We have identified a natural compound that activates apoptosis of epithelial cancer cells through activation of tumor necrosis factor-α (TNF-α), TNF receptor (TNFR)-associated death domain (TRADD), and caspases. The molecule 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde (HDNC, marmelin) was isolated and characterized from ethyl acetate fraction of extracts of Aegle marmelos. HDNC treatment inhibited the growth of HCT-116 colon cancer tumor xenografts in vivo. Immunostaining for CD31 showed that there was a significant reduction in microvessels in the HDNC-treated animals, coupled with decreased cyclooxygenase-2, interleukin-8, and vascular endothelial growth factor mRNA. Using hexoseaminidase assay, we determined that HDNC inhibits proliferation of HCT-116 colon and HEp-2 alveolar epithelial carcinoma cells. Furthermore, the cancer cells showed increased levels of activated caspase-3 and induced G1 cell cycle arrest, which was suppressed by caspase-3 inhibitors. HDNC induced TNF-α, TNFR1, and TRADD mRNA and protein expression. Moreover, caspase-8 and Bid activation, and cytochrome c release, were observed, suggesting the existence of a cross-talk between death receptor and the mitochondrial pathways. HDNC inhibited AKT and extracellular signal-regulated kinase phosphorylation both in cells in culture and in tumor xenografts. In addition, electrophoretic mobility shift assay and luciferase reporter assays showed that HDNC significantly suppressed TNF-α–mediated activation and translocation of nuclear factor-κB (NF-κB). This was further confirmed by Western blot analysis of nuclear extracts wherein levels of RelA, the p65 component of NF-κB, were significantly less in cells treated with HDNC. Together, the data suggest that the novel compound HDNC (marmelin) is a potent anticancer agent that induces apoptosis during G1 phase of the cell cycle and could be a potential chemotherapeutic candidate. [Cancer Res 2008;68(20):8573–81]

Apoptosis is a genetically regulated form of cell death, in which abnormal cells are eliminated from an organism, ensuring normal development of multicellular organisms and maintenance of tissue homeostasis (1). Therefore, an understanding of the molecular mechanisms of apoptosis is not only of scientific importance but may also have therapeutic value. A battery of signaling cascades is involved in the ability of the cell to undergo apoptosis. One such is a family of cysteine proteases, caspases, whose function in apoptotic cell death has been studied in detail (2) and are important mediators of apoptotic processes caused by various inducers (3). They amplify the apoptotic signal and proteolytically process numerous cellular target molecules with different functions (4).

There are two major apoptotic pathways in mammalian cells (5, 6). The extrinsic pathway is initiated by the binding of transmembrane death receptors [e.g., Fas, tumor necrosis factor receptor 1 (TNFR1), and TNF-related apoptosis-inducing ligand (TRAIL) receptors] with cognate extracellular ligands (7). Ligand receptors recruit adaptor proteins [e.g., TNFR-associated death domain (TRADD) and FADD], which interact with and trigger the activation of caspase-8. Activated caspase-8 then cleaves and activates downstream effector caspases, such as caspase-3. In contrast, the intrinsic pathway is characterized by disruption of mitochondrial membrane integrity when cells are exposed to various stresses (e.g., DNA-damaging agents; ref. 8). Mitochondrial membrane integrity triggers apoptosis via both caspase-dependent (e.g., the cytochrome c–caspase-9 pathway) and caspase-independent (e.g., the apoptosis-inducing factor pathway) mechanisms. Cross-talk exists between the extrinsic and intrinsic pathways, as activated caspase-8 can cleave Bid to produce truncated Bid (t-Bid), which then binds to mitochondria and promotes mitochondrial membrane integrity (9, 10). The subsequent release of cytochrome c from mitochondria further facilitates the apoptotic process (1113).

The importance of cell signaling cascades in normal cell regulation and during disease condition has been realized (1416), and many well-known targets at the signaling levels have been identified that are critical for the ability of cancer cells to proliferate rapidly. Furthermore, the availability of high-throughput assays based on the above-said molecular targets has further enhanced the process of drug discovery (17). Most anticancer drugs are natural products or their semisynthetic analogues; well known examples include Taxol, epothilone, camptothecin, daunomycin, and Vinca alkaloids (18, 19). Some of the plant-derived anticancer compounds are beginning to enter clinical trials (19).

Here, we report the identification of a novel compound, 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde (HDNC), from Aegle marmelos, a medicinal plant from India used for treatment of cancer-related symptoms in the Indian system of medicine (20). As a common term, we have named it marmelin. HDNC inhibits the growth of HCT-116 colon cancer cell tumor xenografts and angiogenesis in nude mice. In addition, HDNC suppresses cancer cell proliferation while inducing apoptosis through the induction of activation of TNF-α, TNFR1, TRADD, caspase-8, and t-Bid, resulting in cytochrome c release and activation of caspase-3.

Cells and reagents. HCT-116, HT-29 colon, AGS gastric, and HEp-2 alveolar cancer cells obtained from the American Type Culture Collection were grown in DMEM and RPMI 1640 containing 10% heat-inactivated fetal bovine serum (Sigma Chemical Co.) and 1% antibiotic-antimycotic solution (Mediatech, Inc.) at 37°C in a humidified atmosphere of 5% CO2. The BD RiboQuant multiprobe template set for hCK-3 and hAPO-3c was obtained from BD PharMingen. Caspase-3 Inhibitor IV (Ac-DMQD-CHO) was obtained from EMD Biosciences. Antibodies were obtained from Cell Signaling. The medicinal plant A. marmelos was obtained from south India and the species was authenticated (21, 22).

HCT-116 cell xenograft tumors in mice. Five-week-old male athymic nude mice (The Jackson Laboratory) were maintained with water and standard mouse chow ad libidum and used in protocols approved by the Animal Studies Committee of the University. Animals were injected with 1 × 106 HCT-116 cells in the left and right flank with 100 μL Matrigel and allowed to form xenograft. Marmelin (HDNC, 200 μg/kg body weight) in 5% Na2HCO3 buffer alone was administered i.p. daily for 23 d. Tumors were measured weekly with a Vernier caliper and tumor volumes were calculated according to the formula length × width × depth × 0.5236. At the end of treatment, the animals were sacrificed, and the tumors were removed and weighed for use in histology (H&E and CD31) and gene expression studies.

Proliferation and apoptosis assays. Cells were grown in 96-well plates and treated with increasing doses of extracts or purified HDNC, and proliferation was assessed by hexoseaminidase assay (23). The plates were read at 405 nm in a Synergy HT microtiter plate reader (Bio-Tek Instruments, Inc.). For apoptosis, caspase-3/7 activity was measured using the Apo-ONE Homogeneous Caspase-3/7 Assay kit (Promega Corp.).

Cell cycle analysis. Cells were plated at a density of 5 × 105 per well on six-well plates. After treatment for 24 h, cells were collected into flow cytometry tubes and stained with propidium iodide at 4°C for 30 min in the dark. Cell cycle analysis was performed with a Becton Dickinson FACScan using an FL2 detector with a bandpass filter at specifications of 585 ± 21 nm. In each analysis, 10,000 events were recorded.

RNA preparation and RNase protection assay. RNA was isolated from the cells using Trizol method (Invitrogen); 20 μg/lane were loaded for RNase protection assay (RPA) using radiolabeled RiboQuant multiprobe template set for human cytokines (hCK-3) or human apoptosis-related genes (hAPO-3c), according to the manufacturer's protocol.

Immunoblot analysis. Total lysates from cells or from tumor xenografts and nuclear extracts were prepared and separated on 10% SDS-polyacrylamide gel and blotted onto Immobilon polyvinylidene difluoride membranes (Millipore). Membranes were incubated overnight with the indicated primary antibody followed by secondary antibody (1:5,000) for 1 h. Specific proteins were detected by the enhanced chemiluminescence system (Amersham Pharmacia Biotech).

ELISA assay. TNF-α levels in the medium were determined by sandwich ELISA assays, using ELISA kits, according to the manufacturer's protocol (Pierce Biotechnology). The TNF-α ELISA kit has a detection range of 15.6 to 1,000 pg/mL and has a sensitivity of <2 pg/mL. Briefly, the samples are incubated with the precoated wells followed by biotinylated second antibody. The plates were subsequently incubated with streptavidin-horseradish peroxidase reagent and 3,3′,5,5′-tetramethylbenzidine substrate solution. The absorbance was read at 450 and 550 nm using the Synergy HT plate reader.

Electrophoretic mobility shift assay analysis. HCT-116 cells were pretreated with the HDNC for 2 h before coincubation with TNF-α (10 ng/mL) for 1 h. Nuclear extracts were prepared using the NE-PER kit (Pierce Biotechnology). Protein concentrations were determined using bicinchoninic acid reagent (Pierce Biotechnology). Nuclear extracts (6 μg) were incubated with 32P-labeled double-stranded oligonucleotide containing a consensus NF-κB site (5′-AGTTGAGGGGACTTTCCCAGGC-3′; binding site in bold and underlined) at room temperature for 30 min. The protein-DNA complexes were size separated in a 4% native PAGE in 0.5% Tris-borate EDTA buffer and subjected to autoradiography.

Luciferase assay. HCT-116 cells were seeded in six-well dishes and incubated until 80% confluent. Then, the cells were transfected with 3 μg pNFκB-Luc (Clontech) reporter plasmid using Fugene 6 reagent (Roche Diagnostic). After 4 h of incubation, the cells were treated with extracts or purified HDNC in the presence or absence of TNF-α for 24 h. Cells were lysed and luciferase levels were determined using Luciferase Assay System (Promega).

Statistical analysis. All values are expressed as the mean ± SE. Data were analyzed using a paired two-tailed t test. A P value of <0.05 was considered statistically significant.

HDNC inhibits cancer cell proliferation. Recent studies have shown that administration of hydroalcoholic extract of A. marmelos to Ehrlich ascites carcinoma-bearing Swiss albino mice resulted in a dose-dependent remission of the tumor (21). Furthermore, a recent study showed no toxicity in rats administered extracts of leaves from this plant (22). These data, taken together, suggest that the plant encodes antitumor activity with a low toxicity profile. Accordingly, we aimed to identify an active compound in A. marmelos that is a potent inhibitor of solid tumors. Powdered A. marmelos plant material was extracted with ethanol and fractionated into hexane, dichloromethane, ethyl acetate, and n-butanol soluble fractions (data not shown). The effect of the solvent fractions on the proliferation of cancer cells was evaluated for up to 72 h. Dose-response analysis revealed 10 μg/mL of ethyl acetate fraction (Fig. 1A). The active ethyl acetate fraction was subsequently fractionated by silica column chromatography using a combination of hexane and ethyl acetate solvents. According to their TLC profiles, they were pooled into 12 fractions (data not shown). Fraction 5 showed maximum activity in 24 h. This compound was characterized as HDNC with a molecular mass of 232.23 (Fig. 1B) by nuclear magnetic resonance (NMR) and mass spectrometry (MS) analysis. We have also given the compound a common name marmelin because it was isolated from A. marmelos. Dose-response analysis revealed significant reduction in cell proliferation of multiple cancer cells at a dose of 50 μmol/L HDNC (Fig. 1A; Supplementary Fig. S1). In contrast, HDNC did not inhibit proliferation of normal mouse embryo fibroblasts for doses up to 500 μmol/L (data not shown). Together, these data show that the purified HDNC is a potent antiproliferative molecule with activity against epithelial cancer cells of different origin.

Figure 1.

HDNC inhibits growth of cancer cells. A, dose-response analysis of HEp-2 cells to A. marmelos ethyl acetate fraction and purified HDNC. HEp-2 cells were incubated with increasing concentrations of the ethyl acetate fraction of A. marmelos extracts (0–100 μg/mL) or HDNC (0–500 μmol/L) for 24, 48, and 72 h and cell proliferation was determined using hexoseaminidase activity. The graph is plotted as percent inhibition when compared with control, buffer-treated cells. HDNC treatment resulted in a significant dose-dependent decrease in cell number in the cells when compared with controls. Results are representative of three independent experiments. B, topological structure of HDNC. The structure of the active molecule was determined as HDNC. C, HDNC treatment induces apoptosis in HEp-2 cells. Top, cells incubated with increasing concentrations of HDNC or curcumin (0–10 μmol/L) were analyzed for apoptosis. HDNC treatment increased number of apoptotic cells compared with either curcumin-treated or untreated control. Results are representative of three independent experiments. *, P < 0.001. Bottom, HDNC induces caspase-3, an apoptosis mediator. Lysates from HEp-2 cells were incubated with 10 μg/mL ethyl acetate fraction of A. marmelos extract or 25 μmol/L HDNC and were analyzed by Western blotting for caspase-3 protein expression levels using rabbit anti-caspase-3 antibody. HDNC-treated cells show cleaved (activated) caspase-3, whereas untreated or buffer-treated cells have no cleaved caspase-3. D, DNA content (propidium iodide) and cell cycle analysis of HDNC-treated cells. HEp-2 cells were treated with the ethyl acetate fraction (10 μg/mL) or HDNC (25 μmol/L) for 36 h. M1, sub-G1-G0 peak. Apoptosis was measured as the percentage of cells containing hypodiploid amounts of DNA (sub-G1-G0 peak). HDNC treatment leads to increased number of cells in the sub-G1-G0 phase as indicated by the increase in M1 peak compared with control. However, this is reduced when cells are also treated with the caspase-3 inhibitor AC-DMQD-CHO. Graphs are representative of data collected from at least three experiments.

Figure 1.

HDNC inhibits growth of cancer cells. A, dose-response analysis of HEp-2 cells to A. marmelos ethyl acetate fraction and purified HDNC. HEp-2 cells were incubated with increasing concentrations of the ethyl acetate fraction of A. marmelos extracts (0–100 μg/mL) or HDNC (0–500 μmol/L) for 24, 48, and 72 h and cell proliferation was determined using hexoseaminidase activity. The graph is plotted as percent inhibition when compared with control, buffer-treated cells. HDNC treatment resulted in a significant dose-dependent decrease in cell number in the cells when compared with controls. Results are representative of three independent experiments. B, topological structure of HDNC. The structure of the active molecule was determined as HDNC. C, HDNC treatment induces apoptosis in HEp-2 cells. Top, cells incubated with increasing concentrations of HDNC or curcumin (0–10 μmol/L) were analyzed for apoptosis. HDNC treatment increased number of apoptotic cells compared with either curcumin-treated or untreated control. Results are representative of three independent experiments. *, P < 0.001. Bottom, HDNC induces caspase-3, an apoptosis mediator. Lysates from HEp-2 cells were incubated with 10 μg/mL ethyl acetate fraction of A. marmelos extract or 25 μmol/L HDNC and were analyzed by Western blotting for caspase-3 protein expression levels using rabbit anti-caspase-3 antibody. HDNC-treated cells show cleaved (activated) caspase-3, whereas untreated or buffer-treated cells have no cleaved caspase-3. D, DNA content (propidium iodide) and cell cycle analysis of HDNC-treated cells. HEp-2 cells were treated with the ethyl acetate fraction (10 μg/mL) or HDNC (25 μmol/L) for 36 h. M1, sub-G1-G0 peak. Apoptosis was measured as the percentage of cells containing hypodiploid amounts of DNA (sub-G1-G0 peak). HDNC treatment leads to increased number of cells in the sub-G1-G0 phase as indicated by the increase in M1 peak compared with control. However, this is reduced when cells are also treated with the caspase-3 inhibitor AC-DMQD-CHO. Graphs are representative of data collected from at least three experiments.

Close modal

Caspase-3 activation is essential for HDNC-mediated cell apoptosis. Caspase-3 is a key effector in the apoptosis pathway, amplifying the signal from initiator caspases, such as caspase-8 (24, 25). To determine whether cells undergo apoptosis, we first performed a caspase-3/7 assay after the cells were incubated with purified HDNC. There was a dose-dependent increase in caspase-3/7 activity, suggesting that the compound induces cells to undergo apoptosis (Fig. 1C,, top). To further determine whether the apoptotic activity is due to caspase-3, Western blot analysis was performed with extracts from HEp-2 cells. Significant induction in caspase-3 activation was observed in cells treated with either the ethyl acetate fraction or purified HDNC (Fig. 1C,, bottom). To confirm that caspase-3 activation is required for HDNC-mediated apoptosis, cell cycle was performed. A significant increase in cells in sub-G1 phase was observed when the cells were treated with either the ethyl acetate fraction (10 μg/mL) or purified HDNC (50 μmol/L; Fig. 1D). Furthermore, coincubation with a specific inhibitor of caspase-3, Ac-DMQD-CHO, resulted in a significant reduction in cells in sub-G1 phase. Similar results were obtained with HCT-116 and HT-29 cells (data not shown). These data suggest that HDNC-mediated induction of apoptosis is mediated by caspase-3 activation.

HDNC inhibits tumor growth. To evaluate the role of HDNC in tumor proliferation in vivo, we examined the ability of the compound to suppress the growth of human cancer cell xenografts in nude mice. Colon cancer cell–induced xenograft tumors were allowed to develop and grow to a size of 500 mm3, following which HDNC was administered i.p. for 3 weeks daily. HDNC significantly inhibits the growth of the tumor xenografts (Fig. 2A). Whereas the control tumors grew during the treatment period, reaching a size of 2,000 mm3, tumors in the HDNC-treated animals were smaller throughout. The excised tumors from the HDNC-treated animals averaged ∼1.3 g, whereas those from the control group weighed between 2.0 and 2.5 g (Fig. 2C). In addition, tumor volumes were significantly decreased (Fig. 2B and C). There was no apparent change in liver weight, spleen weight, or body weight in the wild-type animals treated with HDNC (data not shown). These data imply that HDNC is a potential therapeutic for treatment of colon cancers and that it is relatively nontoxic to mice.

Figure 2.

HDNC inhibits cancer tumor xenograft growth in vivo. A, HCT-116 cells were injected to the flanks of nude mice and palpable tumors were allowed to develop for 7 d. Subsequently, HDNC was injected daily i.p. for up to 21 d. On day 22, tumors were excised and subjected to further analyses. Tumor volumes in HDNC-administered mice were smaller than that of control mice. B, tumor size was measured every week. There was a significant reduction in tumor size from HDNC-treated animals when compared with untreated controls. C, HDNC treatment resulted in significantly lower tumor weight and volume when compared with controls. D, HDNC suppresses tumor angiogenesis. Immunohistochemistry shows that HDNC treatment significantly reduced the expression of CD31, an endothelial cell–specific surface marker in tumor xenografts. The proteins are stained brown. Microvessel density was significantly reduced in the xenografts of HDNC-treated animals.

Figure 2.

HDNC inhibits cancer tumor xenograft growth in vivo. A, HCT-116 cells were injected to the flanks of nude mice and palpable tumors were allowed to develop for 7 d. Subsequently, HDNC was injected daily i.p. for up to 21 d. On day 22, tumors were excised and subjected to further analyses. Tumor volumes in HDNC-administered mice were smaller than that of control mice. B, tumor size was measured every week. There was a significant reduction in tumor size from HDNC-treated animals when compared with untreated controls. C, HDNC treatment resulted in significantly lower tumor weight and volume when compared with controls. D, HDNC suppresses tumor angiogenesis. Immunohistochemistry shows that HDNC treatment significantly reduced the expression of CD31, an endothelial cell–specific surface marker in tumor xenografts. The proteins are stained brown. Microvessel density was significantly reduced in the xenografts of HDNC-treated animals.

Close modal

HDNC inhibits angiogenesis. A key feature of tumors is the development of microcapillaries. Given the reduction in the size of the tumors following HDNC treatment, we determined the effect of the compound on tumor vascularization by staining for the endothelial-specific antigen CD31. HDNC treatment leads to lower levels of CD31 staining and to the obliteration of the normal vasculature that is associated with tumor angiogenesis (Fig. 2D). Furthermore, HDNC treatment resulted in a significant reduction in microvessel density (Supplementary Fig. S2). Two key factors that regulate capillary growth in the tumors are vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8), and prostaglandins and the other tumors promoting mediators are known to induce their expression in epithelial cells. Prostaglandin production is regulated by cyclooxygenase-2 (COX-2), a protein that is overexpressed at multiple cancers (2628). Therefore, we next determined the effects of HDNC treatment on COX-2, VEGF, and IL-8 expression. HDNC treatment reduced COX-2, VEGF, and IL-8 mRNA levels in the tumor xenografts (Supplementary Fig. S3).

HDNC induces TNF-α, which exerts its activity through recruitment of TRADD. TNF-α is pleiotropic cytokine involved in inflammatory and apoptotic responses. To determine the effect of HDNC on TNF-α expression, we performed a RPA analysis. TNF-α levels were significantly up-regulated in HEp-2 cells following treatment with HDNC (Fig. 3A). Similarly, incubation of HCT-116 and AGS cells resulted in increased expression of TNF-α at the RNA and protein levels (Fig. 3B and C). Binding of TNF-α to its cognate receptor TNFR1 results in receptor trimerization and subsequent binding to an adapter molecule, TRADD. TRADD in turn recruits TNF-associated factor 2, and subsequent activation of the nuclear factor-κB (NF-κB) and c-Jun NH2-terminal kinase (JNK) pathways. In addition, activated TRADD has been shown to recruit a second adaptor (FADD), which in turn activates caspase-8. To determine whether these proteins are affected by HDNC, a RPA analysis with TNF signaling templates was performed. RPA analysis revealed the activation of TNFR1 (p55) receptor, TRADD, and Fas. Further, we also observed the activation of caspase-8 (Fig. 4A). Immunoblot analysis also showed activation of TRADD on treatment with both ethyl acetate fraction and HDNC compared with the untreated controls (Fig. 4B). Similarly, activation of caspase-8 was confirmed (Fig. 4B).

Figure 3.

HDNC induces TNF-α expression. A, RPA was performed to determine the expression of cytokines in HEp-2 cells following treatment for 6 h with A. marmelos ethyl acetate fraction (10 μg/mL) and pure HDNC (25 μmol/L). TNF-α is significantly induced in the cells. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. B, real-time PCR analysis of TNF-α expression. Total RNA from HCT-116 cells treated with ethyl acetate fraction and pure HDNC was subjected to reverse transcription followed by real-time PCR analyses for TNF-α expression. The data show that TNF-α is significantly induced following HDNC treatment. *, P = 0.003; **, P = 0.001; ***, P < 0.001; ****, P < 0.0001. C, ELISA assay for TNF-α expression in HCT-116 cells following treatment for 30 min with A. marmelos ethyl acetate fraction and pure HDNC. TNF-α is significantly induced in the cells. *, P = 0.003; **, P < 0.001.

Figure 3.

HDNC induces TNF-α expression. A, RPA was performed to determine the expression of cytokines in HEp-2 cells following treatment for 6 h with A. marmelos ethyl acetate fraction (10 μg/mL) and pure HDNC (25 μmol/L). TNF-α is significantly induced in the cells. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. B, real-time PCR analysis of TNF-α expression. Total RNA from HCT-116 cells treated with ethyl acetate fraction and pure HDNC was subjected to reverse transcription followed by real-time PCR analyses for TNF-α expression. The data show that TNF-α is significantly induced following HDNC treatment. *, P = 0.003; **, P = 0.001; ***, P < 0.001; ****, P < 0.0001. C, ELISA assay for TNF-α expression in HCT-116 cells following treatment for 30 min with A. marmelos ethyl acetate fraction and pure HDNC. TNF-α is significantly induced in the cells. *, P = 0.003; **, P < 0.001.

Close modal
Figure 4.

HDNC induces expression of death receptors. A, RPA analysis of total RNA from HEp-2 cells treated for 12 h with ethyl acetate fraction of A. marmelos extract (10 μg/mL) or purified HDNC (25 μmol/L). TNFR1, TRADD, and caspase-8 are induced at 12 h. B, Western blot analysis shows that cells treated with ethyl acetate fraction of A. marmelos extract (10 μg/mL) or purified HDNC (25 μmol/L) have higher levels of TRADD, caspase-8, cleaved Bid (t-Bid), and cytochrome c proteins.

Figure 4.

HDNC induces expression of death receptors. A, RPA analysis of total RNA from HEp-2 cells treated for 12 h with ethyl acetate fraction of A. marmelos extract (10 μg/mL) or purified HDNC (25 μmol/L). TNFR1, TRADD, and caspase-8 are induced at 12 h. B, Western blot analysis shows that cells treated with ethyl acetate fraction of A. marmelos extract (10 μg/mL) or purified HDNC (25 μmol/L) have higher levels of TRADD, caspase-8, cleaved Bid (t-Bid), and cytochrome c proteins.

Close modal

HDNC induces Bid cleavage leading to cytochrome c release. Activated caspase-8 can cleave Bid, a Bcl2-interacting protein, at the COOH terminus. The activated t-Bid subsequently translocates to the mitochondria where it triggers cytochrome c release (29). Because caspase-8 was induced following HDNC treatment, we next analyzed whether Bid is cleaved. Western blot analysis showed significant levels of t-Bid (Fig. 4B). Furthermore, activation of cytosolic cytochrome c was observed following HDNC treatment (Fig. 4B). This observation further strengthens the hypothesis that a cross-talk exists between intrinsic and extrinsic pathways in case of HDNC-induced apoptosis.

Suppression of AKT and extracellular signal-regulated kinase activation in colon cancer cells. AKT is a serine/threonine kinase involved in cell proliferation, survival/apoptosis inhibition, and angiogenesis (30, 31). Similarly, the p42/p44 extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) signaling pathway controls cell growth, differentiation, and survival (32). Because TNF-α modulates cellular responses through MAPK to induce NF-κB, we next determined whether HDNC-mediated induction of TNF-α resulted in increased AKT and/or ERK activity. Western blot analyses showed TNF-α–mediated increase in AKT and ERK phosphorylation (Fig. 5A and B). However, pretreatment with HDNC or ethyl acetate fraction inhibited TNF-α–mediated induction of AKT and ERK phosphorylation (Fig. 5A and B; Supplementary Fig. S4). Further confirmation that HDNC inhibits AKT and ERK phosphorylation was obtained when extracts of tumor xenografts were subjected to Western blot analyses (Fig. 5C and D; Supplementary Fig. S4). Taken together, these data suggest that HDNC is a potent suppressor of TNF-α–mediated signaling.

Figure 5.

HDNC suppressed AKT and ERK activation required for cell survival. A and B, HCT-116 cells were treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (125 nmol/L) in the presence or absence of TNF-α (10 ng/mL) and whole-cell lysates were prepared. Lysates from HDNC treatment showed significant reduction in the TNF-α–mediated AKT and ERK phosphorylation. C and D, AKT and ERK phosphorylation was significantly lower in tumor xenografts from HDNC-treated animals when compared with controls.

Figure 5.

HDNC suppressed AKT and ERK activation required for cell survival. A and B, HCT-116 cells were treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (125 nmol/L) in the presence or absence of TNF-α (10 ng/mL) and whole-cell lysates were prepared. Lysates from HDNC treatment showed significant reduction in the TNF-α–mediated AKT and ERK phosphorylation. C and D, AKT and ERK phosphorylation was significantly lower in tumor xenografts from HDNC-treated animals when compared with controls.

Close modal

Suppression of TNF-α–induced NF-κB activation in colon cancer cells. A key downstream target of TNF-α–mediated NF-κB activation is IL-8 (3335). To determine the effect of HDNC on TNF-α–mediated NF-κB activation, we first determined the expression of IL-8 in HCT-116 cells. Although HDNC did not affect baseline IL-8 expression, it inhibited TNF-α–mediated induction of IL-8 expression in the cells (Fig. 6A). To determine if this is due to loss of NF-κB activation, we performed an electrophoretic mobility shift assay (EMSA) analysis with nuclear extracts of HCT-116 cells. HDNC suppressed TNF-α–induced NF-κB activation (Fig. 6B). Further confirmation of this inhibition was shown by Western blot analysis for RelA, the p65 component of NF-κB. RelA levels were significantly reduced in the nucleus of cells treated with HDNC even in the presence of TNF-α (Fig. 6C,, left). This was even more evident when the expression was quantitated and compared with controls (Fig. 6C,, right). Finally, to show that HDNC inhibits NF-κB activation, we transiently transfected HCT-116 and HT-29 cells with a luciferase reporter gene plasmid under the control of a minimal TATA promoter fused to NF-κB response element. NF-κB transcriptional activity was attenuated in both cell lines treated with either the extract or HDNC when compared with the buffer-treated controls. Furthermore, they also inhibited TNF-α–induced NF-κB transcriptional activity (Fig. 6D). Because TNF-α–mediated inflammatory and pro-proliferative responses involve activation of NF-κB, these data suggest that HDNC enhances TNF-α–mediated apoptotic response in cancer cells by inhibiting the NF-κB activation pathway.

Figure 6.

HDNC suppresses TNF-α–induced NF-κB activation. A, IL-8 expression. Real-time reverse transcription-PCR analysis of total RNA from HCT-116 cells treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (125 nmol/L) in the presence or absence of TNF-α (10 ng/mL) for 24 h shows that IL-8 levels are induced by TNF-α, which is inhibited by both extract or purified molecule. B, EMSA. Nuclear extracts from the cells were subjected to EMSA analysis for the presence of NF-κB. The results reveal suppression of TNF-α–induced NF-κB activation by both ethyl acetate fraction and pure HDNC. C, Western blot analysis. Left, nuclear extracts were Western blotted for RelA, the 65-kDa component of the NF-κB transcription factor complex. As controls for loading, we also probed the membrane for β-actin and topoisomerase II. Right, results from three independent experiments show that both the ethyl acetate extracts and pure HDNC inhibit RelA translocation to the nucleus. D, NF-κB promoter analysis. HCT-116 and HT-29 cells were transfected with plasmid pNFκB-Luc (Mercury Pathway Profiling Luciferase System, Clontech) and subsequently treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (250 nmol/L) in the presence or absence of TNF-α (10 ng/mL). Lysates were prepared and tested for luciferase expression. TNF-α induced NF-κB–mediated luciferase activity, which is inhibited by both extract or purified molecule. *, P < 0.001, compared with TNF-α alone in HCT-116 cells; **, P < 0.001, compared with TNF-α alone in HT-29 cells. Data from three independent experiments.

Figure 6.

HDNC suppresses TNF-α–induced NF-κB activation. A, IL-8 expression. Real-time reverse transcription-PCR analysis of total RNA from HCT-116 cells treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (125 nmol/L) in the presence or absence of TNF-α (10 ng/mL) for 24 h shows that IL-8 levels are induced by TNF-α, which is inhibited by both extract or purified molecule. B, EMSA. Nuclear extracts from the cells were subjected to EMSA analysis for the presence of NF-κB. The results reveal suppression of TNF-α–induced NF-κB activation by both ethyl acetate fraction and pure HDNC. C, Western blot analysis. Left, nuclear extracts were Western blotted for RelA, the 65-kDa component of the NF-κB transcription factor complex. As controls for loading, we also probed the membrane for β-actin and topoisomerase II. Right, results from three independent experiments show that both the ethyl acetate extracts and pure HDNC inhibit RelA translocation to the nucleus. D, NF-κB promoter analysis. HCT-116 and HT-29 cells were transfected with plasmid pNFκB-Luc (Mercury Pathway Profiling Luciferase System, Clontech) and subsequently treated with ethyl acetate fraction (250 ng/mL) or purified HDNC (250 nmol/L) in the presence or absence of TNF-α (10 ng/mL). Lysates were prepared and tested for luciferase expression. TNF-α induced NF-κB–mediated luciferase activity, which is inhibited by both extract or purified molecule. *, P < 0.001, compared with TNF-α alone in HCT-116 cells; **, P < 0.001, compared with TNF-α alone in HT-29 cells. Data from three independent experiments.

Close modal

The ability of cancer cells to avert the apoptotic program has been identified as one of the major mechanisms for the development of cancers (15). In a previous study, extracts from A. marmelos were found to inhibit the proliferation of multiple human tumor cell lines in in vitro assays, including the leukemic K562, erythroleukemic HEL, melanoma Colo38, and breast cancer MCF7 and MDA-MB-231 cell lines (36). In the present study, we have also shown that the ethyl acetate fraction of A. marmelos significantly inhibits proliferation of many cells (Fig. 1A). Subsequently, we have identified a single molecule, which was structurally characterized as HDNC, by detailed NMR and MS analysis (Fig. 1B). This pure compound was also capable of inducing apoptosis to a variety of cell lines. Our in vivo studies have further shown that HDNC inhibits the growth of HCT-116 colon cancer xenografts in nude mice, with a significant reduction in the number of microcapillaries (Fig. 2). Furthermore, we have determined that HDNC acts to inhibit tumor growth by inducing TNF-α–mediated apoptosis program.

Two major apoptotic pathways exist in mammalian cells (5). The extrinsic pathway is initiated by the binding of transmembrane death receptors (e.g., Fas, TNFR1, and TRAIL receptors) with cognate extracellular ligands. Ligand-bound receptors recruit adaptor proteins (e.g., FADD), which interact with and trigger the activation of caspase-8. Activation of TNFR p55 expression by both the crude fraction and pure HDNC and activation of TNF-α imply that this compound was capable of inducing apoptosis. TNF-α is known to trigger the expression of a large battery of genes in diverse cell types (3739), including its specific role in triggering apoptotic death via TNFR1 receptor (40). Our studies show that the presence of the compound causes significantly higher expression levels of TNF-α in the cells coupled with overexpression of TRADD driving the cell into apoptosis via TNF-α–TRADD cascade. In this regard, it should be noted that TNFR1 has been the only receptor expressed in HEp-2 cells (41). However, the colon cancer cells used in the study do express both TNF receptors, but only TNFR1 was found to be induced, further strengthening our conclusion that HDNC acts through the TNFR1 receptor to induce apoptosis.

The intrinsic pathway is characterized by disruption of mitochondrial membrane integrity when cells are exposed to stresses such as DNA-damaging agents (6). Disruption of mitochondrial membrane integrity can trigger apoptosis via both caspase-9–dependent pathway that includes the truncation of Bid protein, which then binds to mitochondria and promotes mitochondrial membrane integrity disruption. Although it is not known whether HDNC can activate caspase-9, our findings suggest the possibility of a cross-talk between the two pathways, as activated caspase-8 can cleave Bid to produce t-Bid.

NF-κB/Rel transcription factors can block apoptosis induced by TNF-α (37, 42, 43). The antiapoptotic activity of NF-κB is also crucial for immunity, tumorigenesis, and cancer chemoresistance (37). With respect to TNF-α, the NF-κB–mediated suppression of apoptosis involves inhibition of JNK cascade. The control of TNFR-induced killing serves as paradigm for the antiapoptotic activity of NF-κB (44, 45). TNF-α normally has less or no cytotoxic effect unless NF-κB activation or protein synthesis is blocked (45). In line with this, EMSA and reporter gene analyses reveal suppression of TNF-α–induced NF-κB activation in colon cancer cells. We infer that this may aid in TNF-α–induced apoptosis by HDNC. However, further studies are required to determine whether knockdown of the NF-κB activation is essential for HDNC activity.

Caspases are activated in a branched protease cascade and control distinct downstream processes in TNF-α–induced and Fas-induced apoptosis (4, 46). To confirm whether induction of apoptosis is due to caspase-3 activation, inhibitor studies using specific caspase-3 reveal marked reduction in the induction of HDNC-mediated apoptosis. It would, however, also be interesting to determine whether additional pathways exist for HDNC-mediated caspase-3 activation by using dominant-negative mutant forms of caspase-8 and caspase-9. Nevertheless, the ability of the compound HDNC from A. marmelos to inhibit cell proliferation and induce apoptosis by a multiprolonged cascade of events suggests that it might be an ideal therapeutic for many solid tumors.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

D. Subramaniam and P. Giridharan contributed equally to this work.

Grant support: Department of Biotechnology, Government of India (A. Balakrishnan) and NIH grants CA109269 (S. Anant) and AT004118 (R.P. Ramanujam).

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
Reed JC. Apoptosis-targeted therapies for cancer.
Cancer Cell
2003
;
3
:
17
–22.
2
Sellers WR, Fisher DE. Apoptosis and cancer drug targeting.
J Clin Invest
1999
;
104
:
1655
–61.
3
Ching LM, Cao Z, Kieda C, Zwain S, Jameson MB, Baguley BC. Induction of endothelial cell apoptosis by the antivascular agent 5,6-dimethylxanthenone-4-acetic acid.
Br J Cancer
2002
;
86
:
1937
–42.
4
Nicholson DW, Thornberry NA. Caspases: killer proteases.
Trends Biochem Sci
1997
;
22
:
299
–306.
5
Cohen GM. Caspases: the executioners of apoptosis.
Biochem J
1997
;
326
:
1
–16.
6
Debatin KM. Apoptosis pathways in cancer and cancer therapy.
Cancer Immunol Immunother
2004
;
53
:
153
–9.
7
Peter ME. The flip side of FLIP.
Biochem J
2004
;
382
:
e1
–3.
8
Johnson CR, Jarvis WD. Caspase-9 regulation: an update.
Apoptosis
2004
;
9
:
423
–7.
9
Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis.
Cell
1998
;
94
:
491
–501.
10
Madesh M, Antonsson B, Srinivasula SM, Alnemri ES, Hajnoczky G. Rapid kinetics of t-Bid-induced cytochrome c and Smac/DIABLO release and mitochondrial depolarization.
J Biol Chem
2002
;
277
:
5651
–9.
11
Bayir H, Fadeel B, Palladino MJ, et al. Apoptotic interactions of cytochrome c: redox flirting with anionic phospholipids within and outside of mitochondria.
Biochim Biophys Acta
2006
;
1757
:
648
–59.
12
Forsburg SL, Guarente L. Communication between mitochondria and the nucleus in regulation of cytochrome genes in the yeast Saccharomyces cerevisiae.
Annu Rev Cell Biol
1989
;
5
:
153
–80.
13
Giege P, Grienenberger JM, Bonnard G. Cytochrome c biogenesis in mitochondria.
Mitochondrion
2008
;
8
:
61
–73.
14
Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer.
Nature
2001
;
411
:
342
–8.
15
Hanahan D, Weinberg RA. The hallmarks of cancer.
Cell
2000
;
100
:
57
–70.
16
Sausville EA, Johnson JI. Molecules for the millennium: how will they look? New drug discovery year 2000.
Br J Cancer
2000
;
83
:
1401
–4.
17
Manly SP, Padmanabha R, Lowe SE. Natural products or not? How to screen for natural products in the emerging HTS paradigm.
Methods Mol Biol
2002
;
190
:
153
–68.
18
Kolewe ME, Gaurav V, Roberts SC. Pharmaceutically active natural product synthesis and supply via plant cell culture technology.
Mol Pharm
2008
;
5
:
243
–56.
19
Saklani A, Kutty SK. Plant-derived compounds in clinical trials.
Drug Discov Today
2008
;
13
:
161
–71.
20
Rana BK, Singh UP, Taneja V. Antifungal activity and kinetics of inhibition by essential oil isolated from leaves of Aegle marmelos.
J Ethnopharmacol
1997
;
57
:
29
–34.
21
Jagetia GC, Venkatesh P, Baliga MS. Aegle marmelos (L.) Correa inhibits the proliferation of transplanted Ehrlich ascites carcinoma in mice.
Biol Pharm Bull
2005
;
28
:
58
–64.
22
Veerappan A, Miyazaki S, Kadarkaraisamy M, Ranganathan D. Acute and subacute toxicity studies of Aegle marmelos Corr., an Indian medicinal plant.
Phytomedicine
2007
;
14
:
209
–15.
23
Subramaniam D, May R, Sureban SM, et al. Diphenyl difluoroketone: a curcumin derivative with potent in vivo anticancer activity.
Cancer Res
2008
;
68
:
1962
–9.
24
Chandra D, Choy G, Deng X, Bhatia B, Daniel P, Tang DG. Association of active caspase 8 with the mitochondrial membrane during apoptosis: potential roles in cleaving BAP31 and caspase 3 and mediating mitochondrion-endoplasmic reticulum cross talk in etoposide-induced cell death.
Mol Cell Biol
2004
;
24
:
6592
–607.
25
Porter AG, Janicke RU. Emerging roles of caspase-3 in apoptosis.
Cell Death Differ
1999
;
6
:
99
–104.
26
Ferrandez A, Prescott S, Burt RW. COX-2 and colorectal cancer.
Curr Pharm Des
2003
;
9
:
2229
–51.
27
Harris RE. Cyclooxygenase-2 (cox-2) and the inflammogenesis of cancer.
Subcell Biochem
2007
;
42
:
93
–126.
28
Marnett LJ, DuBois RN. COX-2: a target for colon cancer prevention.
Annu Rev Pharmacol Toxicol
2002
;
42
:
55
–80.
29
Budihardjo I, Oliver H, Lutter M, Luo X, Wang X. Biochemical pathways of caspase activation during apoptosis.
Annu Rev Cell Dev Biol
1999
;
15
:
269
–90.
30
Luo J, Manning BD, Cantley LC. Targeting the PI3K-Akt pathway in human cancer: rationale and promise.
Cancer Cell
2003
;
4
:
257
–62.
31
Thompson JE, Thompson CB. Putting the rap on Akt.
J Clin Oncol
2004
;
22
:
4217
–26.
32
Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases.
Science
2002
;
298
:
1911
–2.
33
Hanada T, Yoshimura A. Regulation of cytokine signaling and inflammation.
Cytokine Growth Factor Rev
2002
;
13
:
413
–21.
34
Vallee S, Laforest S, Fouchier F, Montero MP, Penel C, Champion S. Cytokine-induced upregulation of NF-κB, IL-8, and ICAM-1 is dependent on colonic cell polarity: implication for PKCδ.
Exp Cell Res
2004
;
297
:
165
–85.
35
Hellmuth M, Wetzler C, Nold M, et al. Expression of interleukin-8, heme oxygenase-1 and vascular endothelial growth factor in DLD-1 colon carcinoma cells exposed to pyrrolidine dithiocarbamate.
Carcinogenesis
2002
;
23
:
1273
–9.
36
Lampronti I, Martello D, Bianchi N, et al. In vitro antiproliferative effects on human tumor cell lines of extracts from the Bangladeshi medicinal plant Aegle marmelos Correa.
Phytomedicine
2003
;
10
:
300
–8.
37
Gupta S. A decision between life and death during TNF-α-induced signaling.
J Clin Immunol
2002
;
22
:
185
–94.
38
Cosman D. A family of ligands for the TNF receptor superfamily.
Stem Cells
1994
;
12
:
440
–55.
39
Wong M, Ziring D, Korin Y, et al. TNFα blockade in human diseases: mechanisms and future directions.
Clin Immunol
2008
;
126
:
121
–36.
40
Inada H, Izawa I, Nishizawa M, et al. Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD.
J Cell Biol
2001
;
155
:
415
–26.
41
Hohmann HP, Remy R, Poschl B, van Loon AP. Tumor necrosis factors-α and -β bind to the same two types of tumor necrosis factor receptors and maximally activate the transcription factor NF-κB at low receptor occupancy and within minutes after receptor binding.
J Biol Chem
1990
;
265
:
15183
–8.
42
Machuca C, Mendoza-Milla C, Cordova E, et al. Dexamethasone protection from TNF-α-induced cell death in MCF-7 cells requires NF-κB and is independent from AKT.
BMC Cell Biol
2006
;
7
:
9
.
43
Nagaki M, Naiki T, Brenner DA, et al. Tumor necrosis factor α prevents tumor necrosis factor receptor-mediated mouse hepatocyte apoptosis, but not fas-mediated apoptosis: role of nuclear factor-κB.
Hepatology
2000
;
32
:
1272
–9.
44
Kucharczak J, Simmons MJ, Fan Y, Gelinas C. To be, or not to be: NF-κB is the answer—role of Rel/NF-κB in the regulation of apoptosis.
Oncogene
2003
;
22
:
8961
–82.
45
Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling.
Cell Death Differ
2003
;
10
:
45
–65.
46
Kondo S. Apoptosis by antitumor agents and other factors in relation to cell cycle checkpoints.
J Radiat Res
1995
;
36
:
56
–62.