Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5

  • Authors:
    • Zakir Hossain
    • Tatsuya Sugawara
    • Takashi Hirata
  • View Affiliations

  • Published online on: January 4, 2013     https://doi.org/10.3892/or.2013.2223
  • Pages: 1201-1207
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Biofunctional marine compounds have recently received substantial attention for their nutraceutical characteristics. In this study, we investigated the apoptosis-inducing effects of sphingoid bases prepared from sea cucumber using human hepatoma HepG2 cells. Apoptotic effects were determined by cell viability assay, DNA fragmentation assay, caspase-3 and caspase-8 activities. The expression levels of apoptosis-inducing death receptor-5 (DR5) and p-AKT were assayed by western blot analysis, and mRNA expression of bax, GADD45 and PPARγ was assayed by quantitative RT-PCR analysis. Sphingoid bases from sea cucumber markedly reduced the cell viability of HepG2 cells. DNA fragmentation indicative of apoptosis was observed in a dose-dependent manner. The expression levels of the apoptosis inducer protein Bax were increased by the sphingoid bases from sea cucumber. GADD45, which plays an important role in apoptosis-inducing pathways, was markedly upregulated by sphingoid bases from sea cucumber. Upregulation of PPARγ mRNA was also observed during apoptosis induced by the sphingoid bases. The expression levels of DR5 and p-AKT proteins were increased and decreased, respectively, as a result of the effects of sphingoid bases from sea cucumber. The results indicate that sphingoid bases from sea cucumber induce apoptosis in HepG2 cells through upregulation of DR5, Bax, GADD45 and PPARγ and downregulation of p-AKT. Our results show for the first time the functional properties of marine sphingoid bases as inducers of apoptosis in HepG2 cells.

Introduction

The highest liver cancer rates are found in East and South East Asia and in Central and Western Africa. Developing countries contribute more than 80% of the cases. Chronic hepatitis B virus infection accounts for approximately 60% of the total liver cancer cases in developing countries and for approximately 23% of the cancer cases in developed countries, while hepatitis C virus infection accounts for approximately 33% of the total liver cancer cases in developing countries and for approximately 20% in developed countries (1,2). Diet is involved in the risk of hepatocellular carcinoma (HCC). It has been shown that red meat and saturated fat are associated with increased chronic liver disease and HCC. A diet that is rich in polyunsaturated fatty acids and, possibly, β-carotene can reduce the risk of HCC (3). Therefore, an appropriate diet might be a potential tool to prevent HCC.

Sphingolipids and their metabolites, including ceramides, sphingosine, and sphingosine-1-phosphate, play important roles in cellular signaling, such as in proliferation, apoptosis, cellular senescence, growth arrest and differentiation in a variety of cells (4,5). Ceramide is generated from sphingomyelin upon diverse apoptotic stimuli and transfers the death signal, which is the so-called second messenger. Ceramide induces apoptosis by inhibiting a variety of pro-growth signal kinases, including AKT and PKC, with direct activation of protein phosphatases (6). Sphingolipids have a dual role within the cell, acting as key structural components of membranes as well as serving as bioactive second messengers. Central to the chemical backbone of all sphingolipids, sphingosine represents an intermediate linking the metabolism of ceramide to sphingosine-1-phosphate. Cellular levels of sphingosine are controlled by the action of ceramide synthase or sphingosine kinase, which are responsible for the formation of ceramide and sphingosine-1-phosphate, respectively. Conversely, sphingosine may be formed by the action of ceramidase from ceramide or sphingosine-1-phosphatase from sphingosine-1- phosphate (7,8).

Physiologically active substances including glycosylceramide (GlcCer) and related compounds have been extracted from a variety of sea cucumber species (9,10). Dry sea cucumber contains ~200 mg glycosylceramides per 100 g (11). GlcCer used as a food ingredient has been isolated from various plant sources, but their content is very low (1–40 mg/100 g dry weight) (12). Thus, sea cucumber may be suitable as a dietary source of sphingolipids. However, the sphingoid base structures in sea cucumber are more complicated than those in mammals and there is little information regarding the nutritional function of these sphingoid bases that are not found in mammals (13,14). Sphingoid bases prepared from wheat flour cerebroside were found to suppress the growth of human colorectal cancer DLD-1 cells through apoptosis (15). Apoptosis was also induced in human colon cancer cells by plant and fungus sphingoid bases (16). In addition, sphingosine induced apoptosis in hippocampal neurons and astrocytes by activating caspase−3/−9 (17).

Peroxisome proliferator-activated receptor γ (PPARγ) has been identified and extensively studied. Several recent studies have shown that most human tumors express PPARγ, and there is evidence that PPARγ ligands have antitumor activity (1825). PPARγ has been the focus of research as a target molecule to prevent cancer (2628). The upregulation of PPARγ by bitter gourd seed oil containing 9-cis, 11-trans, 13-trans-conjugated linolenic acid was observed (29). In addition, the Bax and the growth arrest and DNA damage inducible gene 45 (GADD45) are known to have important roles in the pathways of growth inhibition and apoptosis induction in many types of cancer cells (30,31). Increased expression of GADD family genes is frequently observed in response to agents that trigger apoptosis (32,33). In this study, the possible pathways of apoptosis induced by sphingoid bases from sea cucumber in human hepatoma HepG2 cells were examined.

Materials and methods

Preparation of sphingoid bases

Sphingoid bases were prepared from sea cucumber and maize by a silica gel column after lipid extraction and saponification as previously described (11,34). 4,8-Sphingadienine (d18:2) from maize sphingoid bases and 4,8,10-sphingatrienine (d18:3) and 9-methyl-4,8,10-sphingatrienine (d19:3) from sea cucumber sphingoid bases (Fig. 1) were purified by HPLC (11,34).

Cell culture

HepG2 cells were obtained from the Human Science Research Resources Bank, Osaka, Japan. HepG2 cells were grown in 100-mm dishes containing Dulbecco's modified Eagle's medium with 100 IU/ml penicillin, 100 μg/ml streptomycin, and 10% heat-incubated fetal bovine serum in a 95% air-5% CO2 atmosphere at 37°C. Subculturing was carried out every 3 days at a concentration of 5×104 cells/ml.

Cell viability assay

Aliquots of 1×105 HepG2 cells were seeded into 96-well microplates containing 100 μl of growth medium/well. The plates were preincubated without any treatment for 24 h at 37°C. The sphingoid bases were dissolved in ethanol and diluted into the culture medium (the final concentration was 0.1%). Control was maintained with 0.1% ethanol as vehicle. The plates were incubated for another 24 h at 37°C with the sphingoid bases. The WST-1 reagent (3.3 mg/ml phosphate-buffered saline including 7% 1-methoxy PMS) was added at 10 μl in each well containing 100 μl of medium with the cells. The plates were incubated an additional 2 h at 37°C. Absorbance was measured at 650 nm on a microplate reader (Bio-Rad, Sunnyvale, CA, USA).

Enzyme-linked immunosorbent assay (ELISA) for apoptosis

DNA fragmentation was measured by a biotin-labeled anti-histone-antibody and a peroxidase-conjugated anti-DNA antibody cell death detection ELISA kit (Roche Diagnostics GmbH, Mannheim, Germany). In brief, HepG2 cells (2×103 cells/well) were plated in 96-well plates with 100 μl of medium and incubated for 24 h. After a 24-h incubation, cells were treated with the sphingoid bases for 24 h. The level of cytoplasmic histone-associated DNA fragments (mono- and oligonucleosomes) in HepG2 cells was expressed as an enrichment factor, calculated using the following formula: Enrichment factor = Absorbance of sample (treated)/Absorbance of corresponding control.

Caspase-3 and -8 activities

Caspase-3 and -8 activities were measured using a caspase-3 and -8 colorimetric assay kit (BioVision Research Products, Mountain View, CA, USA). HepG2 cells were treated with the sphingoid bases for 24 h, then harvested and lysed with chilled cell lysis buffer and incubated on ice for 10 min. Cell lysates were clarified by centrifugation at 10,000 × g for 1 min at 4°C. The protein concentration in the supernatant was quantified following the method of Lowry et al(35), and 100 μg of protein was added to 50 μl of lysis buffer for each assay. Fifty microliters of 2X reaction buffer (containing 10 mM dithiothreitol, DTT) was added to each sample. For caspase-3 and -8 assays, samples were incubated with Asp-Glu-Val-Asp-p-nitroanilide (DEVD-pNA) substrate for 2 h at 37°C. Optical density reading was taken at a wavelength of 405 nm on a microplate reader.

Quantitative real-time RT-PCR for mRNA analysis

Quantitative real-time PCR was performed to measure the mRNA levels of the studied genes using a Bio-Rad Thermal Cycler (Bio-Rad Laboratories, Hercules, CA, USA) using SYBR-Green PCR reagents. Total RNA was extracted from the HepG2 cells treated with sphingoid bases using Sepasol-RNA I Super (Nacalai Tesque Inc., Kyoto, Japan). The following primers were used: PPAR-γ, 5′-AAGGCCATTTTC TCAAACGA-3′ (sense), 5′-TGCAACCACTGGATCTGT TC-3′ (antisense); GADD45, 5′-CAGAAGACCGAAAGG ATGGA-3′ (sense), 5′-CAGAAGACCGAAAGGATGGA-3′ (antisense); Bax, 5′-TCTGACGGCAACTTCAACTG-3′ (sense), 5′-CCTGTAATCCCAGGTCCTTG-3′ (antisense). The primer pair of GAPDH was 5′-TGGGATCGAGTGAAG GACCT-3′ (sense) and 5′-CTCCTCCTGCCACTTCTTC TG-3′ (antisense). The reaction solution (20 μl final volume) contained 6 μl of sample, 10 μl SYBR-Green dye and 2 μl of each primer. The thermal cycling conditions were as follows: 48°C for 30 min to prevent carrying over of DNA, an initial denaturation of 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 1 sec and an annealing temperature of 55°C for 1 min.

Western blot analysis

Cells were grown in 100-mm culture dishes, starved of serum for 48 h and treated with sphingoid bases. They were then lysed in cell lysis buffer [62.5 mM Tris-HCl (pH 6.8), 2% SDS, 5% mercaptoethanol, 2 mM phenylmethylsulfonyl fluoride, protease inhibitors (Roche, Mannheim, Germany)], 1 mM Na3VO4, 50 mM NaF and 10 mM EDTA. Ten micrograms of protein per lane was separated by SDS-polyacrylamide gel electrophoresis and blotted onto nitrocellulose membranes, which were then saturated with 5% dried milk in Tris-buffered saline containing 0.4% Tween-20. The blots were incubated with the anti-AKT and anti-phospho-AKT antibodies at a dilution of 1:1,000, and then further incubated with horseradish peroxidase-conjugated secondary antibody. The blots were also incubated with the anti-DR5 and anti-actin antibodies at a dilution of 1:1,000, and then further incubated with horseradish peroxidase-conjugated secondary antibody. Bound antibodies were detected using an Enhanced Chemiluminescence Plus kit (Amersham International, Little Chalfont, UK). The results were analyzed using a Fujifilm visualizer (LAS-3000, Fujifilm Corp., Tokyo, Japan).

Results

Inhibition of HepG2 cell proliferation by sphingoid bases

The effect of sphingoid bases from sea cucumber on the inhibition of HepG2 cell proliferation at various concentrations was tested at 24 h. Sphingoid bases inhibited cell proliferation in a dose-dependent manner. There was significant growth inhibition with 20 or 40 μM of the sphingoid bases when compared with the control (Fig. 2). The growth inhibition with sphingosine and sphingoid bases from sea cucumber was comparable (Fig. 2).

Apoptosis induction in HepG2 cells treated with sphingoid bases

To characterize the apoptosis induced by sphingoid bases, an experiment was conducted using a quantitative cell death ELISA. The enrichment factors in cells treated with 20 μM sphingoid bases (18:3 or 19:3) were increased by 5.0-fold in comparison to the control (Fig. 3). Sphingosine and sphingoid bases from sea cucumber significantly increased caspase-3 and -8 activities compared to the control (Fig. 4).

Bax, GADD45 and PPARγ expression in HepG2 cells treated with sphingoid bases

The expression level of Bax, GADD45 and PPARγ mRNA in HepG2 cells was increased following a 24-h incubation with 20 μM sphingoid bases. Treatment with the sphingoid bases of sea cucumber increased the expression of Bax, GADD45 and PPARγ by only 3.0-, 2.0 and 2.5-fold in the HepG2 cells whereas sphingosine increased their expression levels by 6.0-, 2.0- and 3.0-fold respectively, compared with the control (Fig. 5).

Expression of p-AKT decreased and expression of DR5 increased due to the effects of treatment with the sphingoid bases

To clarify whether the expression levels of p-AKT are associated with the effects of sphingoid bases, we measured expression of AKT and p-AKT by immunoblot analysis. AKT, which is the downstream effector of PI3-kinase was found to be phosphorylated. We found that sphingoid bases induced p-AKT inhibition and triggered apoptosis (Fig. 6A). To clarify whether the expression levels of DR5 are associated with the effects of sphingoid bases, we measured the expression of DR5 by immunoblot analysis. We found that treatment with the sphingoid bases increased expression of DR5 in the HepG2 cells (Fig. 6B).

Discussion

Dietary sphingolipids have gained attention for their potential to protect against the development of colon cancer. Sphingolipids are a family of compounds that have a long-chain (sphingoid) base backbone and include free sphingoid bases (sphingosine and sphinganine), ceramides, sphingomyelins, cerebrosides, sulfatides and gangliosides (36). Several studies have been conducted with milk sphingomyelin, synthetic sphingomyelin, synthetic dihydrosphingomyelin, as well as milk glycosphingolipids to determine 1,2-dimethylhydrazine (DMH)-induced colon cancer protection in CF1 mice. Sphingomyelin at 0.1% of the diet caused a higher percentage of adenomas and a lower percentage of more advanced adenocarcinomas. Notably, synthetic dihydrosphingomyelin (N-palmitoyl dihydrosphingomyelin) more potently reduced the number of aberrant colonic foci than synthetic sphingomyelin (N-palmitoylsphingomyelin) and milk sphingomyelin, suggesting that the 4,5-trans double bond, which is absent in dihydrosphingomyelin, is not required for the suppression of colon carcinogenesis (37). In the current study, we found that the growth suppression effects of sphingosine and sphingoid bases from sea cucumber (mixture of bases, 18:3 and 19:3), and a maize sphingoid base (18:2) in HepG2 cells were comparable. The long chain structure and 3 double bonds present in the bases of sea cucumber may be indispensable for the apoptotic effect of sphingoid bases on cancer cells.

Activation of the caspase cascade, a family of cysteine proteases, is required for apoptotic signaling. In addition to their crucial role as the energy-producing center of the cell, the mitochondria are central to the convergence of the intrinsic or extrinsic cellular signal transduction pathways that result in apoptosis. Essential to this process is the release of cytochrome c and other mitochondrial-related proteins that are normally confined to the mitochondrial intermembrane space by antiapoptotic Bcl-2 family members (38,39). Recent data suggest that oligomeric Bax is a component of the mitochondrial apoptosis-induced channel, and following their interaction, a selective cytochrome c release pore is formed (40). However, the consensus of these theories includes a requirement for Bax/Bak activation to promote pore opening and cytochrome c release. Our data showed an increased mRNA expression of Bax. The release of cytochrome c and other apoptogenic factors from injured mitochondria have been shown to activate caspases (41), and the mitochondrial integrity appears to be regulated, in part, by members of the Bcl-2 family. In response to apoptotic signals, Bax, a proapoptotic member of this family, is redistributed from the cytosol to the mitochondria, where it decreases membrane potential leading to cytochrome c release and caspase activation (42).

Little is known concerning the direct action of sphingolipids on the expression of genes in hepatocytes. In the present study, we also found the increased mRNA expression of PPARγ in HepG2 cells. PPARγ displayed proapoptotic activity, as has been demonstrated in the HL-60 cell line (43) and in the lymphoblastic leukemia cell line (44). PPARγ liagands 15d-PGJ2 and troglitazone suppress DNA synthesis and induce apoptosis in HT-29 colon cancer cells (45). Troglitazone inhibited the growth of liver cancer cells PLC/PRF/5, HepG2 and HuH-7, by inducing apoptosis through caspase-3 activation (46). GADD45α overexpression in normal human fibroblasts and human cancer cells was found to cause G2-M arrest (4749). GADD45α-dependent induction of apoptosis has been observed frequently in cancer cell lines (50,51), but its role in apoptosis induction in normal cells has been controversial. GADD45α has been shown to induce cell cycle arrest in normal fibroblasts but failed to induce apoptosis (48). Others, however, have shown Gadd45α-dependent induction of apoptosis in normal epithelial cells (52). In the present study, we showed that GADD45 expression correlated with a significant increase in apoptosis in HepG2 cells. In this study, we characterized the functional role of AKT in the apoptotic pathway related to the sphingoid bases and we found that inhibition of the activation of AKT is one of the mechanisms by which sphingoid bases induced apoptosis. Our findings concurred with the results of Chang et al(5) who found that sphingosine induced apoptosis by inhibiting AKT expression. AKT is known to inhibit apoptosis by inactivating proapoptotic proteins such as Bad, procaspase-9, and forkhead and by activating antiapoptotic proteins such as NF-κB and cyclic adenosine monophosphate (cAMP)-response element binding protein (53,54). A previous study showed that dephosphorylation of AKT activates the proapoptotic function of Bax (55). Based on these observations, inhibition of AKT phosphorylation by sphingoid bases may lead to inhibition of Bax phosphorylation that promotes Bax translocation into the mitochondrial membrane.

Based on our results, we propose an outline of the sphingoid base-induced apoptotic signaling pathway (Fig. 7). Sphingoid bases induce DNA fragmentation resulting in the upregulation of GADD45, which induces cell cycle arrest in the G2/M phase giving cells the chance to repair DNA damage. In contrast, when DNA damage is not repaired, cells execute the apoptotic pathway. GADD45, DR5 and Bax are upregulated and p-AKT is downregulated, leading to the disruption of the mitochondrial membrane, which in turn causes cytochrome c release from the intramitochondria into the cytosol, thus activating caspase-3 and -8, which then cleaves the death substrates, leading to apoptosis. Sea cucumber is consumed as a food in different regions of the world. Prevention of cancer by supplementing the diet with naturally occurring compounds is considered as a potential approach to reduce cancer.

Acknowledgements

This study was supported by the Japan Society for the Promotion of Science (JSPS) and the Program for Promotion and Applied Research for Innovations in the Bio-Oriented Industry (BRAIN).

Abbreviations:

DR

death receptor

GADD

growth arrest DNA damage

PPAR

peroxisome proliferator-activated receptor

GlcCer

glycosylceramides

References

1 

Parkin DM: The global health burden of infection-associated cancers in the year 2002. Int J Cancer. 118:3030–3044. 2006.PubMed/NCBI

2 

But DY, Lai CL and Yuen MF: Natural history of hepatitis-related hepatocellular carcinoma. World J Gastroenterol. 14:1652–1656. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Liu S, Xie J, Yin J, Zhang H, Zhang Q, Pu R, Li C, Ni W, Wang H and Cao G: A matched case-control study of hepatitis B virus mutations in the preS and core promoter regions associated independently with hepatocellular carcinoma. J Med Virol. 83:45–53. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Huwiler A, Kolter T, Pfeilschifter J and Sandhoff K: Physiology and pathophysiology of sphingolipid metabolism and signaling. Biochim Biophys Acta. 1485:63–99. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Chang HC, Tsai LH, Chuang LY and Hung WC: Role of AKT kinase in sphingosine-induced apoptosis in human hepatoma cells. J Cell Physiol. 188:188–193. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Ruvolo PP: Intracellular signal transduction pathways activated by ceramide and its metabolites. Pharmacol Res. 47:383–392. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Maceyka M, Payne SG, Milstien S and Spiegel S: Sphingosine kinase, sphingosine-1-phosphate, and apoptosis. Biochim Biophys Acta. 1585:193–201. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Orgetmen B and Hannun YA: Biologically active sphingolipids in cancer pathogenesis and treatment. Nat Rev Cancer. 4:604–616. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Yamada K: Chemo-pharmaceutical studies on the glycosphingolipid constituents from echinoderm, sea cucumbers, as medicinal materials. Yakugaku Zasshi. 122:1133–1143. 2002.(In Japanese).

10 

Yamada K, Hamada A, Kisa F, Miyamoto T and Higuchi R: Constituents of holothuroidea, 13. Structure of neuritogenic active ganglioside molecular species from the sea cucumber Stichopus chloronotus. Chem Pharm Bull. 51:46–52. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Sugawara T, Zaima N, Yamamoto A, Sakai S, Noguchi R and Hirata T: Isolation of sphingoid bases of sea cucumber cerebrosides and their cytotoxicity against human colon cancer cells. Biosci Biotechnol Biochem. 70:2906–2912. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Sugawara T and Miyazawa T: Separation and determination of glycolipids from edible plant sources by high-performance liquid chromatography and evaporative light-scattering detection. Lipids. 34:1231–1237. 1999.PubMed/NCBI

13 

Sugawara T, Aida K, Duan J and Hirata T: Analysis of glucosylceramides from various sources by liquid chromatography-ion trap mass spectrometry. J Oleo Sci. 59:387–394. 2010.PubMed/NCBI

14 

Hossain Z, Sugawara T, Aida K and Hirata T: Effect of dietary glucosylceramides from sea cucumber on plasma and liver lipids in cholesterol-fed mice. Fish Sci. 77:1081–1085. 2011. View Article : Google Scholar

15 

Sugawara T, Kinoshita M, Ohnishi M and Miyazawa T: Apoptosis induction by wheat-flour sphingoid bases in DLD-1 human colon cancer cells. Biosci Biotech Biochem. 66:2228–2231. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Aida K, Kinoshita M, Sugawara T, Ono J, Miyazawa T and Ohnishi M: Apoptosis inducement by plant and fungus sphingoid bases in human colon cancer cells. J Oleo Sci. 53:503–510. 2004. View Article : Google Scholar

17 

Kanno T and Nishizaki T: Sphingosine induces apoptosis in hippocampal neurons and astrocytes by activating caspase−3/−9 via a mitochondrial pathway linked to SDK/14-3-3 protein/Bax/cytochrome c. J Cell Physiol. 226:2329–2337. 2011.PubMed/NCBI

18 

Clay CE, Atsumi GI, High KP and Chilton FH: Early de novo gene expression is required for 15-deoxy-Delta 12,14-prostaglandin J2-induced apoptosis in breast cancer cells. J Biol Chem. 276:47131–47135. 2001. View Article : Google Scholar

19 

Debrock G, Vanhentenrijk V, Sciot R, Debiec-Rychter M, Oyen R and Van Oosterom A: A phase II trial with rosiglitazone in liposarcoma patients. Br J Cancer. 89:1409–1412. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Grommes C, Landreth GE and Heneka MT: Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 5:419–429. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Date M, Fukuchi K, Morita S, Takahashi H and Ohura K: 15-Deoxy-delta12,14-prostaglandin J2, a ligand for peroxisome proliferator-activated receptor-gamma, induces apoptosis in human hepatoma cells. Liver Int. 23:460–466. 2003. View Article : Google Scholar

22 

Han S, Sidell N, Fisher PB and Roman J: Up-regulation of p21 gene expression by peroxisome proliferator-activated receptor gamma in human lung carcinoma cells. Clin Cancer Res. 10:1911–1919. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Yang FG, Zhang ZW, Xin DQ, Shi CJ, Wu JP, Guo YL and Guan YF: Peroxisome proliferator-activated receptor gamma ligands induce cell cycle arrest and apoptosis in human renal carcinoma cell lines. Acta Pharmacol Sin. 26:753–761. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Shigeto T, Yokoyama Y, Xin B and Mizunuma H: Peroxisome proliferator-activated receptor alpha and gamma ligands inhibit the growth of human ovarian cancer. Oncol Rep. 18:833–840. 2007.PubMed/NCBI

25 

Lin MS, Chen WC, Bai X and Wang YD: Activation of peroxisome proliferator-activated receptor gamma inhibits cell growth via apoptosis and arrest of the cell cycle in human colorectal cancer. J Dig Dis. 8:82–88. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Sporn MB, Suh N and Mangelsdorf DJ: Prospects for prevention and treatment of cancer with selective PPARγ modulators (SPARMs). Trends Mol Med. 7:395–400. 2001.PubMed/NCBI

27 

Gupta RA and Dubois RN: Controversy: PPARγ as a target for treatment of colorectal cancer. Am J Physiol. 283:G266–G269. 2002.

28 

Girnun GD, Smith WM, Drori S, Sarraf P, Mueller E, Eng C, Nambiar P, Rosenberg DW, Bronson RT, Edelmann W, Kucherlapati R, Gonzalez FJ and Spiegelman BM: APC- dependent suppression of colon carcinogenesis by PPARγ. Proc Natl Acad Sci USA. 99:13771–13776. 2002.PubMed/NCBI

29 

Yasui Y, Hosokawa M, Sahara T, Suzuki R, Ohgiya S, Kohno H, Tanaka T and Miyashita K: Bitter gourd seed fatty acid rich in 9c,11t,13t-conjugated linolenic acid induces apoptosis and up-regulates the GADD45, p53 and PPARgamma in human colon cancer Caco-2 cells. Prostaglandins Leukot Essent Fatty Acids. 73:113–119. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Han C, Demetris AJ, Michalopoulos GK, Zhan Q, Shelhamer JH and Wu T: PPARγ ligands inhibit cholangiocarcinoma cell growth through p53-dependent GADD45 and p21 WAF1/Cip1 pathway. Hepatology. 38:167–177. 2003.

31 

Nagamine M, Okumura T, Tanno S, Sawamukai M, Motomura W, Takahashi N and Kohgo Y: PPARγ ligand-induced apoptosis through a p53-dependent mechanism in human gastric cancer cells. Cancer Sci. 94:338–343. 2003.

32 

Okumura T, Nakamura M, Takata Y, Watanabe S, Kitami Y and Hiwada K: Troglitazone induces apoptosis via the p53 and Gadd45 pathway in vascular muscle cells. Eur J Pharmacol. 407:227–235. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Yin F, Bruemmer D, Blaschke F, Hsueh WA, Law RE and Van Herle AJ: Signaling pathways involved in induction of GADD45 gene expression and apoptosis by troglitazone in human MCF-7 breast carcinoma cells. Oncogene. 23:4614–4623. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Aida K, Kinoshita M, Tanji M, Sugawara T, Tamura M, Ono J, Ueno N and Ohnishi M: Prevention of aberrant crypt foci formation by dietary maize and yeast cerebrosides in 1,2-dimethylhydrazine-treated mice. J Oleo Sci. 54:45–49. 2005. View Article : Google Scholar

35 

Lowry OH, Rosebrough NJ, Farr AL and Randall RJ: Protein measurement with the Folin phenol reagent. J Biol Chem. 193:265–275. 1951.PubMed/NCBI

36 

Merrill AH Jr, Schmelz EM, Dillehay DL, Spiegel S, Shayman JA, Schroeder JJ, Riley RT, Voss KA and Wang E: Sphingolipids: the enigmatic lipid class: biochemistry, physiology, and pathophysiology. Toxicol Appl Pharmacol. 142:208–225. 1997. View Article : Google Scholar : PubMed/NCBI

37 

Schmelz EM, Sullards MS, Dillehay DL and Merrill AH Jr: Colonic cell proliferation and aberrant crypt foci formation are inhibited by dairy glycosphingolipids in 1,2-dimethylhydrazine-treated CF1 mice. J Nutr. 130:522–527. 2000.PubMed/NCBI

38 

Sprick MR and Walczak H: The interplay between the Bcl-2 family and death receptor-mediated apoptosis. Biochim Biophys Acta. 1644:125–132. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Willis SN and Adams JM: Life in the balance: how BH3-only proteins induce apoptosis. Curr Opin Cell Biol. 17:617–625. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Dejean LM, Martinez-Caballero S, Guo L, Hughes C, Teijido O, Ducret T, Ichas F, Korsmeyer SJ, Antonsson B, Jonas EA and Kinnally KW: Oligomeric Bax is a component of the putative cytochrome c release channel MAC, mitochondrial apoptosis-induced channel. Mol Biol Cell. 16:2424–2432. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Scaffidi C, Schmitz I, Zha J, Korsmeyer SJ, Krammer PH and Peter ME: Differential modulation of apoptosis sensitivity in CD95 type I and type II cells. J Biol Chem. 274:22532–22538. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Gross A, Jockel J, Wei MC and Korsmeyer SJ: Enforced dimerization of BAX results in its translocation, mitochondrial dysfunction and apoptosis. EMBO J. 17:3878–3885. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Laurora S, Pizzimenti S, Briatore F, Fraioli A, Maggio M, Reffo P, Ferretti C, Dianzani MU and Barrera G: Peroxisome proliferator-activated receptor ligands affect growth-related gene expression in human leukemic cells. J Pharmacol Exp Ther. 305:932–942. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Liu H, Zang C, Fenner MH, Liu D, Possinger K, Koeffler HP and Elstner E: Growth inhibition and apoptosis in human Philadelphia chromosome-positive lymphoblastic leukemia cell lines by treatment with the dual PPARα/γ ligand TZD18. Blood. 107:3683–3692. 2006.PubMed/NCBI

45 

Shimada T, Kojima K, Yoshiura K, Hiraishi H and Terano A: Characteristics of the peroxisome proliferator activated receptor γ (PPARγ) ligand induced apoptosis in colon cancer cells. Gut. 50:658–664. 2002.

46 

Toyoda M, Takagi H, Horiguchi N, Kakizaki S, Sato K, Takayama H and Mori M: A ligand for peroxisome proliferator activated receptor gamma inhibits cell growth and induces apoptosis in human liver cancer cells. Gut. 50:563–567. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Wang W, Huper G, Guo Y, Murphy SK, Olson JA Jr and Marks JR: Analysis of methylation-sensitive transcriptome identifies GADD45α as a frequently methylated gene in breast cancer. Oncogene. 24:2705–2714. 2005.PubMed/NCBI

48 

Mita H, Tsutsui J, Takekawa M, Witten EA and Saito H: Regulation of MTK1/MEKK4 kinase activity by its N-terminal autoinhibitory domain and GADD45 binding. Mol Cell Biol. 22:4544–4555. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Tront JS, Hoffman B and Liebermann DA: Gadd45α suppresses Ras driven mammary tumorigenesis by activation of c-Jun NH2-terminal kinase and p38 stress signaling resulting in apoptosis and senescence. Cancer Res. 66:8448–8454. 2006.

50 

Zerbini LF and Libermann TA: ADD45 deregulation in cancer: frequently methylated tumor suppressors and potential therapeutic targets. Clin Cancer Res. 11:6409–6413. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Tong T, Ji J, Jin S, Li X, Fan W, Song Y, Wang M, Liu Z, Wu M and Zhan Q: Gadd45α expression induces Bim dissociation from the cytoskeleton and translocation to mitochondria. Mol Cell Biol. 25:4488–4500. 2005.

52 

Hildesheim J, Bulavin DV, Anver MR, Alvord WG, Hollander MC, Vardanian L and Fornace AJ Jr: Gadd45α protects against UV irradiation-induced skin tumors, and promotes apoptosis and stress signaling via MAPK and p53. Cancer Res. 62:7305–7315. 2002.

53 

Zhou H, Li XM, Meinkoth J and Pittman RN: Akt regulates cell survival and apoptosis at a postmitochondrial level. J Cell Biol. 151:483–494. 2000. View Article : Google Scholar : PubMed/NCBI

54 

Testa JR and Bellacosa A: Akt plays a central role in tumorigenesis. Proc Natl Acad Sci USA. 98:10983–10985. 2001. View Article : Google Scholar : PubMed/NCBI

55 

Xin M and Deng X: Nicotine inactivation of proapoptotic function of Bax through phosphorylation. J Biol Chem. 280:10781–10789. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hossain Z, Sugawara T and Hirata T: Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5. Oncol Rep 29: 1201-1207, 2013
APA
Hossain, Z., Sugawara, T., & Hirata, T. (2013). Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5. Oncology Reports, 29, 1201-1207. https://doi.org/10.3892/or.2013.2223
MLA
Hossain, Z., Sugawara, T., Hirata, T."Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5". Oncology Reports 29.3 (2013): 1201-1207.
Chicago
Hossain, Z., Sugawara, T., Hirata, T."Sphingoid bases from sea cucumber induce apoptosis in human hepatoma HepG2 cells through p-AKT and DR5". Oncology Reports 29, no. 3 (2013): 1201-1207. https://doi.org/10.3892/or.2013.2223