Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation

  • Authors:
    • Thanet Sophonnithiprasert
    • Sirinun Nilwarangkoon
    • Yukio Nakamura
    • Ramida Watanapokasin
  • View Affiliations

  • Published online on: October 13, 2015     https://doi.org/10.3892/ijo.2015.3204
  • Pages: 2188-2196
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The combination of TNF-related apoptosis-inducing ligand (TRAIL) and bioactive compound to enhance apoptosis in TRAIL-resistant cancer is one of cancer treatment strategies. TRAIL possesses the unique capacity to selectively induce apoptosis in cancer cells both in vitro and in vivo with little effect on normal cells. Recent studies have reported that there are many TRAIL-resistant cancers. Thus, bioactive compounds that enhance cytotoxicity of TRAIL would be potential candidates for cancer therapeutic application. This study evaluated the cytotoxic and apoptosis induction upon combined treatment of TRAIL and goniothalamin, the natural styryl-lactone compound extracted from plant Goniothalamus spp., in LoVo cells. The results showed that a combination of goniothalamin and TRAIL enhanced caspase-dependent apoptosis induction in LoVo cells via both death receptor- and mitochondrial-mediated apoptosis pathways. In addition, goniothalamin enhanced TRAIL-induced apoptosis through increased death receptor DR5 expression and decreased anti-apoptotic regulator cFLIP. Interestingly, goniothalamin increased translocation of DR5 to cell surface and consequently contributed to the enhancement of TRAIL-induced apoptosis. In conclusion, this is the first report showing the combined treatment of goniothalamin and TRAIL was able to effectively enhance TRAIL-mediated apoptosis induction in TRAIL-refractory colorectal cancer, LoVo cells. Therefore, this study may offer a strategic cancer treatment against TRAIL-resistant cancers.

Introduction

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/APO2L) is a member of the tumor necrosis factor (TNF) family of cytokines and an effective inducer of apoptosis in cancer cells (1). The interaction of TRAIL with death receptors (DRs), including the death receptors TRAILR1 (also known as DR4 and TNFRSF10A) and TRAILR2 (also known as DR5, KILLER and TNFRSF10B), on the surface of cancer cells can trigger apoptotic cell death signaling through death receptor-mediated apoptosis pathway without any harmful effects to normal cells (2,3). TRAIL binding to DRs causes conformational changes in DRs, which leads to the recruitment of the adaptor protein Fas-associated death domain (FADD) and caspase-8 and -10 through the cytoplasmic death domain (DD). This forms the so-called death-inducing signaling complex (DISC). Normally, the DISC fully activates caspase-8 and triggers apoptosis by directly activating the executive caspases, such as caspase-3, -6 and -7, also found in Fas type I cancer cells. However, TRAIL-mediated apoptosis can also induce the mitochondria-mediated apoptosis pathway through the implication of mitochondrial dysfunction and caspase-9 activation via the cleavage of Bid (BH3-interacting domain death agonist) protein into truncated Bid (tBid) by caspase-8. tBid is capable of inducing mitochondrial outer membrane permeabilisation (MOMP) in cells in which the ratio of pro- and anti-apoptotic Bcl-2 family members allows it to do so leading to mitochondrial dysfunction in TRAIL-treated cancer cells, also found in Fas type II cancer cells (47).

However, some cancer cells are resistant to TRAIL-induced apoptosis, especially some highly malignant tumors such as pancreatic cancer, melanoma, neuroblastoma, prostate cancer and colon cancer (8,9). Failure to undergo apoptosis has been implicated in the resistance of cancer cells to TRAIL surveillance and tumor development. The mechanism of resistance to TRAIL-induced apoptosis in cancer can occur at different points in the signaling pathways of TRAIL-induced apoptosis. Dysfunctions or low expression of the DRs can lead to resistance. The defects in FADD and caspase-8 can lead to TRAIL resistance. Another cause of this defect is the high expression of cellular FADD-like interleukin-1β-converting enzyme-inhibitory protein (cFILP) which correlates with TRAIL resistance in several types of cancers because it can bind to FADD and/or caspase-8 and death receptors. This interaction in turn prevents DISC formation and subsequently suppresses the activation of caspase cascade (10,11). High expression of apoptosis inhibitors have been reported to result in TRAIL resistance in mitochondria-dependent type II cancer cells (9). Thus, developing strategies to overcome the TRAIL resistance are the topics of interest. Several observations suggest that the combination of TRAIL with effective small molecule compounds can sensitize the resistant cancer to TRAIL-induced apoptosis. Therefore, it has been assumed as strategy to potentiate the cytotoxicity of TRAIL and its therapeutic applications.

The combined compounds synergize TRAIL-induced apoptosis in cancer through two pathways. First, to increase the death receptors DR4/DR5 expression and trigger its translocation to cell membranes thus increasing TRAIL binding resulted in extrinsic apoptosis pathway. Several chemotherapeutic agents and natural compounds, such as CDDP (12), etoposide (13), PS-341 (bortezomib) (14), tunicamycin (15), rottlerin (16), brandisianins (17), sodium butyrate (18), inostamycin (19) were reported to upregulate the death receptor expression and subsequent sensitization of TRAIL-resistant cancer cells to TRAIL-induced apoptosis. Second, resistant mechanism of TRAIL-induced apoptosis is disrupted through downregulation of cFLIP expression. Natural compounds such as kurarinone (20), icaritin (21), withanolide E (22) were reported to downregulate cFLIP expression and subsequent sensitization of TRAIL-resistant cancer cells to TRAIL-induced apoptosis. Natural compounds, such as silibinin (23), gingerol (24) and indomethacin (25) were reported to possess both mechanisms of sensitizing TRAIL-resistant cancer cells.

The LoVo colorectal cancer (CRC) cell line is derived from left supraclavicular region; stage Dukes' C (26). The CRC is the second most and the third most common cancer in women (representing 9.2% of the total) and men (representing 10.0% of the total) worldwide, respectively (27). The CRC cell lines which resist to TRAIL-induced apoptosis remains a problem in the treatment of these cancers, thus the approaches for enhancing TRAIL-induced apoptosis are urgently required. The LoVo cell line was used as a model of TRAIL-refractory colorectal cancer cells in this study as they were reported to express significantly lower level of cell surface DR5 than the other colon cancer cell lines resulting in resistance to TRAIL treatment (8). Thus, finding the strategy to overcome the TRAIL-insensitive cancer cells is of importance.

Goniothalamin is a major bioactive styryl-lactone compound found in plant Goniothalamus macrophyllus (Blume) Hook. f. & Thomson, indigenous to South East Asia (28). Many reports suggested that goniothalamin showed cytotoxic activity against various cancer cell lines, such as liver, breast, and cervix (2934). Interestingly, our preliminary studies indicated that goniothalamin could increase DR5 expression while decrease cFILP expression in LoVo cells. These preliminary results suggested that goniothalamin has a potential use for combination with TRAIL treatment in TRAIL-resistant LoVo cells. In this study, the mechanisms to overcome the resistance to TRAIL-induced apoptosis were investigated using goniothalamin combining with TRAIL in TRAIL-resistant LoVo cells. This indicated the potential application of goniothalamin as a synergistic agent for combining with TRAIL treatment in colorectal cancer.

Materials and methods

Chemical and antibodies

Goniothalamin (IUPAC name: (2R)-2-[(E)-2-phenylethenyl]-2,3-dihydropyran-6-one) was obtained from Dr Wilawan Mahabusarakam, Faculty of Science, Prince of Songkla University, Thailand in purified powder form. The stems of Goniothalamus macrophyllus were collected from Songkhla province in the southern part of Thailand, in September, 2007. Identification was made by Mr. Ponlawat Pattarakulpisutti, Department of Biology, Faculty of Science, Prince of Songkla University. The specimen (Uraiwan 01) has been deposited in the Herbarium of Department of Biology, Faculty of Science, Prince of Songkla University, Thailand. Recombinant TRAIL was purchased from Merck Millipore Corp. (Merck KGaA, Darmstadt, Germany). Chemicals for cell viability assay including MTT (3-(4,5-dimethyl)-2,5-diphenyl tetrazolium bromide) and dimethylsulfoxide (DMSO) were purchased from Sigma-Aldrich Corp. (St. Louis, MO, USA). Chemicals for flow cytometry analysis including Guava Cell Cycle® reagent and Guava Nexin® reagent were purchased from Merck Millipore Corp. (Merck KGaA), and PE-conjugated DR5 antibody was purchased from eBioscience, Inc. (San Diego, CA, USA). Chemical for fluorescence microscope observation Hoechst 33342 dye was purchased from Fisher Scientific, Inc. (Invitrogen™, Waltham, MA, USA). Chemical for mRNA extraction and cDNA synthesis were purchased from Qiagen N.V. (QIAzol™ lysis reagent, Venlo, LI, The Netherlands) and Thermo Fisher Scientific, Inc. (RevertAid™ First Strand cDNA Synthesis kit, Fermentas™, Waltham, MA, USA), respectively. Chemical for quantitative PCR was obtained from Thermo Fisher Scientific, Inc. (SYBR® Select Master Mix, Applied Biosystems™, Waltham, MA, USA). Antibodies (Abs) for immunoblot analysis including mouse monoclonal Abs against CHOP, and rabbit monoclonal Abs against DR5, PARP, caspase-3, caspase-8, caspase-9, Bcl2, Bax, Bid, Mcl1, and anti-mouse immunoglobulin G and anti-rabbit immunoglobulin G horseradish peroxidase-conjugated secondary antibodies were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA), and mouse monoclonal Abs against phospho-histone H2AX at Ser139 (γ-H2AX), β-actin and rabbit monoclonal Abs against cFLIP were obtained from Merck Millipore Corp. (Merck KGaA).

Cell culture

Human colorectal cancer, LoVo cell line, was obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). It was maintained in RPMI-1640 medium (Gibco Life Technologies, Carlbad, CA, USA) supplemented with 10% fetal bovine serum (GE Healthcare Life Science, Little Chalfont, UK), 100 U/ml penicillin and 100 μg/ml streptomycin (GE Healthcare Life Science, Inc., Little Chalfont, UK) at 37°C in a humidified 5% CO2 atmosphere and used for assays during exponential phase of growth.

Cell viability assay

Cells/well (5×103) were seeded in a 96-well plate. After adherence, culture medium containing 10 and 100 ng/ml of TRAIL alone and in combination with different goniothalamin concentrations 5, 15, 25 and 50 μM were incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. Cytotoxicity of goniothalamin was determined by cell proliferation analysis using MTT assay as described by Denizot and Lang (35). Briefly, after the indicated treatment, 0.5 mg/ml of MTT solution dissolved in culture medium was added and the cells were incubated for 2 h at 37°C with 5% CO2 in the incubator, the MTT solution was removed and 100 μl of DMSO was added to dissolve the formazan crystals, a product of cell respiration as for viable cells, and the absorbance at 540 nm was quantified on Epoch™ Microplate Spectrophotometer and analyzed by Gen5™ Data Analysis software (BioTek, CA, USA).

Chromatin condensation

Cells/well (8×104) were seeded in a 12-well plate. After adherence, culture medium containing 10 and 100 ng/ml of TRAIL alone and in combination with 15μM goniothalamin were incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. Chromatin condensation, a character of apoptosis, was detected by cell staining with a fluorescent dye Hoechst 33342 modified from Oberhammer et al (36). After treatment, the treated cells were washed and fixed with fixative solution (4% paraformaldehyde) for 15 min at room temperature. The fixed cells were washed and then stained with chromatin staining solution (5 μg/ml of Hoechst 33342) for 15 min. After staining, the stained cell were washed and then the plates were observed using a fluorescence microscope IX73 model (Olympus, Tokyo, Japan) with U-MWU2 mirror units for ultraviolet excitation.

Cell cycle determination

Cells/well (2×105) were seeded in each 6-well plate. After adherence, culture medium containing 10 and 100 ng/ml of TRAIL alone and in combination with 15 μM goniothalamin were incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. After treatment, the whole cells were collected and stained according to the manufacturer's instructions (Guava Cell Cycle® reagent from Merck Millipore Corp.; Merck KGaA). The stained cells were then sorted and analyzed for DNA content by a Guava easyCyte™ flow cytometer and GuavaSoft™ software (Merck Millipore Corp.; Merck KGaA), respectively.

Cell surface phosphatidyl-serine determination

Cells/well (2×105) were seeded in a 6-well plate. After adherence, culture medium containing 10 and 100 ng/ml of TRAIL alone and in combination with 15 μM goniothalamin were incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. After treatment, whole cells were collected and stained according to the manufacturer's instructions (Guava Nexin® reagent from Merck Millipore Corp.; Merck KGaA). The stained cells were sorted and analyzed for cell surface phosphatidyl-serine content a Guava easyCyte™ flow cytometer and GuavaSoft™ software (Merck Millipore Corp.; Merck KGaA), respectively.

Cell surface DR5 determination

Cells/well (2×105) were seeded in a 6-well plate. After adherence, culture medium containing different goniothalamin concentrations of 1, 5, 15 and 25 μM was incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. After treatment, the whole cells were collected and resuspended in PBS buffer containing PE-conjugated DR5 antibody, then incubated in the dark for 1 h at room temperature. The cells were washed and resuspended in PBS solution then sorted and analyzed for cell surface DR5 by a Guava easyCyte™ flow cytometer and GuavaSoft software (Merck Millipore Corp.; Merck KGaA), respectively.

mRNA expression analysis

Cells/well (8×104)were seeded in a 12-well plate. After adherence, culture medium containing 15μM goniothalamin was incubated for 24 h at 37°C with 5% CO2. The control group was treated with 0.5% DMSO. Analysis of mRNA expression was performed using the two step quantitative reverse transcriptase (RT)-PCR. After treatment, the whole cells were collected and RNA was extracted by using QIAzol lysis reagent (Qiagen N.V.) and cDNA synthesis by reverse transcription according RevertAid First Strand cDNA Synthesis kit (Fermentas, Thermo Fisher Scientific) with 2 μg of total RNA of each sample. In quantitative PCR step, it was performed with SYBR Select Master Mix (Applied Biosystems, Thermo Fisher Scientific, Inc., Waltham, MA, USA). The primers used for amplification were: DR5 (forward 5′-CACCAGGTGTGATTCAGGTG-3′ and reverse 5′-TACGGCTGCAACTGTGACTC-3′), CHOP (forward 5′-GCGCATGAAGGAGAAAGAAC-3′ and reverse 5′-TCACCATTCGGTCAATCAGA-3′), cFILPL (forward 5′-ATTGCATTGGCAATGAGACAGAGC-3′ and reverse 5′-TCGGTGCT CGGGCATACAGG-3′), cFILPS (forward 5′-GGGCCGAG GCAAGATAAGCAAGG-3′ and reverse 5′-TCAGGACAAT GGGCATAGGGTGT-3′), and GAPDH (forward 5′-AGGTCG GAGTCAACGGATTT-3′ and reverse 5′-TAGTTGAGGTC AATGAAGGG-3′). The PCR amplification was analyzed by CFX96 Touch™ Real-Time PCR Detection system with CFX Manager™ software (Bio-Rad Laboratories, Inc., CA, USA). All steps were performed according to the manufacturer's instructions.

Protein expression analysis by immunoblotting

Cells/well (2×105) were seeded in a 6-well plate. After adherence, the cells were treated with appropriate condition. After treatment, the cells were lysed with RIPA lysis buffer (50 mM Tris-HCl, pH 7.4, 1% NP-40, 0.5% C24H39NaO4, 0.1% SDS, 150 mM NaCl, 2 mM EDTA, 50 mM NaF). The extracted proteins were separated on 8–15% acrylamind gel and transferred onto a polyvinylidene fluoride (PVDF) membrane (Merck Millipore Corp., Merck KGaA). Then, the membranes were blocked with 5% skimmed-milk in TBS-Tween buffer for 1 h at room temperature and incubated with mouse monoclonal Abs against CHOP, γ-H2AX, and rabbit monoclonal Abs against DR5, PARP, caspase-3, caspase-8, caspase-9, Bcl2, Bax, Bid), cFLIP, β-actin overnight at 4°C. Following incubation with anti-mouse immunoglobulin G or anti-rabbit immunoglobulin G horseradish peroxidase-conjugated secondary antibodies for 1 h a room temperature, the signals were developed using Immobilon™ Western chemiluminescent HRP substrate (Merck Millipore Corp., Merck KGaA, Darmstadt, Germany) and detected under Chemiluminescent Imaging system (GeneGnome gel documentation, Synoptics Ltd., Cambridge, UK).

Statistical analysis

To compare the data from different treatments, Student's t-test was used. All data presented were obtained from at least three independent experiments and presented as mean ± standard deviation (SD). A p-value of 0.05 was taken as minimum basis for assigning significance.

Results

Enhanced TRAIL-induced apoptosis in LoVo cells by co-treatment with goniothalamin

Our preliminary study showed that LoVo cells were insensitive to goniothalamin treatment with high IC50 value at 65.25±1.85 μM. However, goniothalamin induced increased DR5 expression at lower concentration than IC50 value indicating that goniothalamin in combination with TRAIL may have a potential to trigger apoptosis induction via the death receptor-TRAIL mediated apoptosis pathway. Thus, we tried to investigate the mechanisms of apoptosis induction upon combination of TRAIL and goniothalamin in LoVo cells.

In this study, we first found that co-treatment of goniothalamin and TRAIL enhanced cytotoxicity induction in LoVo cells. We confirmed this cytotoxic effects and apoptosis induction using the MTT assay and Hoechst 33342 staining to assess chromatin condensation as shown in Fig. 1A and B, respectively. Treatment of LoVo cells with 10 and 100 ng/ml of TRAIL for 24 h showed >80% cell viability, while combined treatment with 15 μM goniothalamin resulted in enhanced cytotoxicity in LoVo cells. The increased chromatin condensation is shown in Fig. 1B upon combining treatment of 15 μM goniothalamin and 10 and 100 ng/ml of TRAIL as compared to a single treatment. Moreover, other apoptotic characteristics, accumulation of subG1 phase population and cell surface phosphatidyl-serine presentation, were studied using flow cytometry technique. As shown in Fig. 2A and C, a significant increased accumulation of a subG1 phase population was detected upon treatment with 15 μM goniothalamin and 10 ng/ml TRAIL as compared to a single treatment, but not for the combination of 15 μM goniothalamin and 100 ng/ml TRAIL as compared to a single 100 ng/ml TRAIL treatment. In addition, the significant increased cell surface phosphatidyl-serine presentation was detected upon combined treatment with both 10 and 100 ng/ml of TRAIL as compared to a single treatment (Fig. 2B and D). Thus, these results indicated that the combined treatment of goniothalamin and TRAIL enhanced cytotoxicity and apoptosis induction in LoVo cells, especially at 15 μM goniothalamin and 10 ng/ml TRAIL, which was selected for use in the next steps to assess apoptosis pathway.

Combined treatment with goniothalamin and TRAIL accelerate apoptosis induction by a caspase activation-dependent pathway involved with both death receptor- and mitochondrial-mediated apoptosis pathways in LoVo cells

To observe whether the combination of goniothalamin and TRAIL leads to activation of caspase-activated apoptosis in TRAIL-resistant LoVo cells, apoptotic-related protein was assessed by immunoblot analysis. As shown in Fig. 3, caspase-activated apoptotic mediators including PARP and caspase-3 as executive apoptosis, caspase-8 and Bid as death receptor mediated apoptosis pathway, caspase-9, Bcl2 and Bax as mitochondrial mediated apoptosis pathway were determined. The results indicated that both extrinsic and intrinsic pathway were enhanced upon combined treatment of 15 μM goniothalamin and 10 or 100 ng/ml of TRAIL, as confirmed by increasing the cleaved form of PARP, caspase-3, caspase-8, caspase-9, Bid, decreased antiapoptotic Bcl2 and increased proapoptotic Bax expression. Moreover, increased phosphorylation of histone the so-called γ-H2AX, was observed upon the combined treatments, these results indicated that goniothalamin induced DNA double-strand breaks and triggered apoptosis-associated γ-H2AX accumulation, which is one of apoptotic characteristics. The results indicated that these combined treatment can induce cytotoxicity resulting in DNA double-strand break in LoVo cells.

Goniothalamin enhances TRAIL-induced apoptosis through DR5 upregulation and cFLIP downregulation

TRAIL-stimulated death signal is initiated by the binding of TRAIL to DR5 resulted in the subsequent caspase-8 activation. As shown in Fig. 4, immunoblot analysis showed that goniothalamin dramatically upregulated the DR5 and CHOP protein but downregulated the cFLIP and Mcl1 protein in a dose-dependent manner (Fig. 4A). Treatment with 15 μM goniothalamin at various time points showed that goniothalamin upregulated the DR5 and CHOP protein while downregulated the antiapoptotic cFLIP and Mcl1 in a time-dependent manner (Fig. 4B). These results corresponded with quantitative RT-PCR analysis (Fig. 4C) indicating that DR5 and CHOP mRNA were upregulated whereas cFLIPL and cFLIPS mRNA was significantly downregulated. Moreover, the translocation of DR5 to cell surface was analyzed and the results indicated that goniothalamin increased cell surface DR5 expression in a dose-dependent manner (Fig. 4D). These results implied that DR5 upregulation induced by increased CHOP expression together with the downregulation of cFLIP and Mcl1 contributed to enhanced TRAIL sensitization.

Discussion

TRAIL, also known as Apo-2L, is a typical member of TNF ligand family that induces apoptosis via death-receptor mediated pathway. TRAIL has potential benefits in cancer therapy because of its potent ability to be selectively toxic in cancer cells. Unlike the other death ligands such as TNF-α or FasL, the treatment of TRAIL causes less toxic in normal cells (37,38). Furthermore, the combined treatment of TRAIL and genotoxic chemotherapeutic agents synergistically inhibited cancer cell growth which are otherwise resistant or less toxicity to treatment with TRAIL or chemotherapy alone (3739). There are several recombinant TRAIL and TRAIL-receptor agonists as an anticancer therapy that have been tested in phase I and II trials in patients with advanced cancer. Clinical studies in TRAIL-receptor agonist are being investigated using combination treatment in patients with advanced cancer stage (40). However, the single TRAIL treatment probably is not feasible since the majority of cancer cells are resistant to TRAIL. Thus, the combination treatment with TRAIL and chemotherapy is essential for use in TRAIL-resistant cancers. We also analyzed in detail that TRAIL combined treatment with cytotoxic agent goniothalamin may enhance cytotoxicity and apoptosis induction in colorectal cancer cells, indicating a potential use for cancer therapy.

In this study, we demonstrated for the first time that goniothalamin upon combined treatment with TRAIL-regulated expression of antiapoptotic- and proapoptotic-related death receptor-mediated apoptotic molecules, including upregulation of DR5 and CHOP, downregulation of cFLIP and Mcl1 resulting in enhancement of the ability of TRAIL in TRAIL refractory LoVo cells. Various studies have reported the increased transcriptional activation of DR5 gene by the upregulation of CHOP expression (4145), these correlated to the upregulation of CHOP expression in goniothalamin treatment. Another mechanism which is involved in sensitization to TRAIL-induced apoptosis is downregulation of cFLIP and Mcl1. cFLIP is the major protein that prevents caspase-8 from activation by death receptors through binding to FADD and/or caspase-8 and TRAIL receptor DR5 in a ligand-dependent and -independent manner and forms an apoptosis inhibitory complex (AIC), then prevents death-inducing signaling complex (DISC) formation and subsequently suppress the activation of caspase cascade (4653). Mcl1 is an antiapoptotic protein involved in death receptor mediated pathway cross-link to mitochondrial mediated pathway by interacting with truncated Bid (tBid) and then strongly inhibits tBid-induced cytochrome c release in mitochondrial mediated apoptosis pathway (5457). Downregulation of cFLIP and Mcl1 expression sensitizes TRAIL-induced apoptosis in various TRAIL refractory cancers. Thus, we speculated that goniothalamin plus TRAIL might play a critical role in goniothalamin-stimulated TRAIL-mediated apoptosis in LoVo cells through both upregulation of DR5 and CHOP and downregulation of cFLIP and Mcl1 expression enhancing TRAIL ability to be selectively cytotoxic to TRAIL-refractory LoVo cells. Moreover, DR5 translocation to cell surface was increased by goniothalamin treatment that may increase potent death receptor-mediated apoptosis induction by TRAIL through binding to DR5 (5860).

Caspase-dependent pathways of these TRAIL-mediated apoptosis are involved in this combination treatment, resulting in a strong enhancement of PARP, caspase-3, -8 and -9 activation. TRAIL triggered death receptor mediated apoptosis pathway via binding to death receptor DR5 resulting in induction cleavage of Bid to tBid, then crosslinking to mitochondria-mediated apoptosis activation (47,61), supported by downregulation of Mcl1 in goniothalamin treatment. Our results showed that apoptotic related molecules triggered activation in both death receptor- and mitochondrial-mediated apoptosis pathways under combination treatment of goniothalamin and TRAIL. Moreover, the increased accumulation of subG1 phase population in cell cycle, increased cell surface phosphatidyl-serine presentation and increased phosphorylation of H2AX were observed under the combined treatment of TRAIL and goniothalamin, indicating apoptosis induction in LoVo cells as compared to a single treatment with TRAIL or goniothalmin alone (6265). Similar reports of other compounds sensitize TRAIL-induced apoptosis include inostamycin (19), delphinidin (66), and parthenolide (67). Therefore, the combined treatment of TRAIL and goniothalamin enhanced cytotoxicity in TRAIL refractory LoVo cells through caspase-dependent apoptosis pathway in both death receptor- and mitochondrial-mediated pathways.

In conclusion, this is the first report that the combination of TRAIL and goniothalamin was able to effectively enhance TRAIL mediated apoptosis induction in TRAIL refractory colorectal cancer, LoVo cells. In addition, we found that goniothalamin enhanced TRAIL sensitization in LoVo cells associated with caspase cascade activation via induction of the DR5 pathway and decreased expression level of antiapoptotic proteins which related to DR5 pathway and subsequently increased apoptosis. From these results, we speculate that combined treatment of TRAIL and goniothalamin provides a possible therapeutic application for treatment of colorectal cancer that are resistant to TRAIL.

Acknowledgements

This study was supported by The Royal Golden Jubilee (RGJ) Ph.D. Program from Thailand Research Fund (TRF) Thailand and the Strategic Wisdom and Research Institute, Srinakharinwirot University, Bangkok, Thailand.

References

1 

Fukuda M, Hamao A, Tanaka A, Kitada M, Suzuki S, Kusama K and Sakashita H: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/APO2L) and its receptors expression in human squamous cell carcinoma of the oral cavity. Oncol Rep. 10:1113–1119. 2003.PubMed/NCBI

2 

Kichev A, Rousset CI, Baburamani AA, Levison SW, Wood TL, Gressens P, Thornton C and Hagberg H: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and cell death in the immature central nervous system after hypoxia-ischemia and inflammation. J Biol Chem. 289:9430–9439. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, Chin W, Jones J, Woodward A, Le T, et al: Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 5:157–163. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Li H, Zhu H, Xu CJ and Yuan J: Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell. 94:491–501. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Gross A, McDonnell JM and Korsmeyer SJ: BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 13:1899–1911. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Schug ZT, Gonzalvez F, Houtkooper RH, Vaz FM and Gottlieb E: BID is cleaved by caspase-8 within a native complex on the mitochondrial membrane. Cell Death Differ. 18:538–548. 2011. View Article : Google Scholar :

7 

Kantari C and Walczak H: Caspase-8 and bid: Caught in the act between death receptors and mitochondria. Biochim Biophys Acta. 1813:558–563. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Galligan L, Longley DB, McEwan M, Wilson TR, McLaughlin K and Johnston PG: Chemotherapy and TRAIL-mediated colon cancer cell death: The roles of p53, TRAIL receptors, and c-FLIP. Mol Cancer Ther. 4:2026–2036. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Zhang L and Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 12:228–237. 2005. View Article : Google Scholar

10 

Lemke J, von Karstedt S, Zinngrebe J and Walczak H: Getting TRAIL back on track for cancer therapy. Cell Death Differ. 21:1350–1364. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Grambihler A, Higuchi H, Bronk SF and Gores GJ: cFLIP-L inhibits p38 MAPK activation: An additional anti-apoptotic mechanism in bile acid-mediated apoptosis. J Biol Chem. 278:26831–26837. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Nagane M, Pan G, Weddle JJ, Dixit VM, Cavenee WK and Huang HJ: Increased death receptor 5 expression by chemotherapeutic agents in human gliomas causes synergistic cytotoxicity with tumor necrosis factor-related apoptosis-inducing ligand in vitro and in vivo. Cancer Res. 60:847–853. 2000.PubMed/NCBI

13 

Sheikh MS, Burns TF, Huang Y, Wu GS, Amundson S, Brooks KS, Fornace AJ Jr and el-Deiry WS: p53-dependent and -independent regulation of the death receptor KILLER/DR5 gene expression in response to genotoxic stress and tumor necrosis factor alpha. Cancer Res. 58:1593–1598. 1998.PubMed/NCBI

14 

Liu X, Yue P, Chen S, Hu L, Lonial S, Khuri FR and Sun SY: The proteasome inhibitor PS-341 (bortezomib) up-regulates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced apoptosis despite up-regulation of c-FLIP and survivin expression in human NSCLC cells. Cancer Res. 67:4981–4988. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Shiraishi T, Yoshida T, Nakata S, Horinaka M, Wakada M, Mizutani Y, Miki T and Sakai T: Tunicamycin enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in human prostate cancer cells. Cancer Res. 65:6364–6370. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Lim JH, Park JW, Choi KS, Park YB and Kwon TK: Rottlerin induces apoptosis via death receptor 5 (DR5) upregulation through CHOP-dependent and PKC delta-independent mechanism in human malignant tumor cells. Carcinogenesis. 30:729–736. 2009. View Article : Google Scholar

17 

Kikuchi H, Ohtsuki T, Koyano T, Kowithayakorn T, Sakai T and Ishibashi M: Brandisianins A-F, isoflavonoids isolated from Millettia brandisiana in a screening program for death-receptor expression enhancement activity. J Nat Prod. 70:1910–1914. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Kim YH, Park JW, Lee JY and Kwon TK: Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis. 25:1813–1820. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Yamamoto K, Makino M, Watanapokasin R, Tashiro E and Imoto M: Inostamycin enhanced TRAIL-induced apoptosis through DR5 upregulation on the cell surface. J Antibiot (Tokyo). 65:295–300. 2012. View Article : Google Scholar

20 

Zhou W, Cao A, Wang L and Wu D: Kurarinone synergizes TRAIL-induced apoptosis in gastric cancer cells. Cell Biochem Biophys. 72:241–249. 2014. View Article : Google Scholar

21 

Han H, Xu B, Hou P, Jiang C, Liu L, Tang M, Yang X, Zhang Y and Liu Y: Icaritin sensitizes human glioblastoma cells to TRAIL-induced apoptosis. Cell Biochem Biophys. 72:533–542. 2015. View Article : Google Scholar

22 

Henrich CJ, Brooks AD, Erickson KL, Thomas CL, Bokesch HR, Tewary P, Thompson CR, Pompei RJ, Gustafson KR, McMahon JB and Sayers TJ: Withanolide E sensitizes renal carcinoma cells to TRAIL-induced apoptosis by increasing cFLIP degradation. Cell Death Dis. 6:e16662015. View Article : Google Scholar : PubMed/NCBI

23 

Son YG, Kim EH, Kim JY, Kim SU, Kwon TK, Yoon AR, Yun CO and Choi KS: Silibinin sensitizes human glioma cells to TRAIL-mediated apoptosis via DR5 up-regulation and down-regulation of c-FLIP and survivin. Cancer Res. 67:8274–8284. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Lee DH, Kim DW, Jung CH, Lee YJ and Park D: Gingerol sensitizes TRAIL-induced apoptotic cell death of glioblastoma cells. Toxicol Appl Pharmacol. 279:253–265. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Tse AK, Cao HH, Cheng CY, Kwan HY, Yu H, Fong WF and Yu ZL: Indomethacin sensitizes TRAIL-resistant melanoma cells to TRAIL-induced apoptosis through ROS-mediated upregulation of death receptor 5 and downregulation of survivin. J Invest Dermatol. 134:1397–1407. 2014. View Article : Google Scholar

26 

Ahmed D, Eide PW, Eilertsen IA, Danielsen SA, Eknæs M, Hektoen M, Lind GE and Lothe RA: Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis. 2:e712013. View Article : Google Scholar : PubMed/NCBI

27 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar

28 

Wattanapiromsakul C, Wangsintaweekul B, Sangprapan P, Itharat A and Keawpradub N: Goniothalamin, a cytotoxic compound, isolated from Goniothalamus macrophyllus (Blume) Hook. f & Thomson var macrophyllus Songklanakarin. J Sci Technol. 27:479–487. 2005.

29 

Inayat-Hussain SH, Annuar BO, Din LB, Ali AM and Ross D: Loss of mitochondrial transmembrane potential and caspase-9 activation during apoptosis induced by the novel styryl-lactone goniothalamin in HL-60 leukemia cells. Toxicol In Vitro. 17:433–439. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Chan KM, Rajab NF, Ishak MH, Ali AM, Yusoff K, Din LB and Inayat-Hussain SH: Goniothalamin induces apoptosis in vascular smooth muscle cells. Chem Biol Interact. 159:129–140. 2006. View Article : Google Scholar

31 

Chen WY, Wu CC, Lan YH, Chang FR, Teng CM and Wu YC: Goniothalamin induces cell cycle-specific apoptosis by modulating the redox status in MDA-MB-231 cells. Eur J Pharmacol. 522:20–29. 2005. View Article : Google Scholar : PubMed/NCBI

32 

de Fátima A, Kohn LK, Antônio MA, de Carvalho JE and Pilli RA: (R)-Goniothalamin: Total syntheses and cytotoxic activity against cancer cell lines. Bioorg Med Chem. 13:2927–2933. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Alabsi AM, Ali R, Ali AM, Al-Dubai SA, Harun H, Abu Kasim NH and Alsalahi A: Apoptosis induction, cell cycle arrest and in vitro anticancer activity of gonothalamin in a cancer cell lines. Asian Pac J Cancer Prev. 13:5131–5136. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Petsophonsakul P, Pompimon W and Banjerdpongchai R: Apoptosis induction in human leukemic promyelocytic HL-60 and monocytic U937 cell lines by goniothalamin. Asian Pac J Cancer Prev. 14:2885–2889. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Denizot F and Lang R: Rapid colorimetric assay for cell growth and survival. Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol Methods. 89:271–277. 1986. View Article : Google Scholar : PubMed/NCBI

36 

Oberhammer FA, Hochegger K, Fröschl G, Tiefenbacher R and Pavelka M: Chromatin condensation during apoptosis is accompanied by degradation of lamin A+B, without enhanced activation of cdc2 kinase. J Cell Biol. 126:827–837. 1994. View Article : Google Scholar : PubMed/NCBI

37 

Shi J, Zheng D, Man K, Fan ST and Xu R: TRAIL: A potential agent for cancer therapy. Curr Mol Med. 3:727–736. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Nagane M, Huang HJ and Cavenee WK: The potential of TRAIL for cancer chemotherapy. Apoptosis. 6:191–197. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Shankar S and Srivastava RK: Enhancement of therapeutic potential of TRAIL by cancer chemotherapy and irradiation: Mechanisms and clinical implications. Drug Resist Updat. 7:139–156. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Buchsbaum DJ, Forero-Torres A and LoBuglio AF: TRAIL-receptor antibodies as a potential cancer treatment. Future Oncol. 3:405–409. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Krajarng A, Imoto M, Tashiro E, Fujimaki T, Shinjo S and Watanapokasin R: Apoptosis induction associated with the ER stress response through up-regulation of JNK in HeLa cells by gambogic acid. BMC Complement Altern Med. 15:262015. View Article : Google Scholar : PubMed/NCBI

42 

Trivedi R, Maurya R and Mishra DP: Medicarpin, a legume phytoalexin sensitizes myeloid leukemia cells to TRAIL-induced apoptosis through the induction of DR5 and activation of the ROS-JNK-CHOP pathway. Cell Death Dis. 5:e14652014. View Article : Google Scholar : PubMed/NCBI

43 

Pennati M, Sbarra S, De Cesare M, Lopergolo A, Locatelli SL, Campi E, Daidone MG, Carlo-Stella C, Gianni AM and Zaffaroni N: YM155 sensitizes triple-negative breast cancer to membrane-bound TRAIL through p38 MAPK- and CHOP-mediated DR5 upregulation. Int J Cancer. 136:299–309. 2015. View Article : Google Scholar

44 

Yi L, Zongyuan Y, Cheng G, Lingyun Z, Guilian Y and Wei G: Quercetin enhances apoptotic effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in ovarian cancer cells through reactive oxygen species (ROS) mediated CCAAT enhancer-binding protein homologous protein (CHOP)-death receptor 5 pathway. Cancer Sci. 105:520–527. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Yoon MJ, Kang YJ, Kim IY, Kim EH, Lee JA, Lim JH, Kwon TK and Choi KS: Monensin, a polyether ionophore antibiotic, overcomes TRAIL resistance in glioma cells via endoplasmic reticulum stress, DR5 upregulation and c-FLIP downregulation. Carcinogenesis. 34:1918–1928. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Zhang S, Shen HM and Ong CN: Down-regulation of c-FLIP contributes to the sensitization effect of 3,3′-diindolylmethane on TRAIL-induced apoptosis in cancer cells. Mol Cancer Ther. 4:1972–1981. 2005. View Article : Google Scholar : PubMed/NCBI

47 

García-García C, Fumarola C, Navaratnam N, Carling D and López-Rivas A: AMPK-independent down-regulation of cFLIP and sensitization to TRAIL-induced apoptosis by AMPK activators. Biochem Pharmacol. 79:853–863. 2010. View Article : Google Scholar

48 

Lin Y, Liu X, Yue P, Benbrook DM, Berlin KD, Khuri FR and Sun SY: Involvement of c-FLIP and survivin down-regulation in flexible heteroarotinoid-induced apoptosis and enhancement of TRAIL-initiated apoptosis in lung cancer cells. Mol Cancer Ther. 7:3556–3565. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Day TW, Huang S and Safa AR: c-FLIP knockdown induces ligand-independent DR5-, FADD-, caspase-8-, and caspase-9-dependent apoptosis in breast cancer cells. Biochem Pharmacol. 76:1694–1704. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Safa AR and Pollok KE: Targeting the anti-apoptotic protein c-FLIP for cancer therapy. Cancers (Basel). 3:1639–1671. 2011. View Article : Google Scholar

51 

Safa AR: c-FLIP, a master anti-apoptotic regulator. Exp Oncol. 34:176–184. 2012.PubMed/NCBI

52 

Safa AR: Roles of c-FLIP in apoptosis, necroptosis, and autophagy. J Carcinog Mutagen. (Suppl 6): pii: 003. 2013.PubMed/NCBI

53 

Wilson TR, McLaughlin KM, McEwan M, Sakai H, Rogers KM, Redmond KM, Johnston PG and Longley DB: c-FLIP: A key regulator of colorectal cancer cell death. Cancer Res. 67:5754–5762. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Lee SJ, Noh HJ, Sung EG, Song IH, Kim JY, Kwon TK and Lee TJ: Berberine sensitizes TRAIL-induced apoptosis through proteasome-mediated downregulation of c-FLIP and Mcl-1 proteins. Int J Oncol. 38:485–492. 2011. View Article : Google Scholar

55 

Murphy AC, Weyhenmeyer B, Noonan J, Kilbride SM, Schimansky S, Loh KP, Kögel D, Letai AG, Prehn JH and Murphy BM: Modulation of Mcl-1 sensitizes glioblastoma to TRAIL-induced apoptosis. Apoptosis. 19:629–642. 2014. View Article : Google Scholar :

56 

Kim SH, Ricci MS and El-Deiry WS: Mcl-1: A gateway to TRAIL sensitization. Cancer Res. 68:2062–2064. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Clohessy JG, Zhuang J, de Boer J, Gil-Gómez G and Brady HJ: Mcl-1 interacts with truncated Bid and inhibits its induction of cytochrome c release and its role in receptor-mediated apoptosis. J Biol Chem. 281:5750–5759. 2006. View Article : Google Scholar

58 

Ozören N and El-Deiry WS: Cell surface death receptor signaling in normal and cancer cells. Semin Cancer Biol. 13:135–147. 2003. View Article : Google Scholar : PubMed/NCBI

59 

Chen JJ, Mikelis CM, Zhang Y, Gutkind JS and Zhang B: TRAIL induces apoptosis in oral squamous carcinoma cells - a crosstalk with oncogenic Ras regulated cell surface expression of death receptor 5. Oncotarget. 4:206–217. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Ren YG, Wagner KW, Knee DA, Aza-Blanc P, Nasoff M and Deveraux QL: Differential regulation of the TRAIL death receptors DR4 and DR5 by the signal recognition particle. Mol Biol Cell. 15:5064–5074. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Wei MC, Lindsten T, Mootha VK, Weiler S, Gross A, Ashiya M, Thompson CB and Korsmeyer SJ: tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes Dev. 14:2060–2071. 2000.PubMed/NCBI

62 

Pietenpol JA and Stewart ZA: Cell cycle checkpoint signaling: Cell cycle arrest versus apoptosis. Toxicology. 181–182:475–481. 2002. View Article : Google Scholar

63 

van Engeland M, Nieland LJ, Ramaekers FC, Schutte B and Reutelingsperger CP: Annexin V-affinity assay: A review on an apoptosis detection system based on phosphatidylserine exposure. Cytometry. 31:1–9. 1998. View Article : Google Scholar : PubMed/NCBI

64 

Kuo LJ and Yang LX: Gamma-H2AX - a novel biomarker for DNA double-strand breaks. In Vivo. 22:305–309. 2008.PubMed/NCBI

65 

Rogakou EP, Nieves-Neira W, Boon C, Pommier Y and Bonner WM: Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. J Biol Chem. 275:9390–9395. 2000. View Article : Google Scholar : PubMed/NCBI

66 

Ko H, Jeong MH, Jeon H, Sung GJ, So Y, Kim I, Son J, Lee SW, Yoon HG and Choi KC: Delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis, by inducing DR5 and causing caspase-mediated HDAC3 cleavage. Oncotarget. 6:9970–9984. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Trang KT, Kim SL, Park SB, Seo SY, Choi CH, Park JK, Moon JC, Lee ST and Kim SW: Parthenolide sensitizes human colorectal cancer cells to tumor necrosis factor-related apoptosis-inducing ligand through mitochondrial and caspase dependent pathway. Intest Res. 12:34–41. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 47 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sophonnithiprasert T, Nilwarangkoon S, Nakamura Y and Watanapokasin R: Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation. Int J Oncol 47: 2188-2196, 2015
APA
Sophonnithiprasert, T., Nilwarangkoon, S., Nakamura, Y., & Watanapokasin, R. (2015). Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation. International Journal of Oncology, 47, 2188-2196. https://doi.org/10.3892/ijo.2015.3204
MLA
Sophonnithiprasert, T., Nilwarangkoon, S., Nakamura, Y., Watanapokasin, R."Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation". International Journal of Oncology 47.6 (2015): 2188-2196.
Chicago
Sophonnithiprasert, T., Nilwarangkoon, S., Nakamura, Y., Watanapokasin, R."Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation". International Journal of Oncology 47, no. 6 (2015): 2188-2196. https://doi.org/10.3892/ijo.2015.3204