A truncated version of retinoid X receptor-α, tRXR-α, promotes cancer cell survival by activating the phosphoinositide 3-kinase (PI3K)/AKT pathway. However, targeting the tRXR-α–mediated survival pathway for cancer treatment remains to be explored. We report here our identification of a new natural product molecule, CF31, a xanthone isolated from Cratoxylum formosum ssp. pruniflorum, and the biologic evaluation of its regulation of the tRXR-α–mediated PI3K/AKT pathway. CF31 binds RXR-α and its binding results in inhibition of RXR-α transactivation. Through RXR-α mutational analysis and computational studies, we show that Arg316 of RXR-α, known to form salt bridges with certain RXR-α ligands, such as 9-cis-retinoic acid (9-cis-RA), is not required for the antagonist effect of CF31, showing a distinct binding mode. Evaluation of several CF31 analogs suggests that the antagonist effect is mainly attributed to an interference with Leu451 of helix H12 in RXR-α. CF31 is a potent inhibitor of AKT activation in various cancer cell lines. When combined with TNF-α, it suppresses TNF-α activation of AKT by inhibiting TNF-α–induced tRXR-α interaction with the p85α regulatory subunit of PI3K. CF31 inhibition of TNF-α activation of AKT also results in TNF-α–dependent activation of caspase-8 and apoptosis. Together, our results show that CF31 is an effective converter of TNF-α signaling from survival to death by targeting tRXR-α in a unique mode and suggest that identification of a natural product that targets an RXR-mediated cell survival pathway that regulates PI3K/AKT may offer a new therapeutic strategy to kill cancer cells. Cancer Res; 73(1); 307–18. ©2012 AACR.

Retinoid X receptor (RXR)-α, a unique member of the nuclear receptor superfamily, regulates diverse biologic processes, including growth, differentiation, apoptosis, and immune response, and ligands for RXR-α show promise as therapeutic agents for many diseases, such as cancer (1–4). Alterations in the expression and function of RXR-α are implicated in the development of a number of diseases and cancer. Targeted disruption of RXR-α gene leads to prostatic preneoplastic lesions (5) and skin abnormalities (6). Dysregulation of RXR-α function by phosphorylation is associated with the development of human liver cancer (7) and colon cancer (8). Levels of RXR-α protein are often reduced in cancer cells and tumor tissues (9–14), suggesting that diminished RXR-α expression is associated with the development of certain malignancies. Numerous studies have shown that proteolytic cleavage of RXR-α protein is primarily responsible for its diminished levels in tumor cells and represents an important mechanism controlling the function and activities of RXR-α (13–18). Recent studies have shown that RXR-α binding to PML/RAR-α is essential for the development of acute promeylocytic leukemia (19, 20), further showing the oncogenic potential of this protein when it acts inappropriately.

Although significant progress has been made, how RXR-α regulates cancer cell growth and how its ligands suppress tumorigenesis is still poorly understood. Like other nuclear receptors, RXR-α interacts with DNA to regulate the transcription of target genes in a ligand-dependent manner (1–4). Aside from its role in DNA binding and transactivation, accumulating evidence indicates that RXR-α also has extranuclear nongenomic actions (15, 21–24). RXR-α resides in the cytoplasm at certain stages during development (25, 26). It migrates from the nucleus to the cytoplasm in response to differentiation (23), apoptosis (21), and inflammation (22, 24). Cytoplasmic localization of RXR-α facilitates nuclear export of its heterodimerization partner Nur77, leading to Bcl-2 conversion and apoptosis (21, 27, 28).

We showed recently that proteolytic cleavage of RXR-α results in production of truncated RXR-α protein, tRXR-α, which acquires new function that is different from the full-length RXR-α (29). Unlike the full-length RXR-α that resides in the nucleus, tRXR-α is cytoplasmic and interacts with the p85α subunit of phosphoinositide 3-kinase (PI3K) in a TNFα–dependent manner, leading to activation of the PI3K/AKT pathway, a major survival pathway important for uncontrolled growth of tumor and as drug resistance (29). Activation of the PI3K/AKT pathway by tRXR-α contributes significantly to anchorage-independent growth of cancer cells in vitro and tumor growth in animals (29), offering an opportunity to suppress cancer cell growth by targeting the tRXR-α–mediated PI3K/AKT pathway.

Epidemiologic studies have shown that dietary phytochemicals provide beneficial effects for cancer prevention, and among them, xanthones are of great interest as cancer chemopreventive agents because of their potent antioxidative and anticancer activity (30–33). We previously purified a series of xanthones from a medicinal plant, Cratoxylum formosum ssp. pruniflorum, which belongs to the Clusiaceae family and is widely distributed in several Southeast Asian countries (34, 35). Here, we report our identification of one of the xanthones, CF31, which suppresses tRXR-α–dependent AKT activation through its unique RXR-α binding. CF31, when combined with TNF-α inhibits TNF-α–induced tRXR-α–p85α interaction and AKT activation. Moreover, the CF31 and TNF-α combination results in synergistic induction of TNF-α–dependent caspase-8 activation and apoptosis in cancer cells. Thus, CF31 represents a new modulator of the tRXR-α survival pathway and a potent converter of TNF-α signaling from survival to death.

Isolation of natural products

Compounds CF31, also called cochinchinone B or 1,3,6,7-tetrahydroxy-2-(3-methyl-2-butenyl)-5-(3,7-dimethyl-2,6-octadienyl)xanthone (36), CF13 (1,3,5-trihydroxy-4-geranylxanthone), and CF26 (pruniflorone Q) were isolated from the stems of Cratoxylum formosum ssp. pruniflorum as described (35).

Plasmids

The pGAL4-RXR-α-LBD plasmid was obtained by inserting RXR-α ligand-binding domain (LBD) sequence (amino acids 198–462) in-frame with the Gal4 DBD coding sequence in the pBIND vector (Promega).

Antibodies and regents

Antibodies for phospho-AKT (Ser473, D9E) and cleaved caspase-8 (p43/p41; Cell Signaling); Bax (6A7; Sigma-Aldrich); p85α (Millipore); AKT1 (C-20), actin, c-Myc (9E10), RXR-α (ΔN197), RXR-α (D20), and PARP (H-250; Santa Cruz Biotechnology) were used. Caspase-3 and -8 activity assay kits were from Biovision.

Stable transfections

GFP-RXR-α/Δ80 was stably transfected into SW480, HepG2, andMCF-7 cells to obtain SW480/RXR-α/Δ80, HepG2/RXR-α/Δ80, and MCF-7/RXR-α/Δ80, respectively.

Ligand-binding assay

RXR-α LBD was incubated with [3H]-9-cis-RA in the presence or absence of unlabeled 9-cis-retinoic acid (9-cis-RA) or CF31. Bound [3H]-9-cis-RA in RXR-α. LBD protein was determined in a scintillation counter (29).

Transient transfection and reporter assays

CV-1 green monkey kidney cells were grown in Dulbecco's Modified Eagle's Medium supplemented with 10% FBS. For chloramphenicol acetyltransferase (CAT) reporter assays, cells were seeded at 5 × 104 cells per well in 24-well plates and transfected with 50 ng TREpal-tk-CAT reporter plasmid (37), 20 ng β-galactosidase expression vector (pCH 110; Amersham), 20 ng RXR-α expression vectors using Lipofectamine 2000 (Invitrogen). Cells were then treated with CF31 at different dose for 20 hours. CAT activity was normalized with β-galactosidase activity. For luciferase reporter assay, 24 hours after transfected with pGL5 luciferase reporter vector (40 ng/well) and pGAL4-RXR-α-LBD expression vector (40 ng/well), cells were incubated with varied concentrations of compounds for 12 hours. Luciferase activities were measured using the Dual-Luciferase Assay System Kit (Promega).

Molecular docking and molecular dynamics simulations

AutoDock version 4.0 was used for the docking screening. The Lamarckian genetic algorithm was selected for ligand conformational searching. Docking parameters were as follows: grid box of 40 Å × 40 Å × 40 Å, xyz-coordinates of gridcenter: 49.192 63.991 −7.523, population size of 150, random starting position and conformation, translation step ranges of 2 Å, rotation step ranges of 50°, elitism of 1, mutation rate of 0.02, cross-over rate of 0.8, local search rate of 0.06, and 2.5 million energy evaluations. Molecular dynamics simulations were adopted to sample possible configurations of each RXR-α LBD complex using the AMBER (version 7.0) program (38). To set up each molecular dynamics simulation, the electrostatic potentials of the ligand was computed by using the Gaussian 98 package at the HF/6-31G* level. Atom-centered partial charges were derived by using the Restrained Electrostatic potential method implemented in the AMBER package.

Immunofluorescence microscopy

HepG2 cells mounted on glass slides were permeabilized with PBS containing 0.1% Triton X-100 and 0.1 mol/L glycine for 15 minutes, and blocked with 1% bovine serum albumin in PBS for 30 minutes at room temperature. Cells were then incubated with primary antibody [anti-Bax (6A7; 1:500)] at 37°C for 1 hour and detected by anti-rabbit immunoglobulin G (IgG) conjugated with Cy3 (1:400) at room temperature for 30 minutes. Cells were costained with 4′6′-diamidino-2-phenylindole (DAPI) to visualize nuclei. The images were taken using LSM-510 confocal laser scanning microscope system (Carl Zeiss).

Western blotting

Equal amounts of the lysates were electrophoresed on 8% SDS-PAGE gel and transferred onto polyvinylidene difluoride membranes. The membranes were blocked with 5% nonfat milk in Tris-Buffered Saline and Tween 20 [50 mmol/L Tris–HCl (pH 7.4), 150 mmol/L NaCl, and 0.1% Tween 20] for 1 hour, incubated with various primary antibodies for 2 hours and detected with either anti-rabbit (1:5,000) or anti-mouse (1:5,000) secondary antibodies for 1 hour, all of which were undertaken under room temperature. The final immunoreactive products were detected by using enhanced chemiluminescence (ECL) system.

Caspase activity assay

Caspase activities were determined by a colorimetric assay based on the ability of caspase-3 and -8 to change acetyl–Asp–Glu–Val–Asp p-nitroanilide (Ac-DEVD-pNA) and acetyl–Ile–Glu–Thr–Asp p-nitroanilide Ac-IETD-pNA) into a yellow formazan product [p-nitroaniline (pNA)], respectively. After treatment with CF31 and/or TNF-α, lysates were prepared and assayed according to the manufacturer's protocol (Biovision Research Products).

RXR-α siRNA

RXR-α siRNA siGENOME SMARTpool (M-003443-02) and siRNA Nonspecific Control IX (D-001206-09-05) were from DHARMACON, and they were transfected into cells by RNAiMAX reagent (Invitrogen).

Coimmunoprecipitation and immunoblotting assays

Cells extracts were cleared by incubation with the Protein A/G plus Agarose beads and then incubated with appropriate antibody and 30 μL of Protein A/G plus Agarose beads overnight at 4°C. Immunoreactive products were detected by chemiluminescence with an ECL system (Amersham).

Colony formation assay

HeLa cells were seeded in 6-well plate (500 cells/well) for 5 days, treated with CF31 in 0.5% serum medium for 3 days, and fixed with 4% paraformaldehyde. Colonies were stained with 0.1% crystal violet.

Statistical analyses

Data were analyzed using an analysis of variance or Student test and were presented as the mean ± SEM of triplicate.

CF31 binds to RXR-α

To identify new agents for regulating the tRXRα survival pathway, we screened a natural product library prepared from Chinese Herbal Medicine for compounds capable of regulating RXR-α transcriptional function. For this purpose, transient transfection assays using a CAT reporter containing TREpal capable of binding to RXR-α homodimer (37) were used. Compounds were evaluated for their effect on TREpal activity in the absence or presence of 9-cis-RA, the natural RXR ligand known to induce the formation of RXR-α homodimer and its activation of TREpal (37). Among compounds that were identified to modulate RXR-α activity, CF31 (Fig. 1A), which was prepared from Cratoxylum formosum ssp. Pruniflorum (35), potently inhibited 9-cis-RA–induced TREpal-reporter activity in a dose-dependent manner (Fig. 1B). CF31 at 10 μmol/L showed a similar inhibitory effect when compared with BI1003 (1 μmol/L), a known RXR-α antagonist (39). To further study the inhibitory effect of CF31, we cloned the LBD of RXR-α as a Gal4 fusion, and used the resulting Gal4-RXR-α/LBD chimera and Gal4 reporter system to evaluate the inhibitory effect of CF31. Gal4-RXR-α/LBD strongly activated the Gal4 reporter in the presence of 9-cis-RA, which was inhibited by 1 μmol/L UVI3003, another RXR-α antagonist (40; Fig. 1C). Similar to UVI3003, treatment of cells with CF31 resulted in inhibition of 9-cis-RA–induced reporter activity in a CF31 dose-dependent manner. Because UVI3003 was derived from a potent and selective RXR-α agonist CD3254 (40), the effect of CF31 on CD3254-induced RXR-α activity was also evaluated. CD3254 strongly induced Gal4-RXR-α/LBD activity, which was comparable with that induced by 9-cis-RA. Interestingly, CD3254-induced reporter activity was similarly inhibited by UVI3003 and CF31 (Fig. 1C). We next determined whether CF31 could bind to RXR-α. Thus, a ligand competition assay with [3H]9-cis-RA binding to RXR-α LBD was used. While unlabeled 9-cis-RA was able to efficiently displace [3H]9-cis-RA from binding to RXR-α LBD with an IC50 of 7.6 nmol/L, CF31 displaced [3H]9-cis-RA with an IC50 of 9.6 μmol/L (Fig. 1D). Together, our data show that CF31 acts as a RXR-α antagonist by binding to the RXR-α LBD.

Figure 1.

CF31 binds to RXR-α in a unique mode. A, structure of CF31. B, inhibitory effect of CF31 on RXR-α transcriptional activity. CV-1 cells transfected with TREpal-tk-CAT and RXR-α expression vector were treated with the indicated concentrations of CF31 in the presence of 10−7 mol/L 9-cis-RA. Reporter activities were measured and normalized. For comparison, the effect of BI-1003 (1 μmol/L) was shown. One of 5 similar experiments is shown. Bars represent means ± SEM. C, inhibition of Gal4-RXR-α-LBD activity by CF31. CV-1 cells transfected with pGL5 luciferase reporter vector and pGAL4-RXR-α-LBD expression vector (40 ng/well) were incubated with or without 9-cis-RA (10−7 mol/L) or CD3254 (10−7 mol/L) in the presence or absence of the indicated concentrations of CF31 or UVI3003 for another 12 hours. Luciferase activities were measured using the Dual-Luciferase Assay System Kit. One of 3 similar experiments is shown. Bars represent means ± SEM. D, CF31 binding to RXR-α in vitro. RXR-α LBD protein was incubated with [3H] 9-cis-RA in the presence or absence of CF31 or unlabeled 9-cis-RA. Bound [3H] 9-cis-RA was quantitated by liquid scintillation counting.

Figure 1.

CF31 binds to RXR-α in a unique mode. A, structure of CF31. B, inhibitory effect of CF31 on RXR-α transcriptional activity. CV-1 cells transfected with TREpal-tk-CAT and RXR-α expression vector were treated with the indicated concentrations of CF31 in the presence of 10−7 mol/L 9-cis-RA. Reporter activities were measured and normalized. For comparison, the effect of BI-1003 (1 μmol/L) was shown. One of 5 similar experiments is shown. Bars represent means ± SEM. C, inhibition of Gal4-RXR-α-LBD activity by CF31. CV-1 cells transfected with pGL5 luciferase reporter vector and pGAL4-RXR-α-LBD expression vector (40 ng/well) were incubated with or without 9-cis-RA (10−7 mol/L) or CD3254 (10−7 mol/L) in the presence or absence of the indicated concentrations of CF31 or UVI3003 for another 12 hours. Luciferase activities were measured using the Dual-Luciferase Assay System Kit. One of 3 similar experiments is shown. Bars represent means ± SEM. D, CF31 binding to RXR-α in vitro. RXR-α LBD protein was incubated with [3H] 9-cis-RA in the presence or absence of CF31 or unlabeled 9-cis-RA. Bound [3H] 9-cis-RA was quantitated by liquid scintillation counting.

Close modal

Arg316 is not required for the antagonist effect of CF31

Molecular conformation analyses showed there was a large degree of structural overlapping between CF31 and LG100754 (Fig. 2A), an antagonist of RAR-α/RXR-α heterodimer (41). This suggested that CF31 might bind to the antagonist form of the RXR-α LBD. However, CF31, unlike many natural and synthetic RXR-α ligands, lacks a carboxylate moiety known to form salt bridges with Arg316 in the L-shaped RXRα ligand-binding pocket (LBP; refs. 41–43). To understand how CF31 binds RXR-α, a docking study using the antagonist structure of the RXR-α LBD in complex with LG100754 (41) was conducted. CF31 docked well with a good score to the RXRα antagonist conformation. Superposing the docked configuration of CF31 and the crystal structure of LG100754 showed that their scaffold orientations were highly conserved (Fig. 2A). An interesting difference, however, is that while the carboxylate group of LG100754 made a strong salt bridge with Arg316 in helix H5, CF31 could not form such a bridge (Fig. 2B).

Figure 2.

Analysis of CF31 RXR-α binding. A, comparison of the docked conformation of CF31 (green and red) with the crystal structure of LG100754 (yellow and red; PDB code: 3A9E). B, the interactions of LG100754 (yellow) and CF31 (green), respectively, with the RXR-α LBP. Only residues closer than 4.2 Å to the ligand are shown (blue). Salt bridge is shown as dotted green line. The secondary structure of the RXR-α-LBD from structure of PDB 3A9E was used and specific residues of RXR-α are labeled. C, role of Arg316 mutation. CV-1 cells transiently transfected with 40 ng of pGL5-Luc reporter vector and 40 ng of Gal4-RXR-α-LBD or Gal4-RXR-α-LBD/R316A were treated with or without 9-cis-RA (10−7 mol/L) or CD3254 (10−7 mol/L) in the presence or absence of the indicated concentrations of CF31 or UVI3003. One of 3 similar experiments is shown. Bars represent means ± SEM. D, the interactions of 9-cis-RA (yellow, PDB 1FBY)) and CD3254 (black, PDB 3FUG), respectively, with Arg316 of RXR-α LBP. Comparison of ligand-residue interaction spectrum for 9-cis-RA and CF31. E, binding free energies were computed using the MM-PB/SA method, including each residue in the RXR-α LBP (from amino acids 260–450) and the overall binding free energy given by the MM-PB/SA method for 9-cis-RA and CF31.

Figure 2.

Analysis of CF31 RXR-α binding. A, comparison of the docked conformation of CF31 (green and red) with the crystal structure of LG100754 (yellow and red; PDB code: 3A9E). B, the interactions of LG100754 (yellow) and CF31 (green), respectively, with the RXR-α LBP. Only residues closer than 4.2 Å to the ligand are shown (blue). Salt bridge is shown as dotted green line. The secondary structure of the RXR-α-LBD from structure of PDB 3A9E was used and specific residues of RXR-α are labeled. C, role of Arg316 mutation. CV-1 cells transiently transfected with 40 ng of pGL5-Luc reporter vector and 40 ng of Gal4-RXR-α-LBD or Gal4-RXR-α-LBD/R316A were treated with or without 9-cis-RA (10−7 mol/L) or CD3254 (10−7 mol/L) in the presence or absence of the indicated concentrations of CF31 or UVI3003. One of 3 similar experiments is shown. Bars represent means ± SEM. D, the interactions of 9-cis-RA (yellow, PDB 1FBY)) and CD3254 (black, PDB 3FUG), respectively, with Arg316 of RXR-α LBP. Comparison of ligand-residue interaction spectrum for 9-cis-RA and CF31. E, binding free energies were computed using the MM-PB/SA method, including each residue in the RXR-α LBP (from amino acids 260–450) and the overall binding free energy given by the MM-PB/SA method for 9-cis-RA and CF31.

Close modal

To study the requirement of Arg316 in CF31 binding, Arg316 was replaced with Glu, and the resulting mutant RXR-α/R316E was evaluated for its activation by 9-cis-RA and CD3254. As shown in Fig. 2C, mutation of Arg316 completely abolished the effect of 9-cis-RA on activating the Gal4 reporter. In contrast, CD3254 could still activate the RXR-α/R316E mutant. Thus, while Arg316 is capable of establishing an ionic interaction with the carboxylate group of 9-cis-RA (43), it fails to form salt bridges with CD3254 (40). Consistently, our computational analysis showed that 9-cis-RA formed 2 shorter hydrogen bonds (2.098 Å and 2.423 Å) with Arg316 than those formed with CD3254 (2.901 Å and 2.553 Å; Fig. 2D). Therefore, Arg316 plays a more important role in the binding of 9-cis-RA than that of CD3254. We then studied whether CF31 could inhibit CD3254-induced RXRα/R316E activity. When RXR-α/R316E was cotransfected with the Gal4 reporter, CD3254-induced reporter activity was potently inhibited by CF31 in a dose-dependent manner (Fig. 2C), showing that Arg316 was not required for CF31 binding.

To identify which amino acid residues in the RXR-α LBP were key to CF31 binding, we used the Molecular Mechanics Poisson–Boltzmann Surface Area (MM-PB/SA) method (44) to evaluate binding free energy of CF31 in complex with the RXRα LBP. For comparison, 9-cis-RA was also studied. Our examination of the interaction spectrum of CF31 with amino acid residues in the RXR-α LBP with that of 9-cis-RA revealed that Arg316 was indeed mainly responsible for the binding of 9-cis-RA but not CF31 (Fig. 2E; Supplementary Table S1). However, CF31 made considerable contacts with several amino acid residues in the RXR-α LBP, including Val265 and Ile268 in H3, Ile310 and Phe313 in H5, V342, I345, F346 in H7, and Leu436 in H11 (Fig. 2E). Thus, the nonpolar van der Waals interactions of CF31 with the hydrophobic residues in the RXRα LBP may serve to stabilize the binding of CF31, revealing its distinct mode of RXRα binding.

In an attempt to gain more understanding of the antagonist effect of CF31, we evaluated 2 additional xanthones isolated from Cratoxylum formosum ssp. pruniflorum, CF13 and CF26 (Fig. 3A), for their antagonist effect (Fig. 3B). While CF13 had no inhibitory effect of 9-cis-RA–induced RXR-α transactivation, the inhibitory effect of CF26 was even stronger than that of CF31 (Fig. 3B). Modeling studies suggested that a potential steric hindrance between side chain of Leu451 in H12 and substituent at position 3 of ring A of these compounds might mediate their antagonist effect (Fig. 3C). The size of the substituent correlated well with their antagonist effect, with 3-methyl-2-butenyl in CF26 being the most effective.

Figure 3.

Antagonist effect of CF31 compounds of RXR-α transactivation. A, structure of compounds CF13, CF31, and CF26. The red moiety of each compound is predicted to mediate its antagonistic activity. B, antagonist effect of CF13, CF31, and CF26. CV-1 cells transfected with TREpal-tk-CAT and RXR-α expression vector were treated with or without 10−7 mol/L 9-cis-RA in the absence or presence of 10 μmol/L CF13, CF26, or CF31. Reporter activities were measured and normalized. One of 3 similar experiments is shown. Bars represent means ± SEM. C, superposition of the docked binding modes of compounds CF13 (purple), CF31 (orange), and CF26 (black) with RXR-α and their interactions with the residue Leu451 of helix 12.

Figure 3.

Antagonist effect of CF31 compounds of RXR-α transactivation. A, structure of compounds CF13, CF31, and CF26. The red moiety of each compound is predicted to mediate its antagonistic activity. B, antagonist effect of CF13, CF31, and CF26. CV-1 cells transfected with TREpal-tk-CAT and RXR-α expression vector were treated with or without 10−7 mol/L 9-cis-RA in the absence or presence of 10 μmol/L CF13, CF26, or CF31. Reporter activities were measured and normalized. One of 3 similar experiments is shown. Bars represent means ± SEM. C, superposition of the docked binding modes of compounds CF13 (purple), CF31 (orange), and CF26 (black) with RXR-α and their interactions with the residue Leu451 of helix 12.

Close modal

CF31 inhibits AKT activation

In the first step to study the effect of CF31 on tRXRα–dependent AKT survival pathway, we determined whether CF31 could regulate AKT activation. Figure 4A shows that CF31 dose-dependently inhibited the activation of AKT in HeLa cells, revealed by its inhibition of the expression of phospho-AKT but not total AKT using Western blotting. Time-course analysis showed that the inhibition of AKT activation by CF31 was observed when cells were treated by 10 μmol/L CF31 for as short as 8 hours (Fig. 4B). The inhibition of AKT activation by CF31 also closely correlated with apoptosis induction as shown by enhanced cleavage of PARP protein (Figs. 4A and B).

Figure 4.

Inhibition of AKT activation and cells survival by CF31. A, dose-dependent effect of CF31 on AKT activation and apoptosis induction. HeLa cells were treated with CF31 (1, 10, and 20 μmol/L) for 12 hours and analyzed by immunoblotting. B, time-course analysis. HeLa cells were treated with CF31 (10 μmol/L) for 8, 16, and 24 hours and analyzed by immunoblotting. C, CF31 inhibits clonogenic survival of HeLa cells. Cells grown in 6-well plates for 5 days were treated with CF31 (50 μmol/L) or K-80003 (50 μmol/L) or sulindac (50 μmol/L) for 3 days. D, RXR-α–dependent enhancement of the apoptotic effect of camptothecine by CF31. HepG2 cells transfected with control or RXR-α siRNA were treated with CF31 (5 μmol/L) and/or camptothecine (10 μmol/L) for 9 hours and analyzed by immunoblotting using anti-PARP and D20 anti-RXR-α antibody.

Figure 4.

Inhibition of AKT activation and cells survival by CF31. A, dose-dependent effect of CF31 on AKT activation and apoptosis induction. HeLa cells were treated with CF31 (1, 10, and 20 μmol/L) for 12 hours and analyzed by immunoblotting. B, time-course analysis. HeLa cells were treated with CF31 (10 μmol/L) for 8, 16, and 24 hours and analyzed by immunoblotting. C, CF31 inhibits clonogenic survival of HeLa cells. Cells grown in 6-well plates for 5 days were treated with CF31 (50 μmol/L) or K-80003 (50 μmol/L) or sulindac (50 μmol/L) for 3 days. D, RXR-α–dependent enhancement of the apoptotic effect of camptothecine by CF31. HepG2 cells transfected with control or RXR-α siRNA were treated with CF31 (5 μmol/L) and/or camptothecine (10 μmol/L) for 9 hours and analyzed by immunoblotting using anti-PARP and D20 anti-RXR-α antibody.

Close modal

The ability of CF31 to inhibit AKT activation prompted us to examine the effect of CF31 on the survival of HeLa cells by colonogenic survival assays. Treatment of cells with CF31 for 3 days completely inhibited colony formation of HeLa cells, which was as effective as K-80003, a sulindac analog that inhibits AKT activation through its binding to tRXR-α (29). In contrast, sulindac showed little effect under the same conditions (Fig. 4C). We also determined whether CF31 inhibition of AKT activation could enhance the apoptotic response of cancer cells to chemotherapeutics. To this end, we examined the death effect of the topoisomerase-I inhibitor camptothecine (CPT), which has shown significant antitumor activity across a broad spectrum of human tumors (45) in the presence or absence of CF31 in HepG2 liver cancer cells. Immunoblotting showed that PARP cleavage induced by camptothecine was significantly enhanced when cells were cotreated with CF31, whereas CF31 alone had undetectable effect on PARP cleavage under the conditions used (Fig. 4D). When cells were transfected with RXR-α siRNA, the enhancing effect of CF31 was impaired. Thus, CF31 may sensitize cancer cells to the apoptotic effect of certain anticancer drugs through its inhibition of AKT activation.

Antagonist effect of CF31 on TNF-α activation of AKT

TNF-α, a multifunctional cytokine, can activate AKT in a tRXR-α–dependent manner in certain cancer cells (29). In HeLa cells, TNF-α treatment enhanced AKT activation, whereas CF31 and K-80003 showed comparable effect on inhibiting AKT activation. Both compounds were much more effective than sulindac when used at 10 μmol/L (Fig. 5A). We then examined whether CF31 could inhibit TNF-α activation of AKT. Treatment of HeLa cells with TNF-α for 30 minutes strongly activated AKT, which was inhibited by either CF31 or K-80003 in a dose-dependent manner (Fig. 5B). Both compounds at 20 μmol/L completely inhibited the effect of TNF-α on activating AKT. CF31 could also inhibit basal and TNF-α–induced AKT activation in other cancer cell lines, including A549 lung cancer and HepG2 liver cancer cells (Fig. 5C). All these cancer cell lines expressed high levels of tRXR-α, as revealed by immunoblotting using ΔN197 anti-RXR-α antibody recognizing the LBD of RXR-α (29; Fig. 5C).

Figure 5.

CF31 inhibits TNF-α–induced AKT activation. A, inhibition of basal AKT activation and induction of apoptosis by CF31. HeLa cells were treated with CF31 (10 μmol/L) or K-80003 (10 μmol/L) or sulindac (10 μmol/L) for 24 hours and analyzed by immunoblotting. B, inhibition of TNF-α–induced AKT activation by CF31. HeLa cells were treated with CF31 (5, 10, and 20 μmol/L) or K-80003 (10 and 20 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes and analyzed by immunoblotting. C, inhibition of AKT activation by CF31. Cells were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes. AKT activation was analyzed by immunoblotting. Expression of RXR-α and tRXR-α was analyzed by immunoblotting using ΔN197 anti-RXR-α antibody. D, inhibition of AKT activation by RXR-α siRNA. A549 cells transfected with control or RXR-α siRNA for 24 hours were treated with CF31 (10 μmol/L) for 12 hours before exposed to TNF-α (10 ng/mL) for 30 minutes. AKT activation was analyzed by immunoblotting. Reduction of RXR-α level was examined by using D20 anti-RXR-α antibody. E, transfection of SW480 cells with GFP-RXR-α/Δ80 sensitizes their response to CF31. SW480 and SW480/RXR-α/Δ80 were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes. AKT activation and RXR-α expression were analyzed by immunoblotting. Expression of endogenous and transfected GFP-RXR-α/Δ80 was examined by using ΔN197 anti-RXR-α antibody. F, CF31 inhibits tRXR-α–p85α interaction. A549 cells treated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes were analyzed for RXR-α–p85α interaction by coimmunoprecipitation using ΔN197 antibody.

Figure 5.

CF31 inhibits TNF-α–induced AKT activation. A, inhibition of basal AKT activation and induction of apoptosis by CF31. HeLa cells were treated with CF31 (10 μmol/L) or K-80003 (10 μmol/L) or sulindac (10 μmol/L) for 24 hours and analyzed by immunoblotting. B, inhibition of TNF-α–induced AKT activation by CF31. HeLa cells were treated with CF31 (5, 10, and 20 μmol/L) or K-80003 (10 and 20 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes and analyzed by immunoblotting. C, inhibition of AKT activation by CF31. Cells were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes. AKT activation was analyzed by immunoblotting. Expression of RXR-α and tRXR-α was analyzed by immunoblotting using ΔN197 anti-RXR-α antibody. D, inhibition of AKT activation by RXR-α siRNA. A549 cells transfected with control or RXR-α siRNA for 24 hours were treated with CF31 (10 μmol/L) for 12 hours before exposed to TNF-α (10 ng/mL) for 30 minutes. AKT activation was analyzed by immunoblotting. Reduction of RXR-α level was examined by using D20 anti-RXR-α antibody. E, transfection of SW480 cells with GFP-RXR-α/Δ80 sensitizes their response to CF31. SW480 and SW480/RXR-α/Δ80 were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes. AKT activation and RXR-α expression were analyzed by immunoblotting. Expression of endogenous and transfected GFP-RXR-α/Δ80 was examined by using ΔN197 anti-RXR-α antibody. F, CF31 inhibits tRXR-α–p85α interaction. A549 cells treated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes were analyzed for RXR-α–p85α interaction by coimmunoprecipitation using ΔN197 antibody.

Close modal

To study the role of tRXR-α in CF31 action, we first assessed the effect of RXR-α siRNA transfection. Transfection of RXR-α siRNAs in A549 cells, which reduced levels of both the full-length RXR-α and tRXR-α (29), abolished the inhibitory effect of CF31 on AKT activation either in the absence or presence of TNF-α (Fig. 5D). To complement this study, we determined whether overexpression of tRXR-α could enhance the inhibitory effect of CF31. For this purpose, we stably expressed GFP-RXR-α/Δ80 (29), a GFP-tagged RXR-α mutant lacking 80 N-terminal amino acid residues, in SW480 colon cancer cells that expressed little tRXR-α protein (Fig. 5E), SW480 cells exhibited low basal AKT activation, and their AKT activation was insensitive to regulation by TNFα and (Fig. 5E). However, stable expression of GFP-RXRα/Δ80 enhanced not only their basal AKT activation but also their response to TNF-α induction of AKT activation (Fig. 5E). Moreover, the sensitivity of SW480 cells to CF31 was restored, as it could effectively inhibit TNF-α–induced AKT activation in SW480/RXR-α/Δ80 cells. Thus, tRXR-α expression is involved in AKT activation by TNF-α and is essential for its inhibition by CF31.

Previously, we reported that tRXR-α binding to p85α was essential for the activation of the PI3K/AKT pathway (29). Thus, we determined whether CF31 inhibition of tRXR-α–dependent AKT activation could be attributed to its interference with tRXR-α interaction with p85α. Consistent with our previous observation, TNF-α induced interaction of tRXR-α with p85α, as revealed by coimmunoprecipitation using the ΔN197 anti-RXR-α antibody (Fig. 5F). However, when cells were cotreated with CF31, TNF-α–induced tRXR-α–p85α interaction was largely abolished. Thus, CF31 may modulate AKT activation through its ability to inhibit the interaction of tRXR-α with p85α.

CF31 induces tRXR-α–dependent apoptosis

To study whether CF31 inhibition of AKT activation could result in apoptosis, we first examined the effect of CF31 on activation of proapoptotic Bcl-2 family member Bax in HepG2 cells in the presence of TNF-α by immunostaining using a Bax conformation-sensitive antibody Bax/6A7 that recognizes active Bax protein (46). As shown in Fig. 6A, immunostaining with Bax/6A7 antibody showed that activated Bax protein was undetectable in control cells. However, when cells were treated with CF31, a strong immunostaining was observed, suggesting the activation of Bax by CF31. The apoptotic effect of CF31 in the presence of TNF-α was also illustrated by its induction of PARP cleavage in HepG2 and HeLa cells (Fig. 6B). Induction of PARP cleavage by TNF-α/CF31 combination treatment was associated with reduction of AKT activation, suggesting that the inhibition of AKT activity plays a role in their induction of apoptosis. In support of the role of AKT inhibition by the combination treatment, transfection of the constitutive-active AKT (CA-AKT) expression vector prevented its induction of PARP cleavage (Fig. 6C).

Figure 6.

CF31 induces RXR-α–dependent apoptosis. A, CF31 activation of Bax. HepG2 cells treated with CF31 (10 μmol/L; 12 hours) were immunostained with Bax/6A7 antibody and DAPI. About 40% CF31-treated cells showed Bax staining. B, CF31 induction of PARP cleavage. Cells were treated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes and analyzed by immunoblotting. C, regulation of CF31/TNF-α–induced PARP cleavage by AKT activation. HepG2 cells transfected with CA-AKT expression vector were treated with TNF-α (10 ng/mL) and/or CF31 (10 μmol/L) for 4 hours and analyzed by immunoblotting. D, inhibition of CF31-induced PARP cleavage by RXR-α siRNA. HeLa cells transfected with control or RXR-α siRNA were treated with CF31 (10 μmol/L) for 12 hours and analyzed by immunoblotting. E, RXR-α/Δ80 expression sensitizes A549 cells to apoptotic effect of CF31. A549 cells were transfected with Myc-RXR-α/Δ80, treated with CF31 (10 μmol/L) for 12 hours and analyzed by immunoblotting. F, RXR-α/Δ80 expression sensitizes MCF-7 cells to apoptotic effect of CF31. MCF-7 and MCF-7 cells transfected with GFP-RXR-α/Δ80 were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours. PARP cleavage were analyzed by immunoblotting.

Figure 6.

CF31 induces RXR-α–dependent apoptosis. A, CF31 activation of Bax. HepG2 cells treated with CF31 (10 μmol/L; 12 hours) were immunostained with Bax/6A7 antibody and DAPI. About 40% CF31-treated cells showed Bax staining. B, CF31 induction of PARP cleavage. Cells were treated with CF31 (10 μmol/L) for 12 hours before exposure to TNF-α (10 ng/mL) for 30 minutes and analyzed by immunoblotting. C, regulation of CF31/TNF-α–induced PARP cleavage by AKT activation. HepG2 cells transfected with CA-AKT expression vector were treated with TNF-α (10 ng/mL) and/or CF31 (10 μmol/L) for 4 hours and analyzed by immunoblotting. D, inhibition of CF31-induced PARP cleavage by RXR-α siRNA. HeLa cells transfected with control or RXR-α siRNA were treated with CF31 (10 μmol/L) for 12 hours and analyzed by immunoblotting. E, RXR-α/Δ80 expression sensitizes A549 cells to apoptotic effect of CF31. A549 cells were transfected with Myc-RXR-α/Δ80, treated with CF31 (10 μmol/L) for 12 hours and analyzed by immunoblotting. F, RXR-α/Δ80 expression sensitizes MCF-7 cells to apoptotic effect of CF31. MCF-7 and MCF-7 cells transfected with GFP-RXR-α/Δ80 were starved overnight and pretreated with CF31 (10 μmol/L) for 12 hours. PARP cleavage were analyzed by immunoblotting.

Close modal

We also determined the role of RXR-α in CF31-induced apoptosis by transfecting control and RXR-α siRNA in HeLa cells. Comparison of the effect of CF31 in cells transfected with control siRNA or RXR-α siRNA showed that transfection of RXR-α siRNA impaired the ability of CF31 to induce PARP cleavage and to inhibit AKT activation (Fig. 6D). To address the role of tRXR-α, we examined the effect of CF31 on PARP cleavage in cells stably expressing RXR-α/Δ80. In A549 cells, CF31 showed little effect on inducing PARP cleavage (Fig. 6E). However, it significantly induced PARP cleavage in cells stably expressing RXR-α/Δ80 tagged with Myc epitope, Myc-RXR-α/Δ80. Similar results were obtained in MCF-7 breast cancer cells stably expressing GFP-RXRα/Δ80 (Fig. 6F). Thus, the expression of tRXR-α plays a role in the apoptosis induction by CF31.

CF31 activates TNF-α–induced extrinsic apoptotic pathway

TNF-α is capable of inducing opposing biologic activities, such as cell survival and death, and its killing effect in cancer cells are often antagonized by its survival function that is mainly mediated by activation of the NF-κB and PI3K/AKT pathways (47, 48). Our observation that both inhibition of AKT activation and induction of apoptosis by CF31 occurred in the presence of TNF-α prompted us to study whether CF31 could convert TNF-α from a survival to a killing molecule. Thus, caspase-3 activation was examined in HeLa cells treated with CF31 or TNF-α alone or their combination. Treatment of cells with either CF31 or TNF-α alone had little effect on caspase-3 activation, whereas the combination treatment resulted in strong caspase-3 activation (Fig. 7A). The combination treatment also strongly induced the activation of caspase-8, the downstream mediator of the TNF-α–dependent apoptotic pathway (47, 48). Caspase-8 activation by CF31/TNF-α combination was also revealed by immunoblotting showing strong induction of cleaved caspase-8 products, p43/p41, an indication of caspase-8 activation (Fig. 7B). The induction of caspase-8 activation and PARP cleavage by the CF31/TNF-α combination was partially impaired by transfection of RXR-α siRNA (Fig. 7B), showing a role of RXR-α.

Figure 7.

Activation of extrinsic apoptotic pathway by CF31. A, induction of caspase-3 and -8 activity by CF31/TNF-α combination. HeLa cells were treated with CF31 (10 μmol/L) and/or TNF-α (10 ng/mL) for 4 hours and analyzed by caspase-3 and -8 activity assay kit. B, inhibition of CF31/TNF-α–induced caspases-8 cleavage by RXR-α siRNA. HeLa cells transfected with control or RXR-α siRNA were treated with TNF-α (10 ng/mL) and CF31 (10 μmol/L) and analyzed by immunoblotting. C and D, inhibition of CF31/TNF-α–induced PARP cleavage by caspase-8 inhibitor and siRNA. HeLa cells transfected with control or caspase-8 siRNA (C) or pretreated with Z-IETD-fmk (40 μmol/L; D) for 1 hour were treated with TNF-α (10 ng/mL) and CF31 (10 μmol/L), then analyzed by immunoblotting.

Figure 7.

Activation of extrinsic apoptotic pathway by CF31. A, induction of caspase-3 and -8 activity by CF31/TNF-α combination. HeLa cells were treated with CF31 (10 μmol/L) and/or TNF-α (10 ng/mL) for 4 hours and analyzed by caspase-3 and -8 activity assay kit. B, inhibition of CF31/TNF-α–induced caspases-8 cleavage by RXR-α siRNA. HeLa cells transfected with control or RXR-α siRNA were treated with TNF-α (10 ng/mL) and CF31 (10 μmol/L) and analyzed by immunoblotting. C and D, inhibition of CF31/TNF-α–induced PARP cleavage by caspase-8 inhibitor and siRNA. HeLa cells transfected with control or caspase-8 siRNA (C) or pretreated with Z-IETD-fmk (40 μmol/L; D) for 1 hour were treated with TNF-α (10 ng/mL) and CF31 (10 μmol/L), then analyzed by immunoblotting.

Close modal

Induction of PARP cleavage by CF31/TNF-α combination treatment was closely correlated with caspase-8 activation (Fig. 7B), suggesting an activation of TNF-α–dependent apoptotic pathway by CF31. To further address the role of caspase-8 activation in apoptosis induction by CF31, we examined the effect of caspase-8 siRNA transfection. Transfection of HeLa cells with caspase-8 siRNA reduced the level of p43/p41 and PARP cleavage (Fig. 7C). In addition, PARP cleavage induced by the CF31/TNF-α combination treatment was largely inhibited in cells treated with the caspase-8 inhibitor Z-IETD-fm (Fig. 7D). These results showed that apoptosis induction by the CF31/TNF-α combination treatment was largely mediated by their activation of TNF-α–mediated extrinsic apoptotic pathway, suggesting the role of CF31 in shifting the TNFα signaling from the survival to death.

Traditional medicinal plants and dietary factors provide a fertile ground for modern drug development, with some compounds, such as paclitaxel, etoposide, camptothecin, and vincristine, being successfully used as anticancer drugs (49). Xanthones, a class of 3-membered heterocyclic ring compounds mainly found as secondary metabolites in higher plants and microorganisms, exert very diverse biologic profiles, including antihypertensive, antioxidative, antithrombotic, and anticancer activity, depending on their diverse structures (30–33). A large number of naturally occurring and synthetic xanthones such as psorospermin (50), dimethylxanthesone-4-acetic acid (DMXAA; ref. 51), and α-mangostin (52) have shown potent anticancer activities. However, the biologic targets of xanthone compounds remain elusive (30–33). Cratoxylum is a small genus distributed in Southeast Asia with some of its species used medicinally, and xanthones are the most characteristic biologically active components of this genus (31, 35, 52, 53). We have previously identified several bioactive xanthones from the stems of Cratoxylum formosum ssp. pruniflorum (34, 35). We report here that CF31, one of the xanthones that we isolated, acts as a potent negative regulator of the tRXR-α–mediated PI3K/AKT survival signaling pathway by binding to RXR-α, thus identifying CF31 as a new lead for a class of anticancer agents targeting this newly identified cancer survival pathway.

CF31 binds to RXR-α in a unique mode and acts as a RXR-α antagonist. The LBP of RXR is highly restrictive to flexible and elongated ligands. The published crystal structures of RXR-α bound to natural or synthetic ligands show that a carboxylate group in these ligands forms salt bridges with basic residue Arg316 at the end of the L-shaped RXR-α LBP to establish anchoring ionic interaction for stabilization (41–43). However, CF31 lacks such a carboxylate moiety (Fig. 1A) and is therefore incapable of interacting with Arg316. This was supported by our mutagenesis study, which showed that Arg316 was not required for its antagonist effect (Fig. 2C). Although Arg316 was not required for CF31 binding, our MM-PB/SA analysis suggested that the binding of CF31 was stabilized by its extensive van der Waals interactions with several hydrophobic residues in the RXR-α (Fig. 2E), thus revealing a distinct binding mode for CF31. It is noteworthy that mutation of Arg316, which completely impaired the transactivation function of 9-cis-RA, did not show much effect on CD3254 (Fig. 2C), implying that CD3254 binding to RXR-α does not require its ionic interaction with Arg316. Very recently, crystal structure studies showed that begelovin, a RXR-α agonist lacking the carboxylate moiety, bound to RXR-α (54) in a mode similar to CF31. Thus, the LBP of RXR-α is more flexible than expected to mediate diverse activities of compounds with different structural features.

CF31 effectively inhibited constitutive and inducible AKT activation and cell survival in several cancer cell lines (Figs. 4 and 5). It was much more effective than sulindac, an nonsteroidal anti-inflammatory drug (NSAID) that was previously reported to inhibit tRXR-α–mediated AKT activation (29), and was comparable with K-80003, an improved sulindac analog (29), on inhibiting AKT activation (Fig. 5A and data not shown). The inhibitory effect of CF31 on AKT activation occurred at concentrations under that CF31 could bind to RXR-α, suggesting that it achieved its inhibitory effect on AKT activation by RXRα binding. In support of this conclusion, we showed that knocking down RXR-α expression by RXR-α siRNA impaired its inhibitory effects on basal and TNF-α–induced AKT activation (Fig. 5D), whereas overexpression of tRXR-α resulted in an enhancement (Fig. 5E).

Another unique property of CF31 is its ability to convert TNF-α from a survival molecule to a killer of cancer cells. TNF-α is a multifunctional cytokine that plays roles in diverse cellular events, such as cell survival and death (47, 48, 55, 56). The apoptotic effect of TNF-α is mediated by caspase-8–dependent apoptotic pathway, whereas its survival function involves activation of PI3K/AKT and NF-κB pathways. Because the death effect of TNF-α is antagonized or suppressed by its abnormally elevated survival function in cancer cells, TNF-α often acts as a survival instead of killer in the cells (47, 48, 55, 56). Because TNF-α is produced by malignant or host cells in the tumor microenvironment but not in normal cells, there has been tremendous interest in developing strategies to change a tumor-promoting microenvironment to a tumor-inhibiting state by shifting TNF-α signaling from survival to death (47, 48, 57–59). Our previous discovery that tRXR-α mediates AKT activation by TNF-α provides an opportunity to convert TNF-α's function from survival to death in cancer cells by targeting tRXR-α (29). Our current study provides several lines of evidence that CF31 could act as such a converter through its ability to suppress tRXRα–mediated AKT activation by TNF-α. First, we found that CF31 could potently induce PARP cleavage in cancer cells when used together with TNF-α (Fig. 6). Moreover, combination of CF31 and TNF-α led to a synergistic effect on PARP cleavage (Fig. 6C) and caspase activation (Fig. 7A), showing that the apoptotic effect of CF31 requires the TNF-α signaling pathway. Second, induction of PARP cleavage by the combination treatment in cancer cells was associated with their inhibition of AKT activation (Fig. 6B), which was also tRXR-α dependent (Fig. 6D–F). In contrast, transfection of CA-AKT abolished the inducing effect of CF31/TNF-α combination on PARP cleavage (Fig. 6C). Thus, inhibition of AKT activation was essential for their induction of apoptosis. Third, induction of caspase-3 activation and PARP cleavage by the CF31/TNF-α combination was mediated by activation of caspase-8 (Fig. 7), a critical mediator of TNF-α–induced intrinsic apoptotic pathway. Thus, CF31 can serve to relieve the antiapoptotic function of AKT, leading to activation of TNF-α–dependent apoptosis. Together, our results identify CF31 as a new converter of TNF-α survival signaling in cancer cells. With its ability to shift TNF-α signaling from survival to death by its unique RXR-α binding, CF31 represents a promising lead for a class of RXR-α modulators that selectively induce apoptosis of cancer cells. Because of the proven natural product drug discovery track record, identification of a natural product that targets an RXR-mediated cell survival pathway that regulates PI3K/AKT may offer a new therapeutic strategy to kill cancer cells.

No potential conflicts of interest were disclosed.

Conception and design: G.-H. Wang, F.-Q. Jiang, J.-Z. Zeng, Y. Su, X.-S. Yao, X.-K. Zhang

Development of methodology: G.-H. Wang, F.-Q. Jiang, Z.-P. Zeng, F. Chen, Y. Dai, J. Liu, J. Liu, H. Zhou, X.-K. Zhang

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.-H. Wang, F.-Q. Jiang, Z.-P. Zeng, Y. Dai

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G.-H. Wang, F.-Q. Jiang, Z.-P. Zeng, F. Chen, Y. Dai, J.-B. Chen, H.-F. Chen, Y. Su, X.-K. Zhang

Writing, review, and/or revision of the manuscript: Z.-P. Zeng, Y. Su, X.-S. Yao, X.-K. Zhang

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Z.-P. Zeng, J.-B. Chen, Y. Su, X.-S. Yao, X.-K. Zhang

Study supervision: X.-K. Zhang

Preparation of natural products and pure compounds for activity test: Y.-H. Duan

This work was supported by Grants from the U.S. Army Medical Research and Material Command (W81XWH-11-1-0677), the NIH (CA140980 and GM089927), the 985 Project from Xiamen University, the National Natural Science Foundation of China (NSFC-91129302, NSFC-30873146 and NSFC-81001664), and the Fundamental Research Funds for the Central Universities (2010121100, 2011121058, and 2010111081).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Altucci
L
,
Gronemeyer
H
. 
The promise of retinoids to fight against cancer
.
Nat Rev Cancer
2001
;
1
:
181
93
.
2.
Dawson
MI
,
Zhang
XK
. 
Discovery and design of retinoic acid receptor and retinoid X receptor class- and subtype-selective synthetic analogs of all-trans-retinoic acid and 9-cis-retinoic acid
.
Curr Med Chem
2002
;
9
:
623
37
.
3.
Liby
KT
,
Yore
MM
,
Sporn
MB
. 
Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer
.
Nat Rev Cancer
2007
;
7
:
357
69
.
4.
Mangelsdorf
DJ
,
Evans
RM
. 
The RXR heterodimers and orphan receptors
.
Cell
1995
;
83
:
841
50
.
5.
Huang
J
,
Powell
WC
,
Khodavirdi
AC
,
Wu
J
,
Makita
T
,
Cardiff
RD
, et al
Prostatic intraepithelial neoplasia in mice with conditional disruption of the retinoid X receptor alpha allele in the prostate epithelium
.
Cancer Res
2002
;
62
:
4812
9
.
6.
Li
M
,
Indra
AK
,
Warot
X
,
Brocard
J
,
Messaddeq
N
,
Kato
S
, et al
Skin abnormalities generated by temporally controlled RXRalpha mutations in mouse epidermis
.
Nature
2000
;
407
:
633
6
.
7.
Matsushima-Nishiwaki
R
,
Okuno
M
,
Adachi
S
,
Sano
T
,
Akita
K
,
Moriwaki
H
, et al
Phosphorylation of retinoid X receptor alpha at serine 260 impairs its metabolism and function in human hepatocellular carcinoma
.
Cancer Res
2001
;
61
:
7675
82
.
8.
Yamazaki
K
,
Shimizu
M
,
Okuno
M
,
Matsushima-Nishiwaki
R
,
Kanemura
N
,
Araki
H
, et al
Synergistic effects of RXR{alpha} and PPAR gamma ligands to inhibit growth in human colon cancer cells—phosphorylated RXR{alpha} is a critical target for colon cancer management 1
.
Gut
2007
;
56
:
1557
63
.
9.
Ariga
N
,
Moriya
T
,
Suzuki
T
,
Kimura
M
,
Ohuchi
N
,
Sasano
H
. 
Retinoic acid receptor and retinoid X receptor in ductal carcinoma in situ and intraductal proliferative lesions of the human breast
.
Jpn J Cancer Res
2000
;
91
:
1169
76
.
10.
Jiang
SY
,
Shen
SR
,
Shyu
RY
,
Yu
JC
,
Harn
HJ
,
Yeh
MY
, et al
Expression of nuclear retinoid receptors in normal, premalignant and malignant gastric tissues determined by in situ hybridization
.
Br J Cancer
1999
;
80
:
206
14
.
11.
Lotan
Y
,
Xu
XC
,
Shalev
M
,
Lotan
R
,
Williams
R
,
Wheeler
TM
, et al
Differential expression of nuclear retinoid receptors in normal and malignant prostates
.
J Clin Oncol
2000
;
18
:
116
21
.
12.
Picard
E
,
Seguin
C
,
Monhoven
N
,
Rochette-Egly
C
,
Siat
J
,
Borrelly
J
, et al
Expression of retinoid receptor genes and proteins in non–small-cell lung cancer
.
J Natl Cancer Inst
1999
;
91
:
1059
66
.
13.
Takiyama
Y
,
Miyokawa
N
,
Sugawara
A
,
Kato
S
,
Ito
K
,
Sato
K
, et al
Decreased expression of retinoid X receptor isoforms in human thyroid carcinomas
.
J Clin Endocrinol Metab
2004
;
89
:
5851
61
.
14.
Zhong
C
,
Yang
S
,
Huang
J
,
Cohen
MB
,
Roy-Burman
P
. 
Aberration in the expression of the retinoid receptor, RXRalpha, in prostate cancer
.
Cancer Biol Ther
2003
;
2
:
179
84
.
15.
Casas
F
,
Daury
L
,
Grandemange
S
,
Busson
M
,
Seyer
P
,
Hatier
R
, et al
Endocrine regulation of mitochondrial activity:involvement of truncated RXRalpha and c-Erb Aalpha1 proteins
.
FASEB J
2003
;
17
:
426
36
.
16.
Nagaya
T
,
Murata
Y
,
Yamaguchi
S
,
Nomura
Y
,
Ohmori
S
,
Fujieda
M
, et al
Intracellular proteolytic cleavage of 9-cis-retinoic acid receptor alpha by cathepsin L-type protease is a potential mechanism for modulating thyroid hormone action
.
J Biol Chem
1998
;
273
:
33166
73
.
17.
Prufer
K
,
Schroder
C
,
Hegyi
K
,
Barsony
J
. 
Degradation of RXRs influences sensitivity of rat osteosarcoma cells to the antiproliferative effects of calcitriol
.
Mol Endocrinol
2002
;
16
:
961
76
.
18.
Nomura
Y
,
Nagaya
T
,
Yamaguchi
S
,
Katunuma
N
,
Seo
H
. 
Cleavage of RXRalpha by a lysosomal enzyme, cathepsin L-type protease
.
Biochem Biophys Res Commun
1999
;
254
:
388
94
.
19.
Zeisig
BB
,
Kwok
C
,
Zelent
A
,
Shankaranarayanan
P
,
Gronemeyer
H
,
Dong
S
, et al
Recruitment of RXR by homotetrameric RARalpha fusion proteins is essential for transformation
.
Cancer Cell
2007
;
12
:
36
51
.
20.
Zhu
J
,
Nasr
R
,
Pérès
L
,
Riaucoux-Lormière
F
,
Honoré
N
,
Berthier
C
, et al
RXR is an essential component of the oncogenic PML/RARA complex in vivo
.
Cancer Cell
2007
;
12
:
23
35
.
21.
Cao
X
,
Liu
W
,
Lin
F
,
Li
H
,
Kolluri
SK
,
Lin
B
, et al
Retinoid X receptor regulates Nur77/TR3-dependent apoptosis [corrected] by modulating its nuclear export and mitochondrial targeting
.
Mol Cell Biol
2004
;
24
:
9705
25
.
22.
Ghose
R
,
Zimmerman
TL
,
Thevananther
S
,
Karpen
SJ
. 
Endotoxin leads to rapid subcellular re-localization of hepatic RXRalpha: a novel mechanism for reduced hepatic gene expression in inflammation
.
Nucl Recept
2004
;
2
:
4
.
23.
Katagiri
Y
,
Takeda
K
,
Yu
ZX
,
Ferrans
VJ
,
Ozato
K
,
Guroff
G
. 
Modulation of retinoid signalling through NGF-induced nuclear export of NGFI-B
.
Nat Cell Biol
2000
;
2
:
435
40
.
24.
Zimmerman
TL
,
Thevananther
S
,
Ghose
R
,
Burns
AR
,
Karpen
SJ
. 
Nuclear export of retinoid X receptor alpha in response to interleukin-1beta-mediated cell signaling: roles for JNK and SER260
.
J Biol Chem
2006
;
281
:
15434
40
.
25.
Dufour
JM
,
Kim
KH
. 
Cellular and subcellular localization of six retinoid receptors in rat testis during postnatal development: identification of potential heterodimeric receptors
.
Biol Reprod
1999
;
61
:
1300
8
.
26.
Fukunaka
K
,
Saito
T
,
Wataba
K
,
Ashihara
K
,
Ito
E
,
Kudo
R
. 
Changes in expression and subcellular localization of nuclear retinoic acid receptors in human endometrial epithelium during the menstrual cycle
.
Mol Hum Reprod
2001
;
7
:
437
46
.
27.
Li
H
,
Kolluri
SK
,
Gu
J
,
Dawson
MI
,
Cao
X
,
Hobbs
PD
, et al
Cytochrome c release and apoptosis induced by mitochondrial targeting of nuclear orphan receptor TR3
.
Science
2000
;
289
:
1159
64
.
28.
Lin
B
,
Kolluri
S
,
Lin
F
,
Liu
W
,
Han
YH
,
Cao
X
, et al
Conversion of Bcl-2 from protector to killer by interaction with nuclear orphan receptor Nur77/TR3
.
Cell
2004
;
116
:
527
40
.
29.
Zhou
H
,
Liu
W
,
Su
Y
,
Wei
Z
,
Liu
J
,
Kolluri
SK
, et al
NSAID sulindac and its analog bind RXRalpha and inhibit RXRalpha-dependent AKT signaling
.
Cancer Cell
2010
;
17
:
560
73
.
30.
Pinto
MM
,
Sousa
ME
,
Nascimento
MS
. 
Xanthone derivatives: new insights in biological activities
.
Curr Med Chem
2005
;
12
:
2517
38
.
31.
Na
Y
. 
Recent cancer drug development with xanthone structures
.
J Pharm Pharmacol
2009
;
61
:
707
12
.
32.
Pedraza-Chaverri
J
,
Cardenas-Rodriguez
N
,
Orozco-Ibarra
M
,
Perez-Rojas
JM
. 
Medicinal properties of mangosteen (Garcinia mangostana)
.
Food Chem Toxicol
2008
;
46
:
3227
39
.
33.
Akao
Y
,
Nakagawa
Y
,
Iinuma
M
,
Nozawa
Y
. 
Anti-cancer effects of xanthones from pericarps of mangosteen
.
Int J Mol Sci
2008
;
9
:
355
70
.
34.
Duan
YH
,
Dai
Y
,
Wang
GH
,
Chen
HF
,
Gao
H
,
Chen
JB
, et al
Xanthone and benzophenone glycosides from the stems of Cratoxylum formosum ssp. pruniflorum
.
Chem Pharm Bull (Tokyo)
2011
;
59
:
231
4
.
35.
Duan
YH
,
Dai
Y
,
Wang
GH
,
Zhang
X
,
Chen
HF
,
Chen
JB
, et al
Bioactive xanthones from the stems of Cratoxylum formosum ssp. pruniflorum
.
J Nat Prod
2010
;
73
:
1283
7
.
36.
Mahabusarakam
W
,
Nuangnaowarat
W
,
Taylor
WC
. 
Xanthone derivatives from Cratoxylum cochinchinense roots
.
Phytochemistry
2006
;
67
:
470
4
.
37.
Zhang
XK
,
Lehmann
J
,
Hoffmann
B
,
Dawson
MI
,
Cameron
J
,
Graupner
G
, et al
Homodimer formation of retinoid X receptor induced by 9-cis retinoic acid
.
Nature
1992
;
358
:
587
91
.
38.
Case
DA
,
Cheatham
TE
,
Darden
T
,
Gohlke
H
,
Luo
R
,
Merz
KM
, et al
The Amber biomolecular simulation programs
.
J Comput Chem
2005
;
26
:
1668
88
.
39.
Lu
J
,
Dawson
MI
,
Hu
QY
,
Xia
Z
,
Dambacher
JD
,
Ye
M
, et al
The effect of antagonists on the conformational exchange of the retinoid X receptor alpha ligand-binding domain
.
Magn Reson Chem
2009
;
47
:
1071
80
.
40.
Nahoum
V
,
Pérez
E
,
Germain
P
,
Rodríguez-Barrios
F
,
Manzo
F
,
Kammerer
S
, et al
Modulators of the structural dynamics of the retinoid X receptor to reveal receptor function
.
Proc Natl Acad Sci U S A
2007
;
104
:
17323
8
.
41.
Sato
Y
,
Ramalanjaona
N
,
Huet
T
,
Potier
N
,
Osz
J
,
Antony
P
, et al
The “Phantom Effect” of the Rexinoid LG100754: structural and functional insights
.
PLoS ONE
2010
;
5
:
e15119
.
42.
Egea
PF
,
Mitschler
A
,
Moras
D
. 
Molecular recognition of agonist ligands by RXRs
.
Mol Endocrinol
2002
;
16
:
987
97
.
43.
Egea
PF
,
Mitschler
A
,
Rochel
N
,
Ruff
M
,
Chambon
P
,
Moras
D
. 
Crystal structure of the human RXRalpha ligand-binding domain bound to its natural ligand: 9-cis retinoic acid
.
EMBO J
2000
;
19
:
2592
601
.
44.
Kollman
PA
,
Massova
I
,
Reyes
C
,
Kuhn
B
,
Huo
S
,
Chong
L
, et al
Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models
.
Acc Chem Res
2000
;
33
:
889
97
.
45.
Pommier
Y
. 
Topoisomerase I inhibitors: camptothecins and beyond
.
Nat Rev Cancer
2006
;
6
:
789
802
.
46.
Kolluri
SK
,
Zhu
X
,
Zhou
X
,
Lin
B
,
Chen
Y
,
Sun
K
, et al
A short Nur77-derived peptide converts Bcl-2 from a protector to a killer
.
Cancer Cell
2008
;
14
:
285
98
.
47.
Balkwill
F
. 
Tumour necrosis factor and cancer
.
Nat Rev Cancer
2009
;
9
:
361
71
.
48.
Mocellin
S
,
Nitti
D
. 
TNF and cancer: the two sides of the coin
.
Front Biosci
2008
;
13
:
2774
83
.
49.
Corson
TW
,
Crews
CM
. 
Molecular understanding and modern application of traditional medicines: triumphs and trials
.
Cell
2007
;
130
:
769
74
.
50.
Kupchan
SM
,
Streelman
DR
,
Sneden
AT
. 
Psorospermin, a new antileukemic xanthone from Psorospermum febrifugum
.
J Nat Prod
1980
;
43
:
296
301
.
51.
Jameson
MB
,
Thompson
PI
,
Baguley
BC
,
Evans
BD
,
Harvey
VJ
,
Porter
DJ
, et al
Clinical aspects of a phase I trial of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a novel antivascular agent
.
Br J Cancer
2003
;
88
:
1844
50
.
52.
Matsumoto
K
,
Akao
Y
,
Kobayashi
E
,
Ohguchi
K
,
Ito
T
,
Tanaka
T
, et al
Induction of apoptosis by xanthones from mangosteen in human leukemia cell lines
.
J Nat Prod
2003
;
66
:
1124
7
.
53.
Pattanaprateeb
P
,
Ruangrungsi
N
,
Cordell
GA
. 
Cytotoxic constituents from Cratoxylum arborescens
.
Planta Med
2005
;
71
:
181
3
.
54.
Zhang
H
,
Li
L
,
Chen
L
,
Hu
L
,
Jiang
H
,
Shen
X
. 
Structure basis of bigelovin as a selective RXR agonist with a distinct binding mode
.
J Mol Biol
2011
;
407
:
13
20
.
55.
Ozes
ON
,
Mayo
LD
,
Gustin
JA
,
Pfeffer
SR
,
Pfeffer
LM
,
Donner
DB
. 
NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase
.
Nature
1999
;
401
:
82
5
.
56.
Pincheira
R
,
Castro
AF
,
Ozes
ON
,
Idumalla
PS
,
Donner
DB
. 
Type 1 TNF receptor forms a complex with and uses Jak2 and c-Src to selectively engage signaling pathways that regulate transcription factor activity
.
J Immunol
2008
;
181
:
1288
98
.
57.
Bertazza
L
,
Mocellin
S
. 
Tumor necrosis factor (TNF) biology and cell death
.
Front Biosci
2008
;
13
:
2736
43
.
58.
van Horssen
R
,
Ten Hagen
TL
,
Eggermont
AM
. 
TNF-alpha in cancer treatment: molecular insights, antitumor effects, and clinical utility
.
Oncologist
2006
;
11
:
397
408
.
59.
Wang
X
,
Lin
Y
. 
Tumor necrosis factor and cancer, buddies or foes?
Acta Pharmacol Sin
2008
;
29
:
1275
88
.