Compounds isolated from members of the Zingiberaceae family are traditionally used as a medicine against inflammatory diseases, but little is known about the mechanism. Here, we report the isolation and structural identification of coronarin D [E-labda-8(17),12-diene-15-ol], a labdane-type diterpene, from Hedychium coronarium and delineate its mechanism of action. Because the transcription factor nuclear factor-κB (NF-κB) is a key mediator of inflammation, apoptosis, invasion, and osteoclastogenesis, we investigated the effect of coronarin D on NF-κB activation pathway, NF-κB-regulated gene products, and NF-κB-regulated cellular responses. The coronarin D inhibited NF-κB activation induced by different inflammatory stimuli and carcinogens. This labdane also suppressed constitutive NF-κB activity in different cell lines and inhibited IκBα kinase activation, thus leading to the suppression of IκBα phosphorylation, degradation, p65 nuclear translocation, and reporter gene transcription. Coronarin D also inhibited the NF-κB-regulated gene products involved in cell survival (inhibitor of apoptosis protein 1, Bcl-2, survivin, and tumor necrosis factor receptor-associated factor-2), proliferation (c-myc, cyclin D1, and cyclooxygenase-2), invasion (matrix metalloproteinase-9), and angiogenesis (vascular endothelial growth factor). Suppression of these gene products by the diterpene enhanced apoptosis induced by TNF and chemotherapeutic agents, suppressed TNF-induced cellular invasion, and abrogated receptor activator of NF-κB ligand-induced osteoclastogenesis. Coronarin D was found to be more potent than its analogue coronarin D acid. Overall, our results show that coronarin D inhibited NF-κB activation pathway, which leads to inhibition of inflammation, invasion, and osteoclastogenesis, as well as potentiation of apoptosis. [Mol Cancer Ther 2008;7(10):3306–17]

Many plants belonging to the Zingiberaceae family are extensively used in traditional systems of medicine in India, China, and Southeast Asian countries. One such plant is the white ginger lily, which originated in the Himalayan region of Nepal and India (Hedychium coronarium and Hedychium flavescens) and is commonly called “dolan champa” in Hindi, “takhellei” in Manipuri, “sonataka” in Marathi, and “suruli sugandhi” in Kannada. The rhizome of this plant is used by certain tribal groups of Bihar (India) as a febrifuge and is considered as an antirheumatic, tonic, and excitant in Moluccas. The base of the stem is chewed and the juice is applied to reduce swelling (1). The native Hawaiians refer to this plant as “awapuhi” (native wild shampoo ginger) and use the juice of mature seeds as a hair and skin treatment. In Cuba, where it is the national flower it is known as the “flor de mariposa” (literally, “butterfly flower”). Extracts from H. coronarium have exhibited anti-inflammatory activity against rat paw edema (2) and diuretic activity in rats (3).

The active principle(s) responsible for these activities is not known. Several labdane-type diterpenes (coronarin A, B, C, D, E, and F) have been isolated from the rhizome of H. coronarium (4, 5), but only little is known about these coronarins. The studies showed that coronarin A inhibited the proliferation of human umbilical vein endothelial cells (6). Coronarin D is known to inhibit the release of β-hexosaminidase from RBL-2H3 cells (7) and acetic acid-induced vascular permeability in mice (8). Whereas coronarin D and E were reported to inhibit nitric oxide production in lipopolysaccharide (LPS)-induced mouse peritoneal macrophages (9), coronarin A has been shown to be cytotoxic in tumor cells (10). However, the pathway by which these diterpenes exert their anti-inflammatory and cytotoxic effects is unclear. One of the major pathways linked with inflammation and growth modulation is the nuclear factor-κB (NF-κB) pathway.

NF-κB is an inducible transcription factor for genes involved in cell survival, adhesion, inflammation, differentiation, and growth. It regulates genes that are critical in inflammation and in the early and late stages of aggressive cancers, including cyclooxygenase-2, 5-lipoxygenase, cyclin D1, apoptosis suppressor proteins such as Bcl-2 and Bcl-xL, and genes required for metastasis and angiogenesis such as matrix metalloproteinase (MMP) and vascular endothelial growth factor (VEGF; ref. 10).

Because NF-κB activation is a mediator of both inflammation and growth modulation, we hypothesized that the anti-inflammatory and cytotoxic effects of coronarin D are due to its ability to inhibit NF-κB and NF-κB-regulated gene expression. In our study, coronarin D inhibited the NF-κB activation induced by different agents, inhibited NF-κB-regulated gene expression, and abrogated receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. It also potentiated chemotherapeutic agent-induced apoptosis.

Isolation of Coronarin D

Dried, powdered rhizomes of H. flavescens (525 g), which belongs to the Zingiberaceae family, were collected from Munnar in Kerala, India, and extracted with 3 L hexane using a Soxhlet extraction apparatus for 24 h. Removal of solvent under reduced pressure using a rotary evaporator yielded 19.5 g of a dark brown residue. This crude extract was subjected to column chromatography using 400 g silica gel (100-200 mesh) and eluted with petroleum ether-ethyl acetate mixture of increasing polarity. Successive fractions were combined based on their behavior on TLC, and the solvent was evaporated to give 17 fraction pools. Fraction pools 10 and 11 (total 3.7 g), obtained by elution of the column with petroleum ether-ethyl acetate in an 80:20 ratio, were found to contain the major product, which was further purified by once again separating it using column chromatography. This yielded the major compound, coronarin D, as colorless needles (2.05 g; crystallized from a petroleum ether-dichloromethane mixture), mp 102-103°C. [a]D(ref 1) +100. IR (KBr, umax/cm): 3455, 3087, 2925, 2856, 1726, 1669, 1642, 1445, 1345, 1208, 939, 889. 1H NMR (300 MHz): d 6.69 (br s, 1H), 5.94 (br s, 1H), 4.81 (s, 1H), 4.36 (br s, 1H), 2.99 (m, 1H), 2.71 (m, 1H), 2.40-2.36 (m, 2H), 2.20 (m, 1H), 2.05-1.08 (m, 12H), 0.89 (s, 3H), 0.82 (s, 3H), 0.71 (s, 3H). 13C NMR (75 MHz): d 170.65, 148.01, 143.45, 124.48, 107.52, 96.46, 56.02, 55.21, 41.90, 39.94, 39.12, 37.68, 33.57, 33.45, 25.58, 23.99, 21.62, 19.21, 14.23. UVlmax methanol nm: 239. LRMS: 318, C20H30O3, require 318. 1H NMR, IR, and UV were identical to that of coronarin D, as reported earlier.

Conversion of Coronarin D to Coronarin D Acid

NaBH4 (100 mg, 2.64 mmol) was added in small portions to a solution of coronarin D (125 mg, 0.4 mmol) in methanol (12 mL) maintained at 15°C, and the mixture was stirred at room temperature (25°C) for 30 min. After the reaction was complete according to TLC, the methanol was completely removed under vacuum pressure and the residue was diluted with water (15 mL) and extracted with dichloromethane (3 × 25 mL). The combined organic extract was washed with brine and dried over anhydrous Na2SO4. Evaporation of solvent yielded the crude product, which was purified by column chromatography over silica gel. On elution with hexane/ethyl acetate (3:1), 98 mg (80%) colorless E-labda-8(17),12-diene-15-ol-16-oic acid (coronarin D acid) crystals was obtained (mp 144-146°C). IR (KBr, umax/cm): 3438, 2942, 2901, 2854, 1682, 1641, 1418, 1264, 1199, 1122, 1046, 998, 880, 762. 1H NMR (300 MHz): d 6.97 (t, 1H, J = 6.40 Hz), 4.83 (s, 1H), 4.39 (s, 1H), 3.74 (t, 2H, J = 6.15 Hz), 2.61 (t, 2H, J = 6.03 Hz), 2.42-1.08 (m, 14H), 0.88 (s, 3H), 0.82 (s, 3H), 0.73 (s, 3H). 13C NMR (75 MHz): d 172.90, 149.95, 148.22, 127.60, 1.7.71, 61.73, 56.56, 55.38, 42.03, 39.54, 39.22, 37.89, 33.57, 30.20, 24.12, 24.08, 21.73, 19.30, 14.30. C/H analysis: Calculated for C20H32O3; C, 74.96%; H, 10.07%; Found: C, 74.88%; H, 10.44%.

Materials

A stock solution (50 mmol/L) of the compound was prepared in DMSO and used for further studies. Bacteria-derived human recombinant tumor necrosis factor (TNF), purified to homogeneity with a specific activity of 5 × 107 units/mg, was kindly provided by Genentech. Cigarette smoke condensate, prepared as described previously (11), was kindly supplied by Dr. G. Gairola (University of Kentucky). Phorbol myristate acetate, okadaic acid, LPS, H2O2, and Taxol were obtained from Sigma. Penicillin, streptomycin, RPMI 1640, Iscove's modified Dulbecco's medium, DMEM, DMEM/F-12, and fetal bovine serum were obtained from Invitrogen. Anti-β-actin was obtained from Sigma-Aldrich. Anti-p65, anti-p50, anti-IκBα, anti-poly(ADP-ribose) polymerase, anti-intercellular adhesion molecule-1, MMP-9, cyclin D1, c-myc, Bcl-2, anti-cyclooxygenase-2, and anti-inhibitor of apoptosis protein 1 were obtained from Santa Cruz Biotechnology. Phosphospecific anti-IκBα (Ser32) and anti-p65 (Ser536) were purchased from Cell Signaling. Anti-IκBα kinase (IKK)-α and -β antibodies were kindly provided by Imgenex.

Cell Lines

The human cell lines KBM-5 (chronic myeloid leukemia), A293 (embryonic kidney carcinoma), H1299 (lung adenocarcinoma), U266 (multiple myeloma), 253JBV (bladder cancer), MCF-7 (breast cancer), SKOV3 (ovarian cancer), HT29 (colon cancer), and PANC-1 (pancreatic cancer) and the murine monocytic cell line RAW 264.7 were obtained from the American Type Culture Collection. Cells were cultured as follows: KBM-5 in Iscove's modified Dulbecco's medium with 15% fetal bovine serum; H1299, K562, MCF-7, SKOV3, and U266 in RPMI 1640; A293, HT29, and PANC-1 in DMEM; and RAW 264.7 in DMEM/F-12 supplemented with 10% fetal bovine serum. Culture medium was supplemented with 100 units/mL penicillin and 100 μg/mL streptomycin.

Electrophoretic Mobility Shift Assay

To determine NF-κB activation, we used electrophoretic mobility shift assays (EMSA) as described previously (12). In brief, nuclear extracts prepared from TNF-treated cells were incubated with 32P-end-labeled 45-mer double-stranded NF-κB oligonucleotide (15 μg protein with 16 fmol DNA) from the HIV long terminal repeat 5′-TTGTTACAAGGGACTTTCCGCTGGGGACTTTCCAGGGAGGCGTGG-3′ (boldface indicates NF-κB binding sites) for 30 min at 37°C. The DNA-protein complex that formed was separated from the free oligonucleotide on 6.6% native polyacrylamide gels. A double-stranded mutated oligonucleotide, 5′-TTGTTACAACTCACTTTCCGCTGCTCACTTTCCAGGGAGGCGTGG-3′, was used to evaluate the specificity of the NF-κB binding to the DNA. The specificity was also determined through competition with the unlabeled oligonucleotide. The dried gels were visualized and the radioactive bands quantitated with a Storm 220 PhosphorImager (Amersham Biosciences) using ImageQuant software Molecular Dynamics.

Western Blot Analysis

To determine the levels of protein expression in the cytoplasm and nucleus, we fractionated extracts using SDS-PAGE as described previously (13). The proteins were then electrotransferred to nitrocellulose membranes and blotted with each antibody, and the expression of proteins was detected with an enhanced chemiluminescence reagent (Amersham Biosciences). We quantitated the bands with NIH imaging software.

IKK Assay

To determine the effects of coronarin D on TNF-induced IKK activation, we used the IKK assay as described previously (13). In brief, to determine the total amounts of IKK-α and IKK-β in each sample, we resolved 50 μg whole-cell protein by using 7.5% SDS-PAGE, electrotransferred the proteins to a nitrocellulose membrane, and blotted them with anti-IKK-α or anti-IKK-β antibodies.

NF-κB-Dependent Reporter Gene Expression Assay

We performed a NF-κB-dependent reporter gene expression assay as described previously (14). The effects of coronarin D on NF-κB-dependent reporter gene transcription as induced by TNF, TNF receptor (TNFR), TNFR-associated death domain (TRADD), TNFR-associated factor 2 (TRAF2), NF-κB-inducing kinase (NIK), IKK-β, and p65 were analyzed using a secretory alkaline phosphatase (SEAP) assay as described previously (14).

Immunocytochemical Analysis of NF-κB p65 Localization

The effects of coronarin D on p65 nuclear translocation were evaluated by an immunocytochemical analysis as described previously (13).

Cytotoxicity Assay

The effect of coronarin D on the cytotoxic effects of chemotherapeutic agents was determined by the MTT uptake method as described previously (15). Briefly, 5,000 cells per well were incubated with different chemotherapeutic agents such as doxorubicin (KBM-5 and U266; 1-100 nmol/L), gemcitabine (PANC-1 and 253JBV; 1-100 nmol/L), 5-fluorouracil (H1299 and HT29; 0.1-5 μmol/L), cisplatin (OSC 19; 0.01-0.5 μg/mL), and docetaxel (SKOV3 and MCF-7; 0.5-5 nmol/L) in combination with coronarin D (10 μmol/L) for 24 h and the cell viability was measured and the IC50 was determined.

Live/Dead Assay

To determine membrane permeability, we used the Live/Dead assay (Molecular Probes), which measures intracellular esterase activity and plasma membrane integrity. This assay was done as described previously (13).

Annexin V Assay

An early indicator of apoptosis is the rapid translocation and accumulation of the membrane phospholipid phosphatidylserine from the cytoplasmic interface of the membrane to the extracellular surface. This loss of membrane asymmetry can be detected by using the binding properties of Annexin V. To identify apoptosis, we used an Annexin V antibody, which was conjugated with a fluorescent dye, FITC, as described previously. Briefly, cells were preincubated with coronarin D, treated with TNF for 16 h at 37°C, and subjected to Annexin V staining. The cells were washed in PBS, resuspended in 100 μL binding buffer containing a FITC-conjugated anti-Annexin V antibody, and then analyzed with a flow cytometer (FACSCalibur, BD Biosciences; ref. 13).

Terminal Deoxynucleotidyl Transferase-Mediated dUTP Nick End Labeling Assay

We also determined cytotoxicity using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling method, which is used to evaluate DNA strand breaks during apoptosis using an in situ cell death detection reagent. This assay was done as described previously (13).

Invasion Assay

Extracellular matrix invasion is a crucial step in tumor metastasis. Therefore, we determined how invasion was affected by coronarin D with an invasion assay as described previously (15). A Matrigel basement membrane matrix extracted from a Engelbreth-Holm-Swarm mouse tumor (BD Biosciences) was reconstituted and used for this assay.

Osteoclast Differentiation Assay

To determine whether coronarin D inhibits osteoclastogenesis, we performed an osteoclast differentiation assay as described previously (16). In brief, 1 × 104 RAW 264.7 cells per well were cultured in 24-well dishes and allowed to adhere overnight. They were then exposed to RANKL, with or without coronarin D, for different numbers of days and examined for osteoclast formation.

We isolated coronarin D from the rhizomes of H. flavascens and confirmed the structure using 1H NMR, IR, and UV analysis. The structure of coronarin D is shown in Fig. 1A. We investigated the effects of coronarin D on the NF-κB activation pathway as induced by various carcinogens and inflammatory stimuli. We also evaluated the effect of coronarin D on NF-κB-regulated gene expression and NF-κB-mediated cellular responses. Because TNF is one of the most potent proinflammatory cytokines and the most potent activator of NF-κB, and because the TNF-induced NF-κB activation pathway has been well characterized, we determined the effects of coronarin D on TNF-induced NF-κB activation.

Figure 1.

A, structure of coronarin D. B, coronarin D blocked NF-κB activation induced by TNF, interleukin-1β, LPS, phorbol myristate acetate (PMA), okadaic acid (OA), H2O2, and cigarette smoke condensate (CSC). Human myeloid leukemia KBM-5 cells (2 × 106) were preincubated for 8 h at 37°C with 50 μmol/L coronarin D and then treated with TNF (0.1 nmol/L, 30 min), H2O2 (500 μmol/L, 2 h), interleukin-1β (100 ng/mL, 30 min), phorbol myristate acetate (25 ng/ml, 1 h), okadaic acid (500 nmol/L, 4 h), LPS (100 ng/ml, 2 h), and cigarette smoke condensate (10 μg/mL, 1 h). Nuclear extracts were prepared and assayed for NF-κB activation using EMSA. Control: untreated cells with activators. C,top, coronarin D suppressed TNF-induced NF-κB activation in a dose-dependent manner. KBM-5 cells (2 × 106) were incubated with different concentrations of coronarin D for 8 h and then treated with 0.1 nmol/L TNF for 30 min. Nuclear extracts were prepared and assayed for NF-κB activation by EMSA. Bottom, coronarin D suppressed TNF-induced NF-κB activation in a time-dependent manner. KBM-5 (2 × 106) cells were incubated with 50 μmol/L coronarin D for the indicated periods and then treated with 0.1 nmol/L TNF for 30 min. Nuclear extracts were then prepared and assayed for NF-κB activation by EMSA. D,top, coronarin D blocked TNF-induced NF-κB activation in human leukemia K562 and lung adenocarcinoma H1299 cells that had been preincubated with 50 μmol/L coronarin D for 8 h and 0.1 nmol/L TNF for 30 min. Bottom, coronarin D suppressed the constitutive activation of NF-κB in U266, 253JBV, and PANC-1 cells. Cells were incubated with 50 μmol/L coronarin D for 8 h. Nuclear extracts were then prepared and assayed for NF-κB activation by EMSA.

Figure 1.

A, structure of coronarin D. B, coronarin D blocked NF-κB activation induced by TNF, interleukin-1β, LPS, phorbol myristate acetate (PMA), okadaic acid (OA), H2O2, and cigarette smoke condensate (CSC). Human myeloid leukemia KBM-5 cells (2 × 106) were preincubated for 8 h at 37°C with 50 μmol/L coronarin D and then treated with TNF (0.1 nmol/L, 30 min), H2O2 (500 μmol/L, 2 h), interleukin-1β (100 ng/mL, 30 min), phorbol myristate acetate (25 ng/ml, 1 h), okadaic acid (500 nmol/L, 4 h), LPS (100 ng/ml, 2 h), and cigarette smoke condensate (10 μg/mL, 1 h). Nuclear extracts were prepared and assayed for NF-κB activation using EMSA. Control: untreated cells with activators. C,top, coronarin D suppressed TNF-induced NF-κB activation in a dose-dependent manner. KBM-5 cells (2 × 106) were incubated with different concentrations of coronarin D for 8 h and then treated with 0.1 nmol/L TNF for 30 min. Nuclear extracts were prepared and assayed for NF-κB activation by EMSA. Bottom, coronarin D suppressed TNF-induced NF-κB activation in a time-dependent manner. KBM-5 (2 × 106) cells were incubated with 50 μmol/L coronarin D for the indicated periods and then treated with 0.1 nmol/L TNF for 30 min. Nuclear extracts were then prepared and assayed for NF-κB activation by EMSA. D,top, coronarin D blocked TNF-induced NF-κB activation in human leukemia K562 and lung adenocarcinoma H1299 cells that had been preincubated with 50 μmol/L coronarin D for 8 h and 0.1 nmol/L TNF for 30 min. Bottom, coronarin D suppressed the constitutive activation of NF-κB in U266, 253JBV, and PANC-1 cells. Cells were incubated with 50 μmol/L coronarin D for 8 h. Nuclear extracts were then prepared and assayed for NF-κB activation by EMSA.

Close modal

Coronarin D Inhibits NF-κB Activation

Because TNF, H2O2, phorbol myristate acetate, okadaic acid, interleukin-1β, LPS, and cigarette smoke condensate are potent activators of NF-κB (10, 1719), we determined the effect of coronarin D on NF-κB activation by all these agents. Although all agents activated NF-κB in human myeloid leukemia KBM-5 cells, coronarin D blocked this activation (Fig. 1B), and cells were fully viable at this concentration and exposure time. These results suggest that coronarin D acts at a step in the NF-κB activation pathway that is common to all these agents.

TNF is one of the most potent activators of NF-κB, and the mechanism of NF-κB activation has been well established; therefore, we determined the effects of coronarin D on TNF-induced NF-κB activation. In KBM-5 cells, coronarin D suppressed TNF-induced NF-κB activation in a dose-dependent manner, and this inhibition was observed at a concentration even as low as 10 μmol/L: TNF-induced NF-κB activation was completely suppressed after 8 h at 50 μmol/L coronarin D (Fig. 1C, top).

Next, we determined the minimum exposure time required for coronarin D to inhibit TNF-mediated NF-κB activation. The cells were exposed to the inhibitor for 2, 4, 8, or 12 h and then treated with TNF for 30 min. We found that coronarin D started to inhibit TNF-induced NF-κB activation after only 2 h and completely suppressed it after 8 h (Fig. 1C, bottom). However, under the same conditions, coronarin D alone (in the absence of TNF) had no effect on NF-κB activation.

Because the signal transduction pathway mediated by NF-κB may be distinct in different cell types, we also determined whether coronarin D blocked TNF-induced NF-κB activation in K562 and H1299 cells (Fig. 1D, top). We found that coronarin D inhibited TNF-induced NF-κB activation in these cells, indicating that the coronarin D-induced inhibition of NF-κB activation is not cell type specific.

Most tumor cells express constitutively active NF-κB (20), although the mechanism of constitutive activation is not well understood. In this study, we found that incubation with coronarin D suppressed the constitutive activation of NF-κB in U266 (multiple myeloma), 253JBV (bladder cancer), and PANC-1 (pancreatic cancer) cells, which are known to express constitutively active NF-κB (Fig. 1D, bottom).

Whether coronarin D-induced inhibition of NF-κB activation is reversible was examined. To determine this, U266 cells were treated with coronarin D for 8 h, washed with PBS two times, resuspended in a fresh medium, harvested after 0, 2, 4, 8, 12, and 24 h, prepared the nuclear extracts, and analyzed for NF-κB activity by EMSA. Our results show that coronarin D-induced NF-κB inhibition is not reversible (Supplementary Fig. S1).3

3

Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

Coronarin D Inhibits TNF-Dependent IκBα Phosphorylation and Degradation

To determine how coronarin D inhibits TNF-induced NF-κB activation, we exposed the cells to coronarin D for 8 h and then treated them with TNF for different periods. Nuclear extracts were prepared and analyzed for NF-κB; cytoplasmic extracts were prepared and analyzed for IκBα. We found that coronarin D completely inhibited TNF-induced NF-κB activation (Fig. 2A) and also suppressed TNF-induced IκBα degradation (Fig. 2B, middle) as determined by Western blot analysis. The results also showed that coronarin D inhibited TNF-induced IκBα phosphorylation (Fig. 2B, top).

Figure 2.

A, coronarin D inhibited the time-dependent TNF-induced activation of NF-κB. KBM-5 (2 × 106) cells were preincubated with 50 μmol/L coronarin D for 8 h. They were then treated with 0.1 nmol/L TNF for the indicated times and analyzed for NF-κB activation by EMSA. B, coronarin D suppressed the TNF-induced phosphorylation and degradation of IκBα. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h for the indicated times. Cytoplasmic extracts were prepared, fractionated by 10% SDS-PAGE, and electrotransferred to a nitrocellulose membrane. A Western blot analysis was performed using anti-IκBα. C, coronarin D suppressed the TNF-induced activation of IKK. KBM-5 cells were pretreated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for the indicated times. Whole-cell extracts were immunoprecipitated with an antibody against IKK-α and analyzed by immune complex kinase assay as described in Materials and Methods. To determine the effect of coronarin D on the level of IKK proteins, whole-cell extracts were fractionated by SDS-PAGE and examined by Western blot analysis using anti-IKK-α and anti-IKK-β antibodies. D, effects of coronarin D on TNF-induced phosphorylation of p65. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 0.1 nmol/L TNF for the indicated times. Nuclear extracts were prepared, fractionated by 10% SDS-PAGE, and electrotransferred to a nitrocellulose membrane. A Western blot analysis was done using phosphospecific p65 (top). Immunocytochemical analysis of p65 localization. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h TNF for 15 min and subjected to immunocytochemical analysis as described in Materials and Methods (bottom).

Figure 2.

A, coronarin D inhibited the time-dependent TNF-induced activation of NF-κB. KBM-5 (2 × 106) cells were preincubated with 50 μmol/L coronarin D for 8 h. They were then treated with 0.1 nmol/L TNF for the indicated times and analyzed for NF-κB activation by EMSA. B, coronarin D suppressed the TNF-induced phosphorylation and degradation of IκBα. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h for the indicated times. Cytoplasmic extracts were prepared, fractionated by 10% SDS-PAGE, and electrotransferred to a nitrocellulose membrane. A Western blot analysis was performed using anti-IκBα. C, coronarin D suppressed the TNF-induced activation of IKK. KBM-5 cells were pretreated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for the indicated times. Whole-cell extracts were immunoprecipitated with an antibody against IKK-α and analyzed by immune complex kinase assay as described in Materials and Methods. To determine the effect of coronarin D on the level of IKK proteins, whole-cell extracts were fractionated by SDS-PAGE and examined by Western blot analysis using anti-IKK-α and anti-IKK-β antibodies. D, effects of coronarin D on TNF-induced phosphorylation of p65. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 0.1 nmol/L TNF for the indicated times. Nuclear extracts were prepared, fractionated by 10% SDS-PAGE, and electrotransferred to a nitrocellulose membrane. A Western blot analysis was done using phosphospecific p65 (top). Immunocytochemical analysis of p65 localization. KBM-5 cells (2 × 106) were preincubated with 50 μmol/L coronarin D for 8 h TNF for 15 min and subjected to immunocytochemical analysis as described in Materials and Methods (bottom).

Close modal

IKK is required for the TNF-induced phosphorylation of IκBα and the phosphorylation of p65 (20). Because coronarin D inhibited the phosphorylation of IκBα, we analyzed its effects on TNF-induced IKK activation using immune complex kinase assays. We found that coronarin D suppressed TNF-induced IKK activation (Fig. 2C). However, neither TNF nor coronarin D affected the expression of IKK proteins. Whether coronarin D inhibits IKK directly was also examined. The results show that coronarin D directly inhibits the IKK activity (Supplementary Fig. S2).3

Above, we have shown that coronarin D can suppress constitutive active NF-κB. Whether this inhibition is mediated through inhibition of IKK was examined. Our results show that coronarin D inhibits constitutive active IKK in these cells (Supplementary Fig. S3).3

Coronarin D Inhibits TNF-Induced p65 Phosphorylation and Nuclear Translocation

The phosphorylation of p65 is required for the transactivation of NF-κB. Using phosphospecific anti-p65 (Ser536) antibody, we determined that TNF did induce p65 phosphorylation, which was substantially inhibited by coronarin D (Fig. 2D, top).

Whether coronarin D inhibits TNF-induced p65 nuclear translocation was examined by the immunocytochemical method. We found that TNF induced the nuclear translocation of p65 and coronarin D blocked the translocation (Fig. 2D, bottom).

Coronarin D Suppresses TNF-Induced NF-κB-Dependent Reporter Gene Expression

Because DNA binding alone is not always related to NF-κB-dependent gene transcription (21), we also analyzed the effects of coronarin D on TNF-induced reporter activity. After cells were transiently transfected with the NF-κB-regulated SEAP reporter construct, incubated with coronarin D, and stimulated with TNF, we found that TNF induced NF-κB reporter activity and that this activity was substantially suppressed by coronarin D (Fig. 3A). These results suggest that coronarin D inhibits TNF-induced gene expression. In addition, DN-IκBα plasmid suppressed TNF-induced reporter activity, indicating the specificity of the plasmid.

Figure 3.

Coronarin D inhibited the TNF-induced expression of the NF-κB-dependent genes TNFR1, TRADD, TRAF2, NIK, IKK-β, and p65. A and B, A293 cells were transiently transfected with an NF-κB-containing SEAP reporter gene plasmid, alone or with the indicated plasmids, for 24 h. After transfection, cells were washed and treated with 50 μmol/L coronarin D for 8 h. To evaluate the effects of TNF, we treated cells with 0.1 nmol/L TNF for an additional 24 h. The supernatants of the culture medium were assayed for SEAP activity as described in Materials and Methods.

Figure 3.

Coronarin D inhibited the TNF-induced expression of the NF-κB-dependent genes TNFR1, TRADD, TRAF2, NIK, IKK-β, and p65. A and B, A293 cells were transiently transfected with an NF-κB-containing SEAP reporter gene plasmid, alone or with the indicated plasmids, for 24 h. After transfection, cells were washed and treated with 50 μmol/L coronarin D for 8 h. To evaluate the effects of TNF, we treated cells with 0.1 nmol/L TNF for an additional 24 h. The supernatants of the culture medium were assayed for SEAP activity as described in Materials and Methods.

Close modal

Coronarin D Inhibits NF-κB Activation Induced by TNFR1, TRADD, TRAF2, NIK, IKK, and p65

TNF-induced NF-κB activation is mediated through the sequential interaction of TNFR and TRADD, TRAF2, NIK, IKK, and p65 and results in the phosphorylation of IκBα (22). Therefore, to further examine the effects of coronarin D on TNF-induced NF-κB activation, we transiently transfected cells with a NF-κB-regulated SEAP reporter construct and TNFR1-, TRADD-, TRAF2-, NIK-, IKK-β-, or p65-expressing plasmids. Cells were then treated with coronarin D and monitored for NF-κB-dependent SEAP expression. We found that coronarin D suppressed the NF-κB activation induced by TNFR1, TRADD, TRAF2, NIK, IKK-β, and p65 (Fig. 3B).

Coronarin D Inhibits TNF-Induced Expression of Proliferative, Antiapoptotic, and Metastatic Gene Products

NF-κB activation has been shown to regulate the expression of c-myc, cyclin D1, and cyclooxygenase-2 (2325), gene products that are involved in the proliferation of various tumor cells. Therefore, we determined whether the expression of these gene products is modulated by coronarin D. As shown in Fig. 4A, TNF induced the expression of all these gene products, and coronarin D suppressed it.

Figure 4.

Coronarin D inhibited the expression of TNF-induced gene expression. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and 1 nmol/L TNF for the indicated times. Whole-cell extracts were prepared and analyzed by Western blot analysis using the indicated antibodies. A, coronarin D inhibited the TNF-induced expression of the proliferative gene c-myc, cyclin D1, and cyclooxygenase-2 (COX-2). B, coronarin D inhibited the TNF-induced, NF-κB-regulated expression of antiapoptotic gene products inhibitor of apoptosis protein 1 (IAP1), TRAF-2, survivin, and Bcl-2. C, coronarin D suppressed the TNF-induced expression of metastatic gene products anti-intercellular adhesion molecule-1 (ICAM-1), MMP-9, and VEGF.

Figure 4.

Coronarin D inhibited the expression of TNF-induced gene expression. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and 1 nmol/L TNF for the indicated times. Whole-cell extracts were prepared and analyzed by Western blot analysis using the indicated antibodies. A, coronarin D inhibited the TNF-induced expression of the proliferative gene c-myc, cyclin D1, and cyclooxygenase-2 (COX-2). B, coronarin D inhibited the TNF-induced, NF-κB-regulated expression of antiapoptotic gene products inhibitor of apoptosis protein 1 (IAP1), TRAF-2, survivin, and Bcl-2. C, coronarin D suppressed the TNF-induced expression of metastatic gene products anti-intercellular adhesion molecule-1 (ICAM-1), MMP-9, and VEGF.

Close modal

NF-κB is key to the survival of tumor cells, as its activation induces the expression of antiapoptotic gene products. Because earlier experiments in this study showed that coronarin D inhibits TNF-induced NF-κB activation, we hypothesized that coronarin D would also inhibit the expression of TNF-induced antiapoptotic gene products, such as cellular inhibitor of apoptosis protein-1, TRAF-2, survivin, and Bcl-2, all known to be regulated by NF-κB (2631). Using Western blot analyses, we found that coronarin D did inhibit the expression of all these proteins (Fig. 4B).

Also using Western blot analysis, we determined that coronarin D inhibited the TNF-induced expression of three metastatic gene products, all of which have been shown elsewhere to be regulated by NF-κB: intercellular adhesion molecule-1, a cell surface adhesion factor (32); MMP-9, which is involved in tumor cell invasion and metastasis (33); and VEGF, the most potent angiogenic factor (ref. 34; Fig. 4C).

Coronarin D Potentiates Apoptosis Induced by TNF and Chemotherapeutic Agents

Because NF-κB activation can inhibit TNF expression and, as shown above, chemotherapeutic agents can induce apoptosis through the expression of the antiapoptotic gene products intercellular adhesion molecule-1, MMP-9, and VEGF (3537), we used several methods to measure different aspects of apoptosis to determine whether coronarin D enhances the apoptosis induced by TNF and other cytotoxic agents. Using a Live/Dead assay, which measures cell membrane permeability, we found that coronarin D enhanced TNF-induced cytotoxicity from 9% to 57% and Taxol-induced cytotoxicity from 12% to 76% (Fig. 5A). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and Annexin V staining also showed that TNF-induced apoptosis was enhanced by incubation with coronarin D (Fig. 5B and C). Using a caspase-3-activated poly(ADP-ribose) polymerase cleavage assay, we found that coronarin D had a dramatic effect on TNF-induced poly(ADP-ribose) polymerase cleavage (Fig. 5D). These results suggest that the apoptotic effects of TNF and Taxol (paclitaxel) are enhanced by coronarin D alone, at a dose at which coronarin D exhibits minimum cytotoxicity.

Figure 5.

Coronarin D enhanced the cytotoxicity induced by TNF and chemotherapeutic agents. A, Live/Dead assay results indicate that coronarin D up-regulated TNF-induced cytotoxicity. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF or 3 nmol/L Taxol (paclitaxel) for 24 h. Cells were stained with a Live/Dead assay reagent for 30 min and then analyzed with a fluorescence microscope as described in Materials and Methods. B, Annexin V staining shows that TNF-induced apoptosis was enhanced by incubation with coronarin D. Cells were pretreated with 50 μmol/L coronarin D for 8 h and then incubated with 1 nmol/L TNF for 16 h. Cells were incubated with anti-Annexin V antibodies conjugated with FITC and then analyzed with a flow cytometer to identify early apoptotic effects. C, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining shows that TNF-induced apoptosis was enhanced by incubation with coronarin D. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for 16 h. Cells were fixed, stained with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay reagent, and analyzed by flow cytometry as described in Materials and Methods. D, coronarin D potentiated TNF-induced poly(ADP-ribose) polymerase (PARP) cleavage. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for the indicated times. Whole-cell extracts were prepared, subjected to SDS-PAGE, and blotted with anti-poly(ADP-ribose) polymerase antibody.

Figure 5.

Coronarin D enhanced the cytotoxicity induced by TNF and chemotherapeutic agents. A, Live/Dead assay results indicate that coronarin D up-regulated TNF-induced cytotoxicity. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF or 3 nmol/L Taxol (paclitaxel) for 24 h. Cells were stained with a Live/Dead assay reagent for 30 min and then analyzed with a fluorescence microscope as described in Materials and Methods. B, Annexin V staining shows that TNF-induced apoptosis was enhanced by incubation with coronarin D. Cells were pretreated with 50 μmol/L coronarin D for 8 h and then incubated with 1 nmol/L TNF for 16 h. Cells were incubated with anti-Annexin V antibodies conjugated with FITC and then analyzed with a flow cytometer to identify early apoptotic effects. C, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining shows that TNF-induced apoptosis was enhanced by incubation with coronarin D. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for 16 h. Cells were fixed, stained with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay reagent, and analyzed by flow cytometry as described in Materials and Methods. D, coronarin D potentiated TNF-induced poly(ADP-ribose) polymerase (PARP) cleavage. KBM-5 cells were preincubated with 50 μmol/L coronarin D for 8 h and then treated with 1 nmol/L TNF for the indicated times. Whole-cell extracts were prepared, subjected to SDS-PAGE, and blotted with anti-poly(ADP-ribose) polymerase antibody.

Close modal

Whether coronarin D can also potentiate the effect of chemotherapeutic agents was also examined. We found that coronarin D potentiated the cytotoxic effects of doxorubicin, gemcitabine, 5-fluorouracil, cisplatin, and docetaxel against different tumor cell types (Table 1).

Table 1.

Coronarin D potentiates the cytotoxic effects of different chemotherapeutic agents against different tumor cell types

CancerCell lineChemotherapeutic agentDrug (IC50)Drug + coronarin D (IC50)
Chronic myeloid leukemia KBM-5 Doxorubicin 58 nmol/L 28 nmol/L 
Myeloma U266 Doxorubicin 72 nmol/L 58 nmol/L 
Pancreatic cancer PANC-1 Gemcitabine 82 nmol/L 44 nmol/L 
Bladder cancer 253JBV Gemcitabine 76 nmol/L 41 nmol/L 
Lung cancer H1299 5-Fluorouracil 1.8 μmol/L 0.8 μmol/L 
Colon cancer HT29 5-Fluorouracil 2.0 μmol/L 0.9 μmol/L 
Head and neck OSC 19 Cisplatin 0.2 μg/mL 0.1 μg/mL 
Ovarian cancer SKOV3 Docetaxel 1.9 nmol/L 1.2 nmol/L 
Breast cancer MCF-7 Docetaxel 1.9 nmol/L 1.4 nmol/L 
CancerCell lineChemotherapeutic agentDrug (IC50)Drug + coronarin D (IC50)
Chronic myeloid leukemia KBM-5 Doxorubicin 58 nmol/L 28 nmol/L 
Myeloma U266 Doxorubicin 72 nmol/L 58 nmol/L 
Pancreatic cancer PANC-1 Gemcitabine 82 nmol/L 44 nmol/L 
Bladder cancer 253JBV Gemcitabine 76 nmol/L 41 nmol/L 
Lung cancer H1299 5-Fluorouracil 1.8 μmol/L 0.8 μmol/L 
Colon cancer HT29 5-Fluorouracil 2.0 μmol/L 0.9 μmol/L 
Head and neck OSC 19 Cisplatin 0.2 μg/mL 0.1 μg/mL 
Ovarian cancer SKOV3 Docetaxel 1.9 nmol/L 1.2 nmol/L 
Breast cancer MCF-7 Docetaxel 1.9 nmol/L 1.4 nmol/L 

NOTE: IC50 is defined as the dose required for 50% cytotoxicity.

Coronarin D Suppresses TNF-Induced Invasion Activity

MMPs have been found to provide critical assistance to tumor cells during metastasis, and because earlier experiments in this study showed that coronarin D suppresses TNF-induced MMP-9 expression, we sought to determine the effects of coronarin D on the TNF-induced MMP-9 invasion. After seeding H1299 cells in the top chamber of a Matrigel invasion chamber in the absence of serum and incubating them with TNF with or without coronarin D for 24 h, we found that coronarin D suppressed TNF-induced cell invasion (Fig. 6A).

Figure 6.

A, coronarin D suppressed TNF-induced invasion activity. H1299 cells were seeded on the top of a Matrigel invasion chamber overnight in the absence of serum, preincubated with 50 μmol/L coronarin D for 8 h, treated with 1 nmol/L TNF for 24 h in the presence of 1% serum, and then subjected to an invasion assay as described in Materials and Methods. B, coronarin D inhibited RANKL-induced osteoclastogenesis. RAW 264.7 cells were incubated alone or with RANKL (5 nmol/L), with or without 5 μmol/L coronarin D, for 5 d and stained for tartrate-resistant acid phosphatase (TRAP) expression. Multinucleated (three nuclei) osteoclasts were counted. Tartrate-resistant acid phosphatase-positive cells were photographed. Original magnification, ×100. C, coronarin D is more active than coronarin D acid in inhibiting TNF-induced NF-κB activity. KBM-5 (2 × 106) cells were preincubated with different concentrations of coronarin D for 8 h. They were then treated with 0.1 nmol/L TNF for the indicated times and analyzed for NF-κB activation by EMSA. D, active chemical group of coronarin D and coronarin D acid.

Figure 6.

A, coronarin D suppressed TNF-induced invasion activity. H1299 cells were seeded on the top of a Matrigel invasion chamber overnight in the absence of serum, preincubated with 50 μmol/L coronarin D for 8 h, treated with 1 nmol/L TNF for 24 h in the presence of 1% serum, and then subjected to an invasion assay as described in Materials and Methods. B, coronarin D inhibited RANKL-induced osteoclastogenesis. RAW 264.7 cells were incubated alone or with RANKL (5 nmol/L), with or without 5 μmol/L coronarin D, for 5 d and stained for tartrate-resistant acid phosphatase (TRAP) expression. Multinucleated (three nuclei) osteoclasts were counted. Tartrate-resistant acid phosphatase-positive cells were photographed. Original magnification, ×100. C, coronarin D is more active than coronarin D acid in inhibiting TNF-induced NF-κB activity. KBM-5 (2 × 106) cells were preincubated with different concentrations of coronarin D for 8 h. They were then treated with 0.1 nmol/L TNF for the indicated times and analyzed for NF-κB activation by EMSA. D, active chemical group of coronarin D and coronarin D acid.

Close modal

Coronarin D Inhibits RANKL-Induced Osteoclastogenesis

Given that the agents that suppress RANKL signaling may inhibit bone resorption or osteoclastogenesis (38), we examined the effect of coronarin D on osteoclastogenesis. Toward that end, we treated RAW 264.7 cells with 5 μmol/mL coronarin D in the presence of RANKL. Cells were then allowed to grow and differentiate into osteoclasts. Using osteoclast differentiation assays, we found that coronarin D substantially decreased RANKL-induced osteoclast differentiation (Fig. 6B). In fact, a 5 μmol/L concentration of coronarin D was sufficient to reduce osteoclastogenesis by more than 70%. Under these conditions, the RAW 264.7 cells remained fully viable (data not shown).

We also compared the activity of coronarin D with its acidic form (Fig. 6C) and found that coronarin D acid also suppressed TNF-induced NF-κB activation but was less active (Fig. 6D).

The present study reports for the first time that coronarin D inhibits NF-κB. Specifically, coronarin D inhibited the NF-κB activation induced by different carcinogens and proinflammatory molecules and inhibited constitutive NF-κB expression. It also inhibited NF-κB by down-regulating IKK activation, which led to the inhibition of IκBα phosphorylation and degradation, p65 phosphorylation, and translocation. Coronarin D also inhibited NF-κB-regulated gene products that are involved in antiapoptotic activity, proliferation, invasion, and angiogenesis. Coronarin D also potentiated TNF- and Taxol-induced apoptosis. The coronarin D-modulated inhibition of NF-κB activity also led to the inhibition of TNF-induced invasion and RANKL-induced osteoclastogenesis.

Our results also showed that coronarin D suppressed the NF-κB activation induced by inflammatory stimuli, such as TNF, interleukin-1β, and LPS; prooxidants, such as H2O2; and carcinogens, such as okadaic acid, tumor promoters, and cigarette smoke condensate. These results suggest that coronarin D acts at a step that is common to all these agents. We also found that coronarin D inhibited NF-κB activation by inhibiting IKK activation, which led to the suppression of IκBα phosphorylation, p65 phosphorylation, and nuclear translocation. However, the mechanism by which coronarin D inhibits IKK activation is still unclear, as it requires an understanding of how IKK is activated, which itself has not been established. Over a dozen different kinases have been linked with IKK activation (39). One of these, TAK1, has been linked with IKK activation through TNF (40, 41). It is possible that coronarin D inhibits IKK through the inhibition of TAK1. However, we found that coronarin D directly inhibits IKK.

Coronarin D also inhibits the constitutive NF-κB activation commonly seen in a wide variety of tumors (4248). Although the reason NF-κB is constitutively active in tumor cells is unknown, various mechanisms have been implicated. These include the constitutive expression of TNF (36, 49) or interleukin-1 (33), increased proteasome activity (50), overexpression of the epidermal growth factor receptor (51), mutation of IκBα (52), and mutation of ras (53). Although most agents activate NF-κB through IKK-dependent mechanisms, some function through IKK-independent mechanisms (54, 55). Further investigation is required to determine whether coronarin D activates NF-κB in various tumor cells through IKK-dependent or IKK-independent mechanisms.

Coronarin D also inhibits the TNF-induced expression of gene products, such as c-myc, cyclin D1, and cyclooxygenase-2, which are essential for cellular proliferation and survival (5658). Expression of the c-myc oncoprotein prevents cell cycle arrest in response to growth-inhibitory signals, differentiation stimuli, and mitogen withdrawal. Moreover, myc activation in quiescent cells is sufficient to induce cell cycle entry in the absence of growth factors. Cyclin D1 exercises powerful control over the mechanisms that regulate the mitotic cell cycle. The inhibition of the expression of these three genes by coronarin D could account for the growth-inhibitory effects linked with certain coronarins (9). The cytotoxic effects of coronarin A reported previously against tumor cells (10) may involve suppression of NF-κB pathway. Moreover, coronarin D potentiated the cytotoxic effects of different chemotherapeutic agents in various tumor cell types. Besides tumor cells, NF-κB has been shown to be active in cancer stem cells. Thus, coronarin D is also likely to enhance the effect of chemotherapeutic agents against cancer stem cell.

Our finding that coronarin D also inhibits the TNF-induced invasion of H1299 cells suggests that coronarin D blocks not only primary tumor development but also malignant progression. The suppression of TNF-induced MMP-9 activity, which plays a key role in invasion, could account for the anti-invasive activity of coronarin D. The suppression of VEGF and intercellular adhesion molecule-1 could also contribute to the anti-invasive activity associated with coronarin D. Our results therefore imply that coronarin D should also exhibit antimetastatic activity in vivo.

RANKL has been shown to play a critical role in osteoclastogenesis, the loss of bone commonly associated with aging and cancer. The results of our study revealed that coronarin D inhibits RANKL-induced osteoclastogenesis, an effect that is most likely mediated through the suppression of NF-κB, as were many of the other effects associated with coronarin D to have.

Our results also show that coronarin D is more active than coronarin D acid. Unlike coronarin D acid, coronarin D can open to give an aldehyde group. The presence of this aldehyde group in coronarin D makes its more active than coronarin D acid.

Previously, it has been shown that coronarin D and E can inhibit nitric oxide production in LPS-induced mouse peritoneal macrophages (9). It is possible that this effect of coronarin D is mediated through suppression of NF-κB-mediated inducible nitric oxide synthase expression as described here. Previous studies have shown that coronarin A inhibits the proliferation of human umbilical vein endothelial cells (6) and acetic acid-induced vascular permeability in mice (8). It is possible that these effects are mediated through suppression of NF-κB-mediated VEGF expression as reported here.

Overall, our results suggest that coronarin D exhibits antiproliferative, proapoptotic, anti-invasive, antiangiogenic, antiosteoclastogenic, and anti-inflammatory effects through the suppression of NF-κB and NF-κB-regulated gene products. However, in vivo studies are needed to validate these findings.

No potential conflicts of interest were disclosed.

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.

We thank Angelique L. Geehan for carefully editing the article.

1
The wealth of India. A dictionary of Indian raw materials and industrial products. Vol. V: H-K. New Delhi: CSIR 1959. p. 11.
2
Shrotriya S, Ali MS, Saha A, Bachar S, Islam MS. Anti-inflammatory and analgesic effects of Hedychium coronarium Koen.
Pak J Pharm Sci
2007
;
20
:
47
–51.
3
Ribeiro Rde A, de Barros F, de Melo MM, et al. Acute diuretic effects in conscious rats produced by some medicinal plants used in the state of Sao Paulo, Brasil.
J Ethnopharmacol
1988
;
24
:
19
–29.
4
Itokawa H, Morita H, Katou I, et al. Cytotoxic diterpenes from the rhizomes of Hedychium coronarium.
Planta Med
1987
;
54
:
311
.
5
Itokawa H, Morita H, Takeya K, Motidome M. Diterpenes from rhizomes of Hedychium coronarium.
Chem Pharm Bull Tokyo
1988
;
36
:
2682
–4.
6
Oh S, Jeong IH, Shin WS, Lee SA. Study on the synthesis of antiangiogenic (+)-coronarin A and congeners from (+)-sclareolide.
Bioorg Med Chem Lett
2003
;
13
:
2009
–12.
7
Morikawa T, Matsuda H, Sakamoto Y, Ueda K, Yoshikawa M. New farnesane-type sesquiterpenes, hedychiols A and B 8,9-diacetate, and inhibitors of degranulation in RBL-2H3 cells from the rhizome of Hedychium coronarium.
Chem Pharm Bull Tokyo
2002
;
50
:
1045
–9.
8
Matsuda H, Morikawa T, Sakamoto Y, Toguchida I, Yoshikawa M. Labdane-type diterpenes with inhibitory effects on increase in vascular permeability and nitric oxide production from Hedychium coronarium.
Bioorg Med Chem
2002
;
10
:
2527
–34.
9
Itokawa H, Morita H, Katou I, et al. Cytotoxic diterpenes from the rhizomes of Hedychium coronarium.
Planta Med
1988
;
54
:
311
–5.
10
Aggarwal BB. Nuclear factor-κB: the enemy within.
Cancer Cell
2004
;
6
:
203
–8.
11
Anto RJ, Mukhopadhyay A, Shishodia S, Gairola CG, Aggarwal BB. Cigarette smoke condensate activates nuclear transcription factor-κB through phosphorylation and degradation of IκB(α): correlation with induction of cyclooxygenase-2.
Carcinogenesis
2002
;
23
:
1511
–8.
12
Chaturvedi MM, Mukhopadhyay A, Aggarwal BB. Assay for redox-sensitive transcription factor.
Methods Enzymol
2000
;
319
:
585
–602.
13
Kunnumakkara AB, Nair AS, Ahn KS, et al. Gossypin, a pentahydroxy glucosyl flavone, inhibits the transforming growth factor β-activated kinase-1-mediated NF-κB activation pathway, leading to potentiation of apoptosis, suppression of invasion, and abrogation of osteoclastogenesis.
Blood
2007
;
109
:
5112
–21.
14
Darnay BG, Ni J, Moore PA, Aggarwal BB. Activation of NF-κB by RANK requires tumor necrosis factor receptor-associated factor (TRAF) 6 and NF-κB-inducing kinase. Identification of a novel TRAF6 interaction motif.
J Biol Chem
1999
;
274
:
7724
–31.
15
Ichikawa H, Takada Y, Murakami A, Aggarwal BB. Identification of a novel blocker of IκBα kinase that enhances cellular apoptosis and inhibits cellular invasion through suppression of NF-κB-regulated gene products.
J Immunol
2005
;
174
:
7383
–92.
16
Bharti AC, Shishodia S, Reuben JM, et al. Nuclear factor-κB and STAT3 are constitutively active in CD138+ cells derived from multiple myeloma patients, and suppression of these transcription factors leads to apoptosis.
Blood
2004
;
103
:
3175
–84.
17
Takada Y, Mukhopadhyay A, Kundu GC, Mahabeleshwar GH, Singh S, Aggarwal BB. Hydrogen peroxide activates NF-κB through tyrosine phosphorylation of IκBα and serine phosphorylation of p65: evidence for the involvement of IκBα kinase and Syk protein-tyrosine kinase.
J Biol Chem
2003
;
278
:
24233
–41.
18
Garg A, Aggarwal BB. Nuclear transcription factor-κB as a target for cancer drug development.
Leukemia
2002
;
16
:
1053
–68.
19
Pahl HL. Activators and target genes of Rel/NF-κB transcription factors.
Oncogene
1999
;
18
:
6853
–66.
20
Shishodia S, Aggarwal BB. Nuclear factor-κB: a friend or a foe in cancer?
Biochem Pharmacol
2004
;
68
:
1071
–80.
21
Campbell KJ, Chapman NR, Perkins ND. UV stimulation induces nuclear factor κB (NF-κB) DNA-binding activity but not transcriptional activation.
Biochem Soc Trans
2001
;
29
:
688
–91.
22
Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword.
Nat Rev Immunol
2003
;
3
:
745
–56.
23
Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS, Jr. NF-κB controls cell growth and differentiation through transcriptional regulation of cyclin D1.
Mol Cell Biol
1999
;
19
:
5785
–99.
24
Yamamoto K, Arakawa T, Ueda N, Yamamoto S. Transcriptional roles of nuclear factor κB and nuclear factor-interleukin-6 in the tumor necrosis factor α-dependent induction of cyclooxygenase-2 in MC3T3-1 cells.
J Biol Chem
1995
;
270
:
31315
–20.
25
Duyao MP, Kessler DJ, Spicer DB, et al. Transactivation of the c-myc promoter by human T cell leukemia virus type 1 tax is mediated by NFκB.
J Biol Chem
1992
;
267
:
16288
–91.
26
Zhu L, Fukuda S, Cordis G, Das DK, Maulik N. Anti-apoptotic protein survivin plays a significant role in tubular morphogenesis of human coronary arteriolar endothelial cells by hypoxic preconditioning.
FEBS Lett
2001
;
508
:
369
–74.
27
Tamatani M, Che YH, Matsuzaki H, et al. Tumor necrosis factor induces Bcl-2 and Bcl-x expression through NFκB activation in primary hippocampal neurons.
J Biol Chem
1999
;
274
:
8531
–8.
28
Xiao CW, Ash K, Tsang BK. Nuclear factor-κB-mediated X-linked inhibitor of apoptosis protein expression prevents rat granulosa cells from tumor necrosis factor α-induced apoptosis.
Endocrinology
2001
;
142
:
557
–63.
29
Catz SD, Johnson JL. Transcriptional regulation of bcl-2 by nuclear factor κB and its significance in prostate cancer.
Oncogene
2001
;
20
:
7342
–51.
30
You M, Ku PT, Hrdlickova R, Bose HR, Jr. ch-IAP1, a member of the inhibitor-of-apoptosis protein family, is a mediator of the antiapoptotic activity of the v-Rel oncoprotein.
Mol Cell Biol
1997
;
17
:
7328
–41.
31
Schwenzer R, Siemienski K, Liptay S, et al. The human tumor necrosis factor (TNF) receptor-associated factor 1 gene (TRAF1) is up-regulated by cytokines of the TNF ligand family and modulates TNF-induced activation of NF-κB and c-Jun N-terminal kinase.
J Biol Chem
1999
;
274
:
19368
–74.
32
Ledebur HC, Parks TP. Transcriptional regulation of the intercellular adhesion molecule-1 gene by inflammatory cytokines in human endothelial cells. Essential roles of a variant NF-κB site and p65 homodimers.
J Biol Chem
1995
;
270
:
933
–43.
33
Esteve PO, Chicoine E, Robledo O, et al. Protein kinase C-ζ regulates transcription of the matrix metalloproteinase-9 gene induced by IL-1 and TNF-α in glioma cells via NF-κB.
J Biol Chem
2002
;
277
:
35150
–5.
34
Kim I, Moon SO, Kim SH, et al. Vascular endothelial growth factor expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin through nuclear factor-κB activation in endothelial cells.
J Biol Chem
2001
;
276
:
7614
–20.
35
Mayo MW, Wang CY, Cogswell PC, et al. Requirement of NF-κB activation to suppress p53-independent apoptosis induced by oncogenic Ras.
Science
1997
;
278
:
1812
–5.
36
Giri DK, Aggarwal BB. Constitutive activation of NF-κB causes resistance to apoptosis in human cutaneous T cell lymphoma HuT-78 cells. Autocrine role of tumor necrosis factor and reactive oxygen intermediates.
J Biol Chem
1998
;
273
:
14008
–14.
37
Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS, Jr. NF-κB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
Science
1998
;
281
:
1680
–3.
38
Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis.
Nature
1999
;
397
:
315
–23.
39
Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-κB activity.
Annu Rev Immunol
2000
;
18
:
621
–63.
40
Sakurai H, Miyoshi H, Toriumi W, Sugita T. Functional interactions of transforming growth factor β-activated kinase 1 with IκB kinases to stimulate NF-κB activation.
J Biol Chem
1999
;
274
:
10641
–8.
41
Takaesu G, Surabhi RM, Park KJ, Ninomiya-Tsuji J, Matsumoto K, Gaynor RB. TAK1 is critical for IκB kinase-mediated activation of the NF-κB pathway.
J Mol Biol
2003
;
326
:
105
–15.
42
Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ. The nuclear factor-κB RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells.
Clin Cancer Res
1999
;
5
:
119
–27.
43
Suh J, Payvandi F, Edelstein LC, et al. Mechanisms of constitutive NF-κB activation in human prostate cancer cells.
Prostate
2002
;
52
:
183
–200.
44
Bhat-Nakshatri P, Sweeney CJ, Nakshatri H. Identification of signal transduction pathways involved in constitutive NF-κB activation in breast cancer cells.
Oncogene
2002
;
21
:
2066
–78.
45
Tai DI. Tsai SL, Chang YH, et al. Constitutive activation of nuclear factor κB in hepatocellular carcinoma.
Cancer
2000
;
89
:
2274
–81.
46
Mitsiades CS, Mitsiades N, Poulaki V, et al. Activation of NF-κB and upregulation of intracellular anti-apoptotic proteins via the IGF-1/Akt signaling in human multiple myeloma cells: therapeutic implications.
Oncogene
2002
;
21
:
5673
–83.
47
Kang S, Kim YB, Kim MH, et al. Polymorphism in the nuclear factor κ-B binding promoter region of cyclooxygenase-2 is associated with an increased risk of bladder cancer.
Cancer Lett
2005
;
217
:
11
–6.
48
Hardwick JC, van den Brink GR, Offerhaus GJ, van Deventer SJ, Peppelenbosch MP. NF-κB, p38 MAPK and JNK are highly expressed and active in the stroma of human colonic adenomatous polyps.
Oncogene
2001
;
20
:
819
–27.
49
Jackson-Bernitsas DG, Ichikawa H, Takada Y, et al. Evidence that TNF-TNFR1-TRADD-TRAF2-RIP-TAK1-IKK pathway mediates constitutive NF-κB activation and proliferation in human head and neck squamous cell carcinoma.
Oncogene
2007
;
10
:
1385
–97.
50
Miyamoto S, Chiao PJ, Verma IM. Enhanced IκBα degradation is responsible for constitutive NF-κB activity in mature murine B-cell lines.
Mol Cell Biol
1994
;
5
:
3276
–82.
51
Sethi G, Ahn KS, Chaturvedi MM, Aggarwal BB. Epidermal growth factor (EGF) activates nuclear factor-κB through IκBα kinase-independent but EGF receptor-kinase dependent tyrosine 42 phosphorylation of IκBα.
Oncogene
2007
;
26
:
7324
–32.
52
Wood KM, Roff M, Hay RT. Defective IκBα in Hodgkin cell lines with constitutively active NF-κB.
Oncogene
1998
;
16
:
2131
–9.
53
Finco TS, Westwick JK, Norris JL, Beg AA, Der CJ, Baldwin AS, Jr. Oncogenic Ha-Ras-induced signaling activates NF-κB transcriptional activity, which is required for cellular transformation.
J Biol Chem
1997
;
272
:
24113
–24116.
54
Cammarano MS, Minden A. Dbl and the Rho GTPases activate NFκB by I κB kinase (IKK)-dependent and IKK-independent pathways.
J Biol Chem
2001
;
276
:
25876
–82.
55
Lou H, Kaplowitz N. Glutathione depletion down-regulates tumor necrosis factor α-induced NF-κB activity via IκB kinase-dependent and -independent mechanisms.
J Biol Chem
2007
;
282
:
29470
–81.
56
Romano M, Claria J. Cyclooxygenase-2 and 5-lipoxygenase converging functions on cell proliferation and tumor angiogenesis: implications for cancer therapy.
FASEB J
2003
;
17
:
1986
–95.
57
Hurlin PJ, Dezfouli S. Functions of myc:max in the control of cell proliferation and tumorigenesis.
Int Rev Cytol
2004
;
238
:
183
–226.
58
Erickson LA, Jin L, Goellner JR, et al. Pathologic features, proliferative activity, and cyclin D1 expression in Hurthle cell neoplasms of the thyroid.
Mod Pathol
2000
;
13
:
186
–92.

Supplementary data