Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

Clinical Cancer Molecular Pathways Research

Molecular Pathways: The Role of NR4A Orphan Nuclear Receptors in Cancer

Helen M. Mohan1,2, Carol M. Aherne3, Ailin C. Rogers1,2, Alan W. Baird1, Des C. Winter2, and Evelyn P. Murphy1

Abstract Nuclear receptors are of integral importance in carcinogenesis. Manipulation of classic ligand-activated nuclear receptors, such as estrogen blockade in breast cancer, is an important established cancer therapy. Orphan nuclear receptors, such as nuclear family 4 subgroup A (NR4A) receptors, have no known natural ligand(s). These elusive receptors are increasingly recognized as molecular switches in cell survival and a molecular link between inflammation and cancer. NR4A receptors act as transcription factors, altering expression of downstream in apoptosis (Fas-ligand, TRAIL), proliferation, DNA repair, metabolism, cell migration, inflammation (interleukin-8), and angiogenesis (VEGF). NR4A receptors are modulated by multiple cell-signaling pathways, including protein kinase A/CREB, NF-kB, phosphoinosi-

tide 3-kinase/AKT, c-jun-NH2-kinase, Wnt, and mitogen-activated protein kinase pathways. NR4A receptor effects are context and tissue specific, influenced by their levels of expression, posttranslational modi- fication, and interaction with other transcription factors (RXR, PPAR-¡). The subcellular location of NR4A "nuclear receptors" is also important functionally; novel roles have been described in the cytoplasm where NR4A proteins act both indirectly and directly on the mitochondria to promote apoptosis via Bcl-2. NR4A receptors are implicated in a wide variety of malignancies, including breast, lung, colon, bladder, and prostate cancer; glioblastoma multiforme; sarcoma; and acute and/or chronic myeloid leukemia. NR4A receptors modulate response to conventional chemotherapy and represent an exciting frontier for chemotherapeutic intervention, as novel agents targeting NR4A receptors have now been developed. This review provides a concise clinical overview of current knowledge of NR4A signaling in cancer and the potential for therapeutic manipulation. Clin Cancer Res; 18(12); 1–6. 2012 AACR.

Background which putative ligands have since been discovered. Diverse Targeting ligand-activated steroid nuclear receptors is an and paradoxical transcriptional and direct roles have been important established cancer therapy. Orphan nuclear described for these receptors; physiologic functions of NR4A receptors are similar to steroid nuclear receptors as they act receptors are context and tissue specific. NR4A receptors as transcription factors to modulate downstream have emerged as important molecular switches in processes expression. However, orphan nuclear receptors have no associated with carcinogenesis, including apoptosis, DNA known natural ligand(s). These intriguing receptors com- repair, proliferation, migration, inflammation, metabolism, prise more than half the total number of nuclear receptors. and angiogenesis (Fig. 1). Unlike classic steroid hormone Nuclear family 4 subgroup A (NR4A) orphan nuclear recep- receptors, which need to be activated by a ligand, these tors NR4A1 (Nur77, 3, nerve growth proteins are constitutively active (1). Emerging techniques in vivo factor 1b), NR4A2 (Nur-related factor 1), and NR4A3 (neu- using cell-specific knockdown of NR4A receptors ron-derived orphan receptor 1, mitogen inducible orphan have recently accelerated discovery of NR4A receptor func- ) are thought to be incapable of classic tions (2). The actions of NR4A receptors depend on NR4A ligand binding due to bulky ligand-binding domains, unlike receptor subcellular localization, levels of NR4A expression, "adopted" receptors, for example, PPAR-g (NR1C3), in transcriptional modulation by coactivators and/or corepres- sors, posttranslational modification, and interaction with other nuclear receptors (3, 4). Authors' Affiliations: 1UCD Veterinary Sciences Centre, University Col- lege Dublin, 2Department of Surgery, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; 3Mucosal Inflammation Program, Department of NR4A Receptors and Cancer Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado Apoptosis Corresponding Author: Helen M. Mohan, St. Vincent's University Hospi- Inhibition of apoptosis. NR4A receptors can promote cell tal, Elm Park, Dublin 4, Ireland. Phone: 353-85-145-2384; Fax: 353-221- growth and survival, activating transcription of downstream 4001; E-mail: [email protected] antiapoptotic and proproliferative genes. Physiologically, doi: 10.1158/1078-0432.CCR-11-2953 NR4A2 is essential for dopaminergic neuronal survival 2012 American Association for Cancer Research. in the central nervous system, and reduced NR4A2 is

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

Mohan et al.

NF-κB PI3K/JNK Mitogens MAP kinase Cytokines cAMP/PKA/CREB Eicosanoids Hypoxia

Cytoplasmic Nuclear NR4A NR4A

P53 PKC/PKD PARP-1 SRC-1 MAP kinase PKA/CREB DNA-PK PGC1α Wnt/β-catenin NF-κB MC1R MAP kinase

VEGF Mitochondria

Apoptosis Proliferation Angiogenesis DNA repair Fatty acid metabolism

Figure 1. The NR4A receptors are involved in a myriad of cellular functions that contribute to cancer, including angiogenesis, apoptosis, metabolism, migration, proliferation, and DNA repair. Expression of the NR4A receptors is induced by a variety of stimuli via multiple cell-signaling pathways, including protein kinase A (PKA)/CREB, NF-kB, phosphoinositide 3-kinase (PI3K)/AKT, c-jun-NH2-kinase (JNK), and MAPK pathways. MAP, mitogen-activated protein; MC1R, melanocortin 1 receptor; PGC1a, peroxisome proliferator-activated receptor g coactivator 1-a; PK, protein kinase; PKC, protein kinase C; PKD, protein kinase D; SRC-1, steroid receptor coactivator 1.

implicated in Parkinson disease (5). In vitro, NR4A2 inhibits proliferation by transcriptional inhibition of Wnt signaling -mediated induction of downstream proapoptotic (11, 12). genes like BAX, a proapoptotic member of the Bcl-2 family Proapoptotic roles for the NR4A receptors. Proapoptotic (6). NR4A2 also inhibits apoptosis via convergence with roles have also been described for the NR4A receptors. Wnt and mitogen-activated protein kinase (MAPK) path- Nuclear export of NR4A1 is important functionally, as ways (7, 8). Kitagawa and colleagues have reported that cytosolic NR4A1 has a nongenomic proapoptotic role in b-catenin binds NR4A2, releasing NR4A2 from the core- cancer cell lines in vitro. NR4A1 induces apoptosis by direct pressor protein Lef-1 in 293F cells, allowing transcription of interaction with Bcl-2 in the mitochondria, exposing proa- downstream Wnt and NR4A2 targets. Physiologically, these poptotic BH3, or indirectly by stimulating other cytosolic interactions are required for normal neuronal development proapoptotic proteins, such as BAX binding to the mito- and the survival of dopaminergic neurons (7). NR4A recep- chondria to initiate the apoptotic cascade (Fig. 1A; refs. 13– tors and b-catenin modulate each other’s transcriptional 16). Bcl-2 modulation by NR4A1 is also important phys- activity in a cell-specific manner (7, 9). In colon cancer cell iologically in the negative selection of T cells (13). Nuclear lines, a bile acid carcinogen (deoxycholic acid) has been NR4A receptors can also have proapoptotic effects by induc- shown to stimulate b-catenin–dependent increased expres- ing proapoptotic and antiproliferative genes (1, 17). sion of NR4A1 (10). Conversely, NR4A1 has been shown to promote degradation of cytoplasmic b-catenin in a tran- DNA repair scription-independent mechanism, whereas in murine A novel function for NR4A receptors in DNA double- models, NR4A1 has been shown to reduce tumor cell strand break (DSB) repair has recently been identified (18).

OF2 Clin Cancer Res; 18(12) June 15, 2012 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

NR4A Receptors in Cancer

NR4A receptors translocate to sites of double-strand DNA activity of NR4A1 and NR4A2 is involved in VEGF-mediated damage in a mechanism dependent on PARP-1 and are angiogenesis (28, 29). phosphorylated by DNA protein kinases. Interestingly, the DNA repair action of NR4A receptors is not dependent on NR4A receptors and chromosomal translocations their transcriptional activity, but rather due to a direct Chromosomal translocation of NR4A receptors can lead interaction at the DNA repair site, the precise mechanisms to oncogenic conversion. For example, translocations of which remain incompletely understood (18). In mela- involving NR4A3 in extraskeletal myxoid chondrosarcoma, noma, repression of NR4A receptors impairs UV-induced most commonly between Ewing sarcoma region-1 (EWS) DNA damage repair via the melanocortin-1 receptor (19). and NR4A3 t(9, 22)(q22; q12), result in a fusion protein, These studies suggest a pro–cell-survival role for NR4As by EWS-NOR1, which activates NR4A3 target genes, such as regulating DNA repair. Conversely, a role for NR4A1 in PPAR-g, and leads to oncogenesis (30). inhibition of DNA repair has been described in hepatocel- lular carcinoma cell lines (20). Clinical–Translational Advances Inflammation NR4A receptors: identified roles in human cancer NR4A receptors are an important molecular link between Altered NR4A receptor expression has been identified in inflammation and cancer. Proinflammatory effects of many solid tumors, with a plethora of in vitro roles described NR4A receptors are seen in the tumor-like growth of (Table 1). Meanwhile, reduced expression of NR4A1 and pannus in rheumatoid arthritis, in which inflammatory NR4A3 receptors is associated with hematologic malignan- synovial hyperplasia becomes an invasive front of destruc- cies, including acute myeloid leukemia and chronic mye- tive tissue causing cartilage damage (3). Conversely, anti- lodysplastic and/or myeloproliferative disease (31). inflammatory effects have been shown in transformed Novel drug targets. Drug development targeting NR4A normal vascular macrophages, endothelial cells in athero- receptor signaling is challenging due to the lack of a natural sclerosis, and in the central nervous system (21, 22). Proin- ligand, their bulky ligand-binding domains, the diversity of flammatory prostaglandins are strongly implicated in can- their cell- and context-specific functions, and redundancy cer; prostaglandin E2 (PGE2) induces NR4A2 in colon between members of the NR4A family. Despite these chal- cancer, which leads to a myriad of downstream procancer lenges, several strategies have now been developed to enable effects. Meanwhile, COX-2 inhibitors repress NR4A2 therapeutic targeting of NR4A receptor signaling (1). These expression and NR4A-regulated genes, including osteopon- strategies include targeting their expression, nuclear export, tin (23). This repression may represent a mechanism for the and interaction with coactivators and/or repressors. reduced colon cancer risk observed in population studies The proapoptotic nongenomic action of NR4A1in the of nonsteroidal anti-inflammatory drugs (NSAID). NR4A cytoplasm has therapeutic potential and has been manip- receptors are downstream targets of the cAMP-responsive ulated in 2 ways. The first way is by drugs inducing nuclear element binding protein (CREB); binding sites for CREB export of NR4A1. Several drugs already in clinical use are have been identified in the promoter region of all 3 NR4A now recognized to induce nuclear export of NR4A1, includ- receptors. PGE2 induction of NR4A receptors involves CREB ing 5-fluorouracil (5-FU) and certain NSAIDs (15). Novel and NF-kB signaling, as PGE2 induces phosphorylation of drugs targeting nuclear export of NR4A1 include n-butyle- CREB; this phosphorylated CREB can then bind to NR4A nephthalide (BP) and cytosporone B. BP and its derivatives promoters and enhance gene transcription (23). (PCH4) have shown therapeutic potential in glioblastoma multiforme, in vitro and in vivo, and in oral squamous cell Metabolism and angiogenesis carcinoma in vitro (32). PCH4 induces apoptosis in oral NR4A receptors are involved in fatty acid oxidation and squamous cell carcinoma and glioblastoma multiforme cell hepatic glucose metabolism (24). In normal skeletal lines in a mechanism dependent on PCH4-mediated muscle physiology, NR4A3 has been shown to promote increased NR4A1 expression and cytoplasmic translocation fatty acid oxidative pathways and is induced by b-adren- (32, 33). Cytosporone B, a compound isolated from fungi, ergic signaling via protein kinase A, MAPK, and CREB- is a ligand for NR4A1 and induces apoptosis by transactiva- dependent pathways (25, 26). In colon cancer, NR4A2 tion of NR4A1 target genes and by inducing expression and with peroxisome proliferator-activated receptor g coacti- mitochondrial localization of NR4A1 in vivo and in vitro vator 1a (PGC1a) induces expression of fatty acid oxi- (34). dation enzymes, allowing cells to switch to alternative Second, a novel nanopeptide (nu-BCP9) has been oxidative pathways, promoting cell survival (27). derived from NR4A1, which mimics the action of NR4A1 Tumor growth also depends on formation of new blood on mitochondrial Bcl-2 to promote apoptosis (14). As the vessels (angiogenesis) to facilitate delivery of oxygen and functions of the NR4A receptors are further elucidated, nutrients to the tumor. NR4A receptors are downstream further peptide mimetics may be developed to emulate or targets of VEGF, promoting proliferation of endothelial cells block specific actions of the NR4A receptors, at both a in vitro and in vivo (25, 28). Transcription of NR4A receptors nongenomic and transcriptional level. is increased by VEGF, hypoxia, and CREB activation in Another class of drugs that can promote cell death via vascular endothelial cells (25). In vivo, the transcriptional NR4A receptors is the diindoylmethane derivatives (CDIM).

www.aacrjournals.org Clin Cancer Res; 18(12) June 15, 2012 OF3

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

Mohan et al.

Table 1. Summary of NR4A receptors in tumors, showing altered expression of NR4A receptors in tumor tissue and functional effects of NR4A receptors shown in vitro

NR4A expression Cancer (reference) in tumors Function in vitro Drugs

Colon (15, 23, 27, 39) NR4A1þ NR4A2þ Apoptosis A1,A2 proliferation CDIMs, 5-FU, butyrate A1,A2þ metabolism A2þ angiogenesis A1þ Migration A1þ NSAIDs/COX-2 inhibitors Breast (40–43) NR4A1þ Apoptosis A1þ A3þ A2 migration A1 Nu-BCP9, retinoids, CDIMs Melanoma (19, 44, 45) NR4A3þ Damage repair A1þ apoptosis A1, A2,A3þ, angiogenesis A1þ Bladder (4, 46) NR4A1þ NRA2þ Apoptosis A1þ,A2, migration A2þ CDIMs Liver (20, 47) NR4A1þ (HCC), Apoptosis A1þ DNA repair A1 BP NR4A2 (mets) Thyroid (48) NR4A1, NR4A3 Apoptosis A1þ A3þ Lithium Lung (1, 49–51) NR4A1þ (NSCLT) Proliferation A1, apoptosis CDIMs, retinoids, e.g., A1, paraneoplastic CD437 (SCLT-ACTH) Glioblastoma multiforme (32) Apoptosis A1þ BP/PCH4 Cytosporone B Pancreatic (1) NR4A1þ Apoptosis A1 proliferation A1 CDIMs Prostate (1) Apoptosis A1 proliferation A1 CDIMs Gastric (52) NR4A2 Apoptosis A1þ Cytosporone B, chenodeoxycholic acid Extraskeletal myxoid T(9;22)q22; Apoptosis A3, proliferation A3þ chondrosarcoma (30) q12 NR4A3/EWS1 Cervical (45) Proliferation A2þ, apoptosis A1þ, A2, migration A2þ

Ovarian (1, 53) Apoptosis A1þ Vitamin K2 Oral squamous cell carcinoma (33, 37) NR4A2þ Apoptosis A1þ,A2 BP/PCH4, 5-FU (resistance)

NOTE: Drugs targeting NR4A receptors are listed. þ, increased; , decreased. Abbreviations: ACTH, adrenocorticotropic hormone; CDIM, diindoylmethane derivative; HCC, hepatocellular carcinoma; NSCLT, non– small cell lung cancer therapy; SCLT, small cell lung cancer therapy.

CDIMs are unusual because they can cause apoptosis via Chemotherapy resistance. NR4A receptors contribute to induction or inhibition of NR4A1 transcriptional activity, in resistance to chemotherapy; understanding the mechan- addition to NR4A-independent induction of apoptosis isms of this resistance may enable therapeutic targeting. via endoplasmic reticular (ER) stress. The derivative DIM- Induction of NR4A2 by PGE2 in a cAMP/protein kinase C-pPhOCH3 [1, 1-bis (30-indoyl)-1-(p-anisyl)] methane is A–dependent manner promotes resistance to 5-FU in squa- an "activator" of NR4A1-mediated transcription of proapop- mous cell carcinoma (37). This finding has potential rele- totic genes, for example, cystathionase, p21, and ATF3, vance for colorectal cancer, as PGE2-mediated induction of leading to apoptosis in pancreatic cancer cell lines in vitro. NR4A2 also occurs in colon cancer cell lines, whereas 5-FU Intriguingly, DIM-C-pPhOH [1, 1-bis (30-indoyl)-1-p-hydro- is a commonly used chemotherapeutic agent in colorectal xyphenylmethane], an "inhibitor" of NR4A1 nuclear trans- cancer. Similarly, Riggins and colleagues have suggested activation, also causes apoptosis in pancreatic cancer cell roles for NR4A receptors in mediating resistance to doxo- lines, but by reducing transcription of NR4A1-dependent rubicin in breast cancer (6). The DNA repair effect of NR4A antiapoptotic and proproliferative genes (1, 17, 35, 36). receptors may contribute to resistance to radiotherapy or This modulation of the opposing effects of NR4A1 on cell chemotherapy (e.g., bleomycin; refs. 18, 20). Manipulation survival by CDIMS has potential for selective receptor mod- of NR4A/DSB binding may be a strategy to increase tumor ulation in pancreatic cancer. Interestingly, in bladder cancer, sensitivity to chemoradiotherapy. a CDIM (DIM-C-pPhCl) can induce apoptosis and inhibit Other nuclear receptors, like b growth via activation of NR4A2 (1). (RARb), can influence the expression and function of NR4A

OF4 Clin Cancer Res; 18(12) June 15, 2012 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

NR4A Receptors in Cancer

receptors and are targetable. For example, by combining a Disclosure of Potential Conflicts of Interest histone deacetylase inhibitor with fenretinide (a synthetic No potential conflicts of interest were disclosed. retinoid), the expression, interaction, and nuclear export of NR4A1 and RARb are increased, leading to apoptosis in Authors' Contributions Conception and design: H.M. Mohan, A.C. Rogers, A.W. Baird, D.C. Winter hepatocellular cell lines in vitro, which are otherwise - Development of methodology: H.M. Mohan, D.C. Winter tively insensitive to fenretinide (38). Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H.M. Mohan Analysis and interpretation of data (e.g., statistical analysis, biosta- tistics, computational analysis): H.M. Mohan, D.C. Winter Conclusions Writing, review, and/or revision of the manuscript: H.M. Mohan, C.M. Aherne, A.C. Rogers, A.W. Baird, D.C. Winter, E.P. Murphy In summary, the study of NR4A receptors represents an Administrative, technical, or material support (i.e., reporting or orga- exciting new chapter in our understanding of the molecular nizing data, constructing databases): H.M. Mohan, A.C. Rogers changes that occur during carcinogenesis. NR4A receptors Study supervision: A.W. Baird, D.C. Winter have diverse cellular effects (15) and function as molecular sensors, which, depending on their cellular microenviron- Grant Support H.M. Mohan, IBD Research Foundation; C.M. Aherne, Crohns and Colitis ment, may promote or inhibit cell death. Targeting expres- Foundation of America (CCFA) Fellowship award #2865; A.C. Rogers, Irish sion levels, activity, and nuclear export of NR4A receptors, Research Council for Science, Engineering and Technology (IRCSET); and in a tissue-dependent manner, to manipulate their role in E.P. Murphy, Science Foundation Ireland (SFI). cell survival, has potential in the development of novel Received December 20, 2011; revised March 27, 2012; accepted March 28, anticancer strategies. 2012; published OnlineFirst May 7, 2012.

References 1. Safe S, Kim K, Lee S-O. NR4A Orphan receptors and cancer. Nucl 16. Wu Q, Liu S, Ye XF, Huang ZW, Su WJ. Dual roles of Nur77 in selective Recept Signal 2011;9:e002. regulation of apoptosis and cell cycle by TPA and ATRA in gastric 2. Sekiya T, Kashiwagi I, Inoue N, Morita R, Hori S, Waldmann H, et al. The cancer cells. Carcinogenesis 2002;23:1583–92. nuclear orphan receptor Nr4a2 induces Foxp3 and regulates differ- 17. Yoon K, Lee SO, Cho SD, Kim K, Khan S, Safe S. Activation of nuclear entiation of CD4þ T cells. Nat Commun 2011;2:269. TR3 (NR4A1) by a diindolylmethane analog induces apoptosis and 3. McMorrow JP, Murphy EP. Inflammation: a role for NR4A orphan proapoptotic genes in pancreatic cancer cells and tumors. Carcino- nuclear receptors? Biochem Soc Trans 2011;39:688–93. genesis 2011;32:836–42. 4. Inamoto T, Czerniak BA, Dinney CP, Kamat AM. Cytoplasmic mis- 18. Malewicz M, Kadkhodaei B, Kee N, Volakakis N, Hellman U, Viktorsson localization of the orphan nuclear receptor Nurr1 is a prognostic factor K, et al. Essential role for DNA-PK-mediated phosphorylation of NR4A in bladder cancer. Cancer 2010;116:340–6. nuclear orphan receptors in DNA double-strand break repair. Genes 5. Zhang T, Wang P, Ren H, Fan J, Wang G. NGFI-B nuclear orphan Dev 2011;25:2031–40. receptor Nurr1 interacts with p53 and suppresses its transcriptional 19. Smith AG, Luk N, Newton RA, Roberts DW, Sturm RA, Muscat GE. activity. Mol Cancer Res 2009;7:1408–15. Melanocortin-1 receptor signaling markedly induces the expression of 6. Riggins RB, Mazzotta MM, Maniya OZ, Clarke R. Orphan nuclear the NR4A nuclear receptor subgroup in melanocytic cells. J Biol Chem receptors in breast cancer pathogenesis and therapeutic response. 2008;283:12564–70. Endocr Relat Cancer 2010;17:R213–31. 20. Zhao BX, Chen HZ, Du XD, Luo J, He JP, Wang RH, et al. Orphan 7. Kitagawa H, Ray WJ, Glantschnig H, Nantermet PV, Yu Y, Leu CT, et al. receptor TR3 enhances p53 transactivation and represses DNA dou- A regulatory circuit mediating convergence between Nurr1 transcrip- ble-strand break repair in hepatoma cells under ionizing radiation. Mol tional regulation and Wnt signaling. Mol Cell Biol 2007;27:7486–96. Endocrinol 2011;25:1337–50. 8. Nordzell M, Aarnisalo P, Benoit G, Castro DS, Perlmann T. Defining an 21. Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, et al. N-terminal activation domain of the orphan nuclear receptor Nurr1. A Nurr1/CoREST pathway in microglia and astrocytes protects dopa- Biochem Biophys Res Commun 2004;313:205–11. minergic neurons from inflammation-induced death. Cell 2009;137: 9. Rajalin AM, Aarnisalo P. Cross-talk between NR4A orphan nuclear 47–59. receptors and b-catenin signaling pathway in osteoblasts. Arch Bio- 22. You B, Jiang YY, Chen S, Yan G, Sun J. The orphan nuclear receptor chem Biophys 2011;509:44–51. Nur77 suppresses endothelial cell activation through induction of 10. Wu H, Lin Y, Li W, Sun Z, Gao W, Zhang H, et al. Regulation of Nur77 IkappaBalpha expression. Circ Res 2009;104:742–9. expression by b-catenin and its mitogenic effect in colon cancer cells. 23. Holla VR, Mann JR, Shi Q, DuBois RN. Prostaglandin E2 regulates the FASEB J 2011;25:192–205. nuclear receptor NR4A2 in colorectal cancer. J Biol Chem 2006;281: 11. Chen HZ, Liu QF, Li L, Wang WJ, Yao LM, Yang M, et al. The orphan 2676–82. receptor TR3 suppresses intestinal tumorigenesis in mice by down- 24. Pearen MA, Muscat GE. Minireview: Nuclear 4A regulating Wnt signalling. Gut 2011;61:714–24. signaling: implications for metabolic disease. Mol Endocrinol 2010;24: 12. Sun Z, Cao X, Jiang MM, Qiu Y, Zhou H, Chen L, et al. Inhibition of b- 1891–903. catenin signaling by nongenomic action of orphan nuclear receptor 25. Rius J, Martínez-Gonzalez J, Crespo J, Badimon L. NOR-1 is involved Nur77. Oncogene. 2011 Oct 10. [Epub ahead of print]. in VEGF-induced endothelial cell growth. Atherosclerosis 2006;184: 13. Thompson J, Winoto A. During negative selection, Nur77 family pro- 276–82. teins translocate to mitochondria where they associate with Bcl-2 and 26. Pearen MA, Myers SA, Raichur S, Ryall JG, Lynch GS, Muscat GE. The expose its proapoptotic BH3 domain. J Exp Med 2008;205:1029–36. orphan nuclear receptor, NOR-1, a target of beta-adrenergic signaling, 14. Kolluri SK, Zhu X, Zhou X, Lin B, Chen Y, Sun K, et al. A short Nur77- regulates gene expression that controls oxidative metabolism in skel- derived peptide converts Bcl-2 from a protector to a killer. Cancer Cell etal muscle. Endocrinology 2008;149:2853–65. 2008;14:285–98. 27. Holla VR, Wu H, Shi Q, Menter DG, DuBois RN. Nuclear orphan 15. Wilson AJ, Arango D, Mariadason JM, Heerdt BG, Augenlicht LH. TR3/ receptor NR4A2 modulates fatty acid oxidation pathways in colorectal Nur77 in colon cancer cell apoptosis. Cancer Res 2003;63:5401–7. cancer. J Biol Chem 2011;286:30003–9.

www.aacrjournals.org Clin Cancer Res; 18(12) June 15, 2012 OF5

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

Mohan et al.

28. Zhao D, Desai S, Zeng H. VEGF stimulates PKD-mediated CREB- death of the human breast cancer cell line MCF-7. Mol Cell Endocrinol dependent orphan nuclear receptor Nurr1 expression: role in VEGF- 2000;162:151–6. induced angiogenesis. Int J Cancer 2011;128:2602–12. 42. Chintharlapalli S, Burghardt R, Papineni S, Ramaiah S, Yoon K, Safe S. 29. Zeng H, Qin L, Zhao D, Tan X, Manseau EJ, Van Hoang M, et al. Orphan Activation of Nur77 by selected 1,1-Bis(3¢-indolyl)-1-(p-substituted nuclear receptor TR3/Nur77 regulates VEGF-A-induced angiogenesis phenyl)methanes induces apoptosis through nuclear pathways. J Biol through its transcriptional activity. J Exp Med 2006;203:719–29. Chem 2005;280:24903–14. 30. Ohkura N, Nagamura Y, Tsukada T. Differential transactivation by 43. Ye X, Wu Q, Liu S, Lin X, Zhang B, Wu J, et al. Distinct role and orphan nuclear receptor NOR1 and its fusion gene product EWS/ functional mode of TR3 and RARalpha in mediating ATRA-induced NOR1: possible involvement of poly(ADP-ribose) polymerase I, PARP- signalling pathway in breast and gastric cancer cells. Int J Biochem Cell 1. J Cell Biochem 2008;105:785–800. Biol 2004;36:98–113. 31. Ramirez-Herrick AM, Mullican SE, Sheehan AM, Conneely OM. 44. Rosengren Pielberg G, Golovko A, Sundstrom€ E, Curik I, Lennartsson Reduced NR4A gene dosage leads to mixed myelodysplastic/myelo- J, Seltenhammer MH, et al. A cis-acting regulatory mutation causes proliferative neoplasms in mice. Blood 2011;117:2681–90. premature hair graying and susceptibility to melanoma in the horse. 32. Chang LF, Lin PC, Ho LI, Liu PY, Wu WC, Chiang IP, et al. Over- Nat Genet 2008;40:1004–9. expression of the orphan receptor Nur77 and its translocation induced 45. Ke N, Claassen G, Yu DH, Albers A, Fan W, Tan P, et al. Nuclear by PCH4 may inhibit malignant glioma cell growth and induce cell hormone receptor NR4A2 is involved in cell transformation and apo- apoptosis. J Surg Oncol 2011;103:442–50. ptosis. Cancer Res 2004;64:8208–12. 33. Liu PY, Sheu JJ, Lin PC, Lin CT, Liu YJ, Ho LI, et al. Expression of Nur77 46. Cho SD, Lee SO, Chintharlapalli S, Abdelrahim M, Khan S, Yoon K, induced by an n-butylidenephthalide derivative promotes apoptosis et al. Activation of nerve growth factor-induced B alpha by meth- and inhibits cell growth in oral squamous cell carcinoma. Invest New ylene-substituted diindolylmethanes in bladder cancer cells Drugs 2012;30:79–89. induces apoptosis and inhibits tumor growth. Mol Pharmacol 2010; 34. Zhan Y, Du X, Chen H, Liu J, Zhao B, Huang D, et al. Cytosporone B is 77:396–404. an agonist for nuclear orphan receptor Nur77. Nat Chem Biol 47. Chen YL, Jian MH, Lin CC, Kang JC, Chen SP, Lin PC, et al. The 2008;4:548–56. induction of orphan nuclear receptor Nur77 expression by n-butyle- 35. Cho SD, Lei P, Abdelrahim M, Yoon K, Liu S, Guo J, et al. 1,1-bis(3¢- nephthalide as pharmaceuticals on hepatocellular carcinoma cell indolyl)-1-(p-methoxyphenyl)methane activates Nur77-independent therapy. Mol Pharmacol 2008;74:1046–58. proapoptotic responses in colon cancer cells. Mol Carcinog 2008; 48. Camacho CP, Latini FR, Oler G, Hojaij FC, Maciel RM, Riggins GJ, 47:252–63. et al. Down-regulation of NR4A1 in follicular thyroid carcinomas is 36. Lee S, Abdelrahim M, Yoon K, Chintarlapalli S, Papineni S, Kim K, restored following lithium treatment. Clin Endocrinol (Oxf) 2009;70: et al. Inactivation of the orphan nuclear receptor TR3/Nur77 inhibits 475–83. pancreatic cancer cell and tumor growth. Cancer Res 2010;70: 49. Kolluri SK, Bruey-Sedano N, Cao X, Lin B, Lin F, Han YH, et al. 6824–36. Mitogenic effect of orphan receptor TR3 and its regulation by MEKK1 37. Shigeishi H, Higashikawa K, Hatano H, Okui G, Tanaka F, Tran TT, et al. in lung cancer cells. Mol Cell Biol 2003;23:8651–67.

PGE2 targets squamous cell carcinoma cell with the activated epider- 50. Ueda Y, Bandoh S, Fujita J, Sato M, Yamaji Y, Takahara J. Expression mal growth factor receptor family for survival against 5-fluorouracil of nerve growth factor-induced clone B subfamily and pro-opiomela- through NR4A2 induction. Cancer Lett 2011;307:227–36. nocortin gene in lung cancer cell lines. Am J Respir Cell Mol Biol 38. Yang H, Zhan Q, Wan YJ. Enrichment of Nur77 mediated by retinoic 1999;20:1319–25. acid receptor b leads to apoptosis of human hepatocellular carcinoma 51. Lee SO, Andey T, Jin UH, Kim K, Sachdeva M, Safe S. The nuclear cells induced by fenretinide and histone deacetylase inhibitors. Hepa- receptor TR3 regulates mTORC1 signaling in lung cancer cells expres- tology 2011;53:865–74. sing wild-type p53. Oncogene. 2011 Nov 14. [Epub ahead of print]. 39. Cho SD, Yoon K, Chintharlapalli S, Abdelrahim M, Lei P, Hamilton S, 52. Jeong JH, Park JS, Moon B, Kim MC, Kim JK, Lee S, et al. Orphan et al. Nur77 agonists induce proapoptotic genes and responses in nuclear receptor Nur77 translocates to mitochondria in the early phase colon cancer cells through nuclear receptor-dependent and nuclear of apoptosis induced by synthetic chenodeoxycholic acid derivatives receptor-independent pathways. Cancer Res 2007;67:674–83. in human stomach cancer cell line SNU-1. Ann N Y Acad Sci 40. Alexopoulou AN, Leao M, Caballero OL, Da Silva L, Reid L, Lakhani SR, 2003;1010:171–7. et al. Dissecting the transcriptional networks underlying breast cancer: 53. Sibayama-Imazu T, Fujisawa Y, Masuda Y, Aiuchi T, Nakajo S, Itabe H, NR4A1 reduces the migration of normal and breast cancer cell lines. et al. Induction of apoptosis in PA-1 ovarian cancer cells by vitamin K2 Breast Cancer Res 2010;12:R51. is associated with an increase in the level of TR3/Nur77 and its 41. Ohkubo T, Ohkura N, Maruyama K, Sasaki K, Nagasaki K, Hanzawa H, accumulation in mitochondria and nuclei. J Cancer Res Clin Oncol et al. Early induction of the orphan nuclear receptor NOR-1 during cell 2008;134:803–12.

OF6 Clin Cancer Res; 18(12) June 15, 2012 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst May 7, 2012; DOI: 10.1158/1078-0432.CCR-11-2953

Molecular Pathways: The Role of NR4A Orphan Nuclear Receptors in Cancer

Helen M. Mohan, Carol M. Aherne, Ailin C. Rogers, et al.

Clin Cancer Res Published OnlineFirst May 7, 2012.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-11-2953

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2012/06/01/1078-0432.CCR-11-2953. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2012 American Association for Cancer Research.