMC1472905] [PubMed: 16702404]

    62. Zhang CL, Zou Y, He W, Gage FH, Evans RM. A role for adult TLX-positive neural stem cells in learning and behaviour. Nature.

    2008;451:1004–1007 [PubMed: 18235445]

    63. Li W, Sun G, Yang S, Qu Q, Nakashima K, Shi Y. Nuclear receptor TLX regulates cell cycle progression in neural stem cells of the

    developing brain. Mol Endocrinol. 2008;22:56–64 [PMCID: PMC2194628] [PubMed: 17901127]

    64. Niu W, Zou Y, Shen C, Zhang CL. Activation of postnatal neural stem cells requires nuclear receptor TLX. J Neurosci. 2011;31:13816– 

    13828 [PMCID: PMC3192402] [PubMed: 21957244]

    65. Sim FJ, Keyoung HM, Goldman JE, et al. Neurocytoma is a tumor of adult neuronal progenitor cells. J Neurosci. 2006;26:12544–12555[PubMed: 17135416]

    66. Phillips HS, Kharbanda S, Chen R, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease

     progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–173 [PubMed: 16530701]

    67. Liu HK, Wang Y, Belz T, et al. The nuclear receptor tailless induces long-term neural stem cell expansion and brain tumor initiation. Genes

    Dev. 2010;24:683–695 [PMCID: PMC2849125] [PubMed: 20360385]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    21/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 21/36

    68. Park HJ, Kim JK, Jeon HM, et al. The neural stem cell fate determinant TLX promotes tumorigenesis and genesis of cells resembling glioma

    stem cells. Mol Cells. 2010;30:403–408 [PubMed: 20814749]

    69. Zou Y, Niu W, Qin S, Downes M, Burns DK, Zhang CL. The nuclear receptor TLX is required for gliomagenesis within the adult

    neurogenic niche. Mol Cell Biol. 2012;32:4811–4820 [PMCID: PMC3497606] [PubMed: 23028043]

    70. Zeng ZJ, Johansson E, Hayashi A, et al. TLX controls angiogenesis through interaction with the von Hippel-Lindau protein. Biol Open.

    2012;1:527–535 [PMCID: PMC3509445] [PubMed: 23213445]

    71. Kobayashi M, Takezawa S, Hara K, et al. Identification of a photoreceptor cell-specific nuclear receptor. Proc Natl Acad Sci U S A.

    1999;96:4814–4819 [PMCID: PMC21774] [PubMed: 10220376]

    72. Milam AH, Rose L, Cideciyan AV, et al. The nuclear receptor NR2E3 plays a role in human retinal photoreceptor differentiation and

    degeneration. Proc Natl Acad Sci USA. 2002;99:473–478 [PMCID: PMC117584] [PubMed: 11773633]

    73. Eichen JG, Dalmau J, Demopoulos A, Wade D, Posner JB, Rosenfeld MR. The photoreceptor cell-specific nuclear receptor is an

    autoantigen of paraneoplastic retinopathy. J Neuroophthalmol. 2001;21:168–172 [PubMed: 11725181]

    74. Bracci PM, Zhou M, Young S, Wiemels J. Serum autoantibodies to pancreatic cancer antigens as biomarkers of pancreatic cancer in a SanFrancisco Bay Area case-control study. Cancer. 2012;118:5384–5394 [PMCID: PMC3414682] [PubMed: 22517435]

    75. Park YY, Kim K, Kim SB, et al. Reconstruction of nuclear receptor network reveals that NR2E3 is a novel upstream regulator of ESR1 in

     breast cancer. EMBO Mol Med. 2012;4:52–67 [PMCID: PMC3376834] [PubMed: 22174013]

    76. Tsai SY, Tsai MJ. Chick ovalbumin upstream promoter-transcription factors (COUP-TFs): coming of age. Endocr Rev. 1997;18:229–240

    [PubMed: 9101138]

    77. Lin FJ, Qin J, Tang K, Tsai SY, Tsai MJ. Coup d'Etat: an orphan takes control. Endocr Rev. 2011;32:404–421 [PMCID: PMC3365794]

    [PubMed: 21257780]

    78. Litchfield LM, Klinge CM. Multiple roles of COUP-TFII in cancer initiation and progression. J Mol Endocrinol. 2012;49:R135–R148

    [PMCID: PMC3469769] [PubMed: 22966133]

    79. Qin J, Wu SP, Creighton CJ, et al. COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis. Nature.

    2013;493:236–240 [PMCID: PMC4022346] [PubMed: 23201680]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    22/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 22/36

    80. Perets R, Kaplan T, Stein I, et al. Genome-wide analysis of androgen receptor targets reveals COUP-TF1 as a novel player in human

     prostate cancer. PLoS One. 2012;7:e46467. [PMCID: PMC3464259] [PubMed: 23056316]

    81. Nagasaki S, Suzuki T, Miki Y, et al. Chicken ovalbumin upstream promoter transcription factor II in human breast carcinoma: possible

    regulator of lymphangiogenesis via vascular endothelial growth factor-C expression. Cancer Sci. 2009;100:639–645 [PubMed: 19154418]

    82. Litchfield LM, Riggs KA, Hockenberry AM, et al. Identification and characterization of nucleolin as a COUP-TFII coactivator of retinoic

    acid receptor β transcription in breast cancer cells. PLoS One. 2012;7:e38278. [PMCID: PMC3365040] [PubMed: 22693611]

    83. Hawkins SM, Loomans HA, Wan YW, et al. Expression and functional pathway analysis of nuclear receptor NR2F2 in ovarian cancer. J

    Clin Endocrinol Metab. 2013;98:E1152–1162 [PMCID: PMC3701283] [PubMed: 23690307]

    84. De Sousa Damião R, Fujiyama Oshima CT, Stávale JN, Gonçalves WJ. Analysis of the expression of estrogen receptor, progesterone

    receptor and chicken ovalbumin upstream promoter-transcription factor I in ovarian epithelial cancers and normal ovaries. Oncol Rep.

    2007;18:25–32 [PubMed: 17549341]

    85. Shin SW, Kwon HC, Rho MS, Choi HJ, Kwak JY, Park JI. Clinical significance of chicken ovalbumin upstream promoter-transcription

    factor II expression in human colorectal cancer. Oncol Rep. 2009;21:101–106 [PubMed: 19082449]

    86. Jonk LJ, de Jonge ME, Pals CE, et al. Cloning and expression during development of three murine members of the COUP family of nuclear 

    orphan receptors. Mech Dev. 1994;47:81–97 [PubMed: 7947324]

    87. Miyajima N, Kadowaki Y, Fukushige S, et al. Identification of two novel members of erbA superfamily by molecular cloning: the gene

     products of the two are highly related to each other. Nucleic Acids Res. 1988;16:11057–11074 [PMCID: PMC338996] [PubMed: 2905047]

    88. Kruse SW, Suino-Powell K, Zhou XE, et al. Identification of COUP-TFII orphan nuclear receptor as a retinoic acid-activated receptor.

    PLoS Biol. 2008;6:e227. [PMCID: PMC2535662] [PubMed: 18798693]

    89. Warnecke M, Oster H, Revelli JP, Alvarez-Bolado G, Eichele G. Abnormal development of the locus coeruleus in Ear2(Nr2f6)-deficientmice impairs the functionality of the forebrain clock and affects nociception. Genes Dev. 2005;19:614–625 [PMCID: PMC551581]

    [PubMed: 15741322]

    90. Sakhinia E, Glennie C, Hoyland JA, et al. Clinical quantitation of diagnostic and predictive gene expression levels in follicular and diffuse

    large B-cell lymphoma by RT-PCR gene expression profiling. Blood. 2007;109:3922–3928 [PubMed: 17255358]

    91. Qin J, Chen X, Xie X, Tsai MJ, Tsai SY. COUP-TFII regulates tumor growth and metastasis by modulating tumor angiogenesis. Proc Natl

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    23/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 23/36

    Acad Sci USA. 2010;107:3687–3692 [PMCID: PMC2840495] [PubMed: 20133706]

    92. Maxwell MA, Muscat GE. The NR4A subgroup: immediate early response genes with pleiotropic physiological roles. Nucl Recept Signal.

    2006;4:e002. [PMCID: PMC1402209] [PubMed: 16604165]

    93. Pearen MA, Muscat GE. Minireview: Nuclear hormone receptor 4A signaling: implications for metabolic disease. Mol Endocrinol.

    2010;24:1891–1903 [PubMed: 20392876]

    94. Lee SO, Andey T, Jin UH, et al. The nuclear receptor TR3 regulates mTORC1 signaling in lung cancer cells expressing wild-type p53.Oncogene. 2012;31:3265–3276 [PMCID: PMC3299891] [PubMed: 22081070]

    95. Cho SD, Yoon K, Chintharlapalli S, et al. Nur77 agonists induce proapoptotic genes and responses in colon cancer cells through nuclear 

    receptor-dependent and nuclear receptor-independent pathways. Cancer Res. 2007;67:674–683 [PubMed: 17234778]

    96. Lee SO, Abdelrahim M, Yoon K, et al. Inactivation of the orphan nuclear receptor TR3/Nur77 inhibits pancreatic cancer cell and tumor 

    growth. Cancer Res. 2010;70:6824–6836 [PMCID: PMC2988472] [PubMed: 20660371]

    97. Alexopoulou AN, Leao M, Caballero OL, et al. Dissecting the transcriptional networks underlying breast cancer: NR4A1 reduces the

    migration of normal and breast cancer cell lines. Breast Cancer Res. 2010;12:R51. [PMCID: PMC2949640] [PubMed: 20642837]

    98. Mullican SE, Zhang S, Konopleva M, et al. Abrogation of nuclear receptors Nr4a3 and Nr4a1 leads to development of acute myeloid

    leukemia. Nat Med. 2007;13:730–735 [PubMed: 17515897]

    99. Ke N, Claassen G, Yu DH, et al. Nuclear hormone receptor NR4A2 is involved in cell transformation and apoptosis. Cancer Res.

    2004;64:8208–8212 [PubMed: 15548686]

    100. Inamoto T, Czerniak BA, Dinney CP, Kamat AM. Cytoplasmic mislocalization of the orphan nuclear receptor Nurr1 is a prognostic factor 

    in bladder cancer. Cancer. 2010;116:340–346 [PubMed: 19908257]

    101. Llopis S, Singleton B, Duplessis T, Carrier L, Rowan B, Williams C. Dichotomous roles for the orphan nuclear receptor NURR1 in breast

    cancer. BMC Cancer. 2013;13:139. [PMCID: PMC3617898] [PubMed: 23517088]

    102. Wang J, Yang J, Zou Y, Huang GL, He ZW. Orphan nuclear receptor nurr1 as a potential novel marker for progression in human prostate

    cancer. Asian Pac J Cancer Prev. 2013;14:2023–2028 [PubMed: 23679312]

    103. Han Y, Cai H, Ma L, et al. Nuclear orphan receptor NR4A2 confers chemoresistance and predicts unfavorable prognosis of colorectal

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    i i h i d i h h C 2013 49 3420 3430 [ b d 23809 6 ]

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    24/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 24/36

    carcinoma patients who received postoperative chemotherapy. Eur J Cancer. 2013;49:3420–3430 [PubMed: 23809767]

    104. Han Y, Cai H, Ma L, et al. Expression of orphan nuclear receptor NR4A2 in gastric cancer cells confers chemoresistance and predicts an

    unfavorable postoperative survival of gastric cancer patients with chemotherapy. Cancer. 2013;119:3436–3445 [PubMed: 23821160]

    105. Kelly VR, Xu B, Kuick R, Koenig RJ, Hammer GD. Dax1 up-regulates Oct4 expression in mouse embryonic stem cells via LRH-1 and

    SRA. Mol Endocrinol. 2010;24:2281–2291 [PMCID: PMC2999479] [PubMed: 20943815]

    106. Zhang H, Thomsen JS, Johansson L, Gustafsson JA, Treuter E. DAX-1 functions as an LXXLL-containing corepressor for activatedestrogen receptors. J Biol Chem. 2000;275:39855–39859 [PubMed: 11053406]

    107. Ito M, Yu R, Jameson JL. DAX-1 inhibits SF-1-mediated transactivation via a carboxy-terminal domain that is deleted in adrenal

    hypoplasia congenita. Mol Cell Biol. 1997;17:1476–1483 [PMCID: PMC231873] [PubMed: 9032275]

    108. Sun C, Nakatake Y, Akagi T, et al. Dax1 binds to Oct3/4 and inhibits its transcriptional activity in embryonic stem cells. Mol Cell Biol.

    2009;29:4574–4583 [PMCID: PMC2725734] [PubMed: 19528230]

    109. Crawford PA, Dorn C, Sadovsky Y, Milbrandt J. Nuclear receptor DAX-1 recruits nuclear receptor corepressor N-CoR to

    steroidogenic factor 1. Mol Cell Biol. 1998;18:2949–2956 [PMCID: PMC110674] [PubMed: 9566914]

    110. Holter E, Kotaja N, Mäkela S, et al. Inhibition of androgen receptor (AR) function by the reproductive orphan nuclear receptor DAX-1.

    Mol Endocrinol. 2002;16:515–528 [PubMed: 11875111]

    111. Zhou J, Oakley RH, Cidlowski JA. DAX-1 (dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X-

    chromosome, gene 1) selectively inhibits transactivation but not transrepression mediated by the glucocorticoid receptor in a LXXLL-dependent

    manner. Mol Endocrinol. 2008;22:1521–1534 [PMCID: PMC2453603] [PubMed: 18417736]

    112. Xu B, Yang WH, Gerin I, Hu CD, Hammer GD, Koenig RJ. Dax-1 and steroid receptor RNA activator (SRA) function as transcriptional

    coactivators for steroidogenic factor 1 in steroidogenesis. Mol Cell Biol. 2009;29:1719–1734 [PMCID: PMC2655620] [PubMed: 19188450]

    113. Nedumaran B, Hong S, Xie YB, et al. DAX-1 acts as a novel corepressor of orphan nuclear receptor HNF4α and negatively regulates

    gluconeogenic enzyme gene expression. J Biol Chem. 2009;284:27511–27523 [PMCID: PMC2785680] [PubMed: 19651776]

    114. Nedumaran B, Kim GS, Hong S, et al. Orphan nuclear receptor DAX-1 acts as a novel corepressor of liver X receptor α and inhibits

    hepatic lipogenesis. J Biol Chem. 2010;285:9221–9232 [PMCID: PMC2838341] [PubMed: 20080977]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    115 B ̸ A S l S G t f JA T t E T i ti l i b SHP l l h i d h i l i l

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    25/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 25/36

    115. Ba̸vner A, Sanyal S, Gustafsson JA, Treuter E. Transcriptional corepression by SHP: molecular mechanisms and physiological

    consequences. Trends Endocrinol Metab. 2005;16:478–488 [PubMed: 16275121]

    116. Xiao J, Gong AY, Eischeid AN, et al. miR-141 modulates androgen receptor transcriptional activity in human prostate cancer cells through

    targeting the small heterodimer partner protein. Prostate. 2012;72:1514–1522 [PubMed: 22314666]

    117. Yang Z, Zhang Y, Kemper JK, Wang L. Cross-regulation of protein stability by p53 and nuclear receptor SHP. PLoS One.

    2012;7:e39789. [PMCID: PMC3380837] [PubMed: 22737255]

    118. Zhao BX, Chen HZ, et al. p53 mediates the negative regulation of MDM2 by orphan receptor TR3. EMBO J. 2006;25:5703–5715

    [PMCID: PMC1698882] [PubMed: 17139261]

    119. Seol W, Hanstein B, Brown M, Moore DD. Inhibition of estrogen receptor action by the orphan receptor SHP (short heterodimer 

     partner). Mol Endocrinol. 1998;12:1551–1557 [PubMed: 9773978]

    120. Klinge CM, Jernigan SC, Risinger KE. The agonist activity of tamoxifen is inhibited by the short heterodimer partner orphan nuclear 

    receptor in human endometrial cancer cells. Endocrinology. 2002;143:853–867 [PubMed: 11861507]

    121. Dussault I, Giguère V. Differential regulation of the N-myc proto-oncogene by ROR α and RVR, two orphan members of the superfamilyof nuclear hormone receptors. Mol Cell Biol. 1997;17:1860–1867 [PMCID: PMC232033] [PubMed: 9121434]

    122. Downes M, Carozzi AJ, Muscat GE. Constitutive expression of the orphan receptor, Rev-erbA α, inhibits muscle differentiation and

    abrogates the expression of the myoD gene family. Mol Endocrinol. 1995;9:1666–1678 [PubMed: 8614403]

    123. Chini CC, Escande C, Nin V, Chini EN. DBC1 (Deleted in Breast Cancer 1) modulates the stability and function of the nuclear receptor 

    Rev-erbα. Biochem J. 2013;451:453–461 [PMCID: PMC3630992] [PubMed: 23398316]

    124. Negoro H, Okinami T, Kanematsu A, Imamura M, Tabata Y, Ogawa O. Role of Rev-erbα domains for transactivation of the connexin43

     promoter with Sp1. FEBS Lett. 2013;587:98–103 [PubMed: 23201262]

    125. Wang J, Yin L, Lazar MA. The orphan nuclear receptor Rev-erb α regulates circadian expression of plasminogen activator inhibitor type

    1. J Biol Chem. 2006;281:33842–33848 [PubMed: 16968709]

    126. Raspé E, Duez H, Mansén A, et al. Identification of Rev-erbα as a physiological repressor of apoC-III gene transcription. J Lipid Res.

    2002;43:2172–2179 [PubMed: 12454280]

    7/3/2016 Minireview: Role Of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    127 Yang DR Ding XF Luo J et al Increased chemosensitivity via targeting testicular nuclear receptor 4 (TR4) Oct4 interleukin 1 receptor

  • 8/18/2019 Minireview_ Role of Orphan Nuclear Receptors in Cancer and Potential as Drug Targets

    26/36

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3896638/?report=printable 26/36

    127. Yang DR, Ding XF, Luo J, et al. Increased chemosensitivity via targeting testicular nuclear receptor 4 (TR4)-Oct4-interleukin 1 receptor 

    antagonist (IL1Ra) axis in prostate cancer CD133+ stem/progenitor cells to battle prostate cancer. J Biol Chem. 2013;288:16476–16483

    [PMCID: PMC3675583] [PubMed: 23609451]

    128. Shyr CR, Hu YC, Kim E, Chang C. Modulation of estrogen receptor-mediated transactivation by orphan receptor TR4 in MCF-7 cells. J

    Biol Chem. 2002;277:14622–14628 [PubMed: 11844790]

    129. Hu YC, Shyr CR, Che W, Mu XM, Kim E, Chang C. Suppression of estrogen receptor-mediated transcription and cell growth by

    interaction with TR2 orphan receptor. J Biol Chem. 2002;277:33571–33579 [PubMed: 12093804]

    130. Mu X, Chang C. TR2 orphan receptor functions as negative modulator for androgen receptor in prostate cancer cells PC-3. Prostate.

    2003;57:129–133 [PubMed: 12949936]

    131. Lin TM, Young WJ, Chang C. Multiple functions of the TR2–11 orphan receptor in modulating activation of two key c