Elsevier

Biochemical Pharmacology

Volume 83, Issue 10, 15 May 2012, Pages 1445-1455
Biochemical Pharmacology

Structure-dependent activation of NR4A2 (Nurr1) by 1,1-bis(3′-indolyl)-1-(aromatic)methane analogs in pancreatic cancer cells

https://doi.org/10.1016/j.bcp.2012.02.021Get rights and content

Abstract

NR4A2 (Nurr1) is an orphan nuclear receptor with no known endogenous ligands and is highly expressed in many cancer cell lines including Panc1 and Panc28 pancreatic cancer cells. Structure-dependent activation of NR4A2 by a series of 1,1-bis(3′-indolyl)-1-(aromatic)methane (C-DIM) analogs was determined in pancreatic cancer cells transfected with yeast GAL4-Nurr1 chimeras and a UASx5-luc reporter gene or constructs containing response elements that bind NR4A2. Among 23 different structural analogs, phenyl groups containing p-substituted trifluoromethyl, t-butyl, cyano, bromo, iodo and trifluoromethoxy groups were the most active compounds in transactivation assay. The p-bromophenyl analog (DIM-C-pPhBr) was used as a model for structure–activity studies among a series of ortho-, meta- and para-bromophenyl isomers and the corresponding indole 2- and N-methyl analogs. Results show that NR4A2 activation was maximal with the p-bromophenyl analog and methylation of the indole NH group abrogated activity. Moreover, using GAL4-Nurr1 (full length) or GAL-Nurr1-A/B and GAL4-Nurr1-(C-F) chimeras expressing N- and C-terminal domains of Nurr1, respectively, DIM-C-pPhBr activated all three constructs and these responses were differentially affected by kinase inhibitors. DIM-C-pPhBr also modulated expression of several Nurr1-regulated genes in pancreatic cancer cells including vasoactive intestinal peptide (VIP), and the immunohistochemical and western blot analyses indicated that DIM-C-pPhBr activates nuclear NR4A2.

Introduction

The nuclear receptor (NR) superfamily of transcription factors are characterized by their structural homology which includes N- and C-terminal domains (A/B and E/F, respectively), a DNA binding domain (C), and an adjacent hinge region (D) [1], [2], [3]. Both N- and C-terminal regions may contain activation functions (AFs), and the ligand binding domain (LBD) resides in the C-terminus of NRs [1], [2], [3], [4]. The 48 members of the NR superfamily can be subdivided into three broad categories, namely, the endocrine nuclear receptors, adopted orphan receptors, and orphan receptors such as NR4A for which cognate ligands have not yet been identified [1], [2], [3], [4]. The NR4A orphan receptor subfamily includes NR4A1 (Nur77, NGFI-B, TR3), NR4A2 (Nurr1, NOT), and NR4A3 (Nor-1, MINOR) [5], [6], [7], [8], and it has been suggested that the failure to identify an endogenous ligand may be due to the lack of a typical NR ligand binding pocket in the LBD domain of NR4A receptors [9].

NR4A receptors are immediate-early genes that are induced by diverse stimuli in multiple tissues, and there is increasing evidence that these receptors play important roles in maintaining tissue homeostasis and in pathophysiological processes including cancer [5], [6], [7], [8]. NR4A receptors have specific functions in the brain, T-cells/thymocytes, adipose tissue, steroidgenesis, muscle, blood vessels, macrophages, and cardiovascular system; however, the role of NR4A receptors in carcinogenesis is less well understood [7], [8]. Knockdown of NR4A1 by RNA interference in cancer cell lines induced apoptosis or inhibited growth in multiple cell lines (reviewed in [8]) and NR4A2 knockdown also induced apoptosis and decreased metastasis in cancer cells lines [10], [11], [12].

The effects and mechanisms of action of drug-induced activation or deactivation of NR4A receptors are complex and dependent on both cell context and structure. Studies with phorbol esters, retinoids and other apoptosis-inducing agents have unraveled a novel NR4A1-dependent proapoptotic pathway that involves nuclear export of the receptor which in some cell lines form a mitochondrial proapoptotic bcl-2–NR4A1 complex [13], [14]. Studies in this laboratory have demonstrated that among 1,1-bis(3′-indolyl)-1-(p-substituted phenyl)methanes (C-DIMs), the p-methoxy (DIM-C-pPhOCH3) and p-hydroxy (DIM-C-pPhOH) derivatives induce nuclear NR4A1-dependent apoptosis and growth inhibition of colon, pancreatic and bladder cancer cells through activation or deactivation of the receptor [15], [16], [17], [18]. Cytosporone B and related compounds bind directly the receptor and appear to activate nuclear NR4A1-mediated transcription; these receptor agonists also induce nuclear NR4A1 export [19], [20].

6-Mercaptopurine (6-MP) activates NR4A2 in CV1 and HEK293 cells through the N-terminal A/B domain and this pathway involves metabolic activation of 6-MP [21]. Several benzimidazoles also induce NR4A2-dependent transactivation, and a p-chloro-substituted C-DIM analog (DIM-C-pPhCI) also activates Nurr1 in bladder cancer cells [22], [23]. In this study, we have investigated the structure-dependent activation of NR4A2 in pancreatic cancer cells by twenty-three C-DIM analogs containing various p-substituted phenyl and heteroaromatic substituents and their activity was compared to 6-MP. Among the most active compounds were the p-trifluoromethyl (DIM-C-pPhCF3), p-bromo (DIM-C-pPhBr), p-t-butyl (DIM-C-pPhtBu), p-cyano (DIM-C-pPhCN), p-iodo (DIM-C-pPhI), and p-trifluoromethoxy (DIM-C-phOCF3) analogs. Using one or more NR4A2-active C-DIMs as models, these compounds also induced transactivation in cells transfected with constructs containing three copies of an NGFI-B response element (NBRE3-luc) and three copies of a Nur response element (NuREx3-luc). Nurr1-active C-DIMs also activated a wild-type GAL4-Nurr1 variant and GAL4-Nurr1-(A/B) and GAL4-Nurr1-(C-F) chimeras containing N- and C-terminal regions of NR4A2, respectively, and the prototypical model NR4A2 activator DIM-C-pPhBr induced expression of several NR4A2-dependent genes in Panc1 and Panc28 that were confirmed by RNA interference. These studies demonstrate that the C-DIM structure is an excellent scaffold for developing NR4A2-active compounds.

Section snippets

Cell lines and cell culture

Panc1 and Panc28 pancreatic cancer cell lines were obtained from the American Type Culture Collection (Manassas, VA) and maintained in Dulbecco's modified Eagle's medium (DMEM) nutrient mixture with Ham's F-12 (DMEM/Ham's F-12; Sigma–Aldrich, St Louis, MO) supplemented with 5% fetal bovine serum (FBS), 0.22% sodium bicarbonate and 10 mL/L 100× antibiotic antimycotic solution (Invitrogen, Carlsbad, CA). Cells were maintained at 37 °C in the presence of 5% CO2 and the solvent [dimethyl sulfoxide

Results

Panc1 and Panc28 pancreatic cancer cell lines were used in these studies for screening a series of C-DIM compounds as activators of NR4A2. Transfection of Panc28 cells with a UASx5-luc construct containing 5 tandem GAL4 response elements gave relatively low basal luciferase activity compared to Panc1 cells. For the C-DIM screening assay, Panc28 cells were transfected with UASx5-luc and the GAL4-Nurr1 chimera (containing the yeast GAL4 DNA binding domain fused to wild type NR4A2) and treated

Discussion

Although endogenous ligands for the NR4A orphan receptors have not been identified, there is increasing evidence that several agents can modulate nuclear NR4A-dependent transactivation [8], [9], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20]. Cytosporone B and related analogs have been extensively investigated as NR4A1 (TR3) agonists and they activate nuclear NR4A1 and induce nuclear export of this receptor which acts directly on mitochondria to induced apoptosis [14], [20]. The

Conflicts of interest

There are no conflicts of interest to disclose.

Acknowledgements

This research was supported by the National Institutes of Health (R01CA124998) and Texas AgriLife Research.

References (35)

  • M.A. Maxwell et al.

    The NR4A subgroup: immediate early response genes with pleiotropic physiological roles

    Nucl Recept Signal

    (2006)
  • M.A. Pearen et al.

    Minireview: nuclear hormone receptor 4A signaling—implications for metabolic disease

    Mol Endocrinol

    (2010)
  • S.O. Lee et al.

    Targeting NR4A1 (TR3) in cancer cells and tumors

    Expert Opin Ther Targets

    (2011)
  • Z. Wang et al.

    Structure and function of Nurr1 identifies a class of ligand-independent nuclear receptors

    Nature

    (2003)
  • N. Ke et al.

    Nuclear hormone receptor NR4A2 is involved in cell transformation and apoptosis

    Cancer Res

    (2004)
  • T. Inamoto et al.

    Cytoplasmic mislocalization of the orphan nuclear receptor Nurr1 is a prognostic factor in bladder cancer

    Cancer

    (2010)
  • Q.X. Li et al.

    NR4A1,2,3—an orphan nuclear hormone receptor family involved in cell apoptosis and carcinogenesis

    Histol Histopathol

    (2006)
  • Cited by (65)

    • Pharmacological activation of Nr4a rescues age-associated memory decline

      2020, Neurobiology of Aging
      Citation Excerpt :

      Even though we do not have direct evidence that the memory enhancement by C-DIM drugs are due directly to activation of Nr4a transcription factors in vivo, previous studies show that C-DIM compounds activate Nr4a-mediated transcription (Hammond et al., 2018; Hedrick et al., 2019; Li et al., 2012). Notably, C-DIM12 fails to activate Nr4a downstream gene expression following Nr4a2 knockdown in cultured cell lines (Hammond et al., 2015; Inamoto et al., 2008; Li et al., 2012). In addition, in vitro reporter assays and computational modeling further support the idea that C-DIM12 directly activates Nr4a (Hammond et al., 2018; Inamoto et al., 2008).

    • A novel diindolylmethane analog, 1,1-bis(3’-indolyl)-1-(p-chlorophenyl) methane, inhibits the tumor necrosis factor-induced inflammatory response in primary murine synovial fibroblasts through a Nurr1-dependent mechanism

      2018, Molecular Immunology
      Citation Excerpt :

      In this study, we investigated one of a novel series of paraphenyl substituted dinndolylmethane compounds (C-DIMs) that has been previously demonstrated to be a transcriptional modulator of NR4A family nuclear receptors (De Miranda et al., 2015; Lee et al., 2014; Yoon et al., 2011; Inamoto et al., 2008). C-DIM12 activates Nurr1 in dopaminergic neurons (Hammond et al., 2015), microglial cells (De Miranda et al.,2015), pancreatic cells (Li et al., 2012) and in urothelial carcinoma cells (Inamoto et al., 2008). However, it is unknown whether C-DIM12 directly inhibits inflammatory gene transcription in primary synovial fibroblasts.

    View all citing articles on Scopus
    View full text