Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1226-7155(Print)
ISSN : 2287-6618(Online)
International Journal of Oral Biology Vol.39 No.3 pp.159-167
DOI : https://doi.org/10.11620/IJOB.2014.39.3.159

Apoptotic Effect of Co-treatment with Curcumin and Cisplatin on SCC25 Human Tongue Squamous Cell Carcinoma Cell Line

Hyeon-Jin Sohn, In-Ryoung Kim, Yong-Ho Kim, Gyoo-Cheon Kim, Hyun-Ho Kwak, Bong-Soo Park*
Department of Oral Anatomy, School of Dentistry, Pusan National University
Correspondence to: Bong-Soo Park, Department of Oral Anatomy, School of Dentistry, Pusan National University, Yangsan, 626-870, Korea, Tel.: +82-51-510-8242, Fax: +82-51-510-8241, E-mail: parkbs@pusan.ac.kr
August 14, 2014 September 3, 2014 September 3, 2014

Abstract

Curcumin is a widely used flavoring agent in food, and it has been reported to inhibit cell growth, to induce apoptosis, and to have antitumor activity in many cancers. Cisplatin is one of the most potent known anticancer agents and shows significant clinical activity against a variety of solid tumors. This study was undertaken to investigate the synergistic apoptotic effects of co-treatment with curcumin and cisplatin on human tongue SCC25 cells. To investigate whether the co-treatment efficiently reduced the viability of the SCC25 cells compared with the two treatments separately, an MTT assay was conducted. The induction and the augmentation of apoptosis were confirmed by DNA electrophoresis, Hoechst staining, and an analysis of DNA hypoploidy. Western blot, MMP and immunofluorescence tests were also performed to evaluate the expression levels and the translocation of apoptosis-related proteins following the co-treatment.

In this study, following the co-treatment with curcumin and cisplatin, the SCC25 cells showed several forms of apoptotic manifestation, such as nuclear condensation, DNA fragmentation, reduction of MMP, increased levels of Bax, decreased levels of Bcl-2, and decreased DNA content. In addition, they showed a release of cytochrome c into the cytosol, translocation of AIF and DFF40 (CAD) to the nuclei, and activation of caspase-7, caspase-3, PARP, and DFF45 (ICAD). In contrast, separate treatments of 5 μM of curcumin or 4 μg/ml of cisplatin, for 24 hours, did not induce apoptosis.

Therefore, our data suggest that combination therapy with curcumin and cisplatin could be considered as a novel therapeutic strategy for human oral squamous cell carcinoma.


초록


    Pusan National University

    Introduction

    Cells undergoing apoptosis usually exhibit characteristic changes, including nuclear condensation and degradation of DNA into oligonucleosomal fragments [1]. Apoptotic cell death is thought to result ultimately from the proteolytic actions of caspase [2], and alterations in mitochondrial function play a key role in the regulation of apoptosis [3]. Moreover, the proteasome system has been implicated as a negative or positive mediator of apoptosis. The proteasome pathway is known to work mostly upstream of mitochondrial alterations and caspase activation [4].

    Carcinoma of the oral cavity, especially oral squamous cell carcinoma (OSCC), is one of the leading causes of cancer-related death, affecting nearly 500,000 patients annually worldwide. OSCC is a major malignancy, which remains incurable with current therapies [5]. OSCC patients are treated with classical modalities consisting of surgery, radiotherapy, and/or chemotherapy. Given the high mortality rates with OSCC, new therapeutic approaches continue to be investigated. The most promising approach is the use of natural agents with known anticancer effects.

    Curcumin is a natural component of the spice turmeric. It is derived from the rhizome of the Indian plant Curcuma longa and traditionally has been used in the treatment of digestion problems, parasites, gallstones, and arthritis in Southeast Asia [6]. Several studies have reported that curcumin has anti-oxidant, anti-inflammatory and anti-cancer effects in humans [7-9]. Other studies reported that curcumin inhibits cell proliferation and induces apoptosis in many cancer cells [10-12]. Cisplatin, which belongs to a class of platinum containing anti-cancer compounds, is an extremely potent anticancer agent, showing significant clinical activity against a variety of solid tumors [13]. It is a representative anticancer drug used to treat certain types of head and neck cancer, cervical carcinoma, lung cancer, neurological cancers, and a wide variety of other cancers. However, resistance to cisplatin remains a significant barrier to the survival of cancer patients.

    Recent studies have demonstrated that co-treatment of an antitumor agent with a natural product with anticancer effects may be a potential therapeutic strategy to reduce the extent and the severity of cancer treatment-related toxicity [14-18]. A systemic study of synergistic apoptotic effects of co-treatment with curcumin and cisplatin on a human oral squamous carcinoma cell line is lacking. Therefore, the present study investigated the synergistic apoptotic effect of co-treatment with curcumin and the anticancer drug cisplatin on a human oral squamous cell carcinoma (SCC25) cell line.

    Materials and methods

    Reagents

    The following reagents were obtained commercially: curcumin, cisplatin, dimethyl sulfoxide (DMSO), Hoechst 33342, RNase A, proteinase K, aprotinin, leupeptin, PMSF, thiazolyl blue tetrazolium bromide and propidium iodide (PI) were from Sigma (St. Louis, MO, USA); Mouse monoclonal anti-human caspase-3, caspase-7, poly(ADP-ribose) polymerase (PARP), cytochrome c, apoptosis-inducing factor (AIF) antibodies, and rabbit polyclonal anti-human DFF40 (CAD), DFF45 (ICAD), GAPDH antibodies, and FITC-conjugated goat anti-mouse and anti-rabbit IgGs were from Santa Cruz Biotechnology (Santa Cruz, CA, USA); HRP-conjugated sheep anti-mouse and anti-rabbit IgGs were from Amersham GE Healthcare (Little Chalfront, UK). 5,5',6,6'-tetrachloro- 1,1',3,3'-tetraethylbenzimidazol carbocyanine iodide (JC-1) was from Molecular Probes (Eugene, OR, USA). Dulbecco's modified Eagle's medium (DMEM) and FBS were from Gibco (Gaithersburg, MD, USA). SuperSignal West Femto enhanced chemiluminescence Western blotting detection reagent was from Pierce (Rockford, IL, USA).

    Cell culture

    The SCC25 human oral saquamous cell carcinoma cell line was purchased from ATCC (Rockville, USA). Cells were maintained at 37°C with 5% CO2 in air atmosphere in DMEM/F12 with 4 mM L-glutamine, 1.5 g/l sodiumbicarbonate, 4.5 g/l glucose and 1.0 mM, sodiumpyruvate supplemented with 10% FBS.

    Treatment of curcumin and cisplatin

    Stock solutions of the curcumin (100 mM) and cisplatin (25 mg/ml) made by dissolving them in DMSO were kept frozen at -20°C until use. Twenty four hours after SCC25 cells were subcultured, the original medium was removed. The cells were washed with PBS and then incubated in the same fresh medium. Since single treatment of 5 μM curcumin and 4 μg/ml cisplatin for 24 h showed slight induction of cell death in MTT assay, this single concentration was utilized for further assessment of apoptosis for co-treatment.

    MTT assay

    SCC25 cells were placed in a 96-well plate and were incubated for 24 h. Then, the cells were treated with each drugs for various time points. After cells were treated with 500 μg/ml of thiazolyl blue tetrazolium bromide (MTT solution), they were incubated at 37°C with 5% CO2 for 4 h. And then the medium was aspirated and formed formazan crystals were dissolved in DMSO. Cell viability was measured by an ELISA reader (Tecan, Männedorf, Switzerland) at 570 nm excitatory emission wavelength.

    Hoechst staining

    Cells were harvested and cell suspension was centrifuged onto a clean, fat-free glass slide with a cytocentrifuge. The samples were stained in 4 μg/ml Hoechst 33342 for 30 min at 37°C and fixed for 10 min with 4% paraformaldehyde.

    DNA electrophoresis

    2 x 106 cells were resuspended in 1.5 mL of lysis buffer [10 mM Tris (pH 7.5), 10 mM EDTA (pH 8.0), 10 mM NaCl and 0.5% SDS] into which proteinase K (200 μg/ml) was added. After samples were incubated overnight at 48°C, 200 μl of ice cold 5 M NaCl was added and the supernatant containing fragmented DNA was collected after centrifugation. The DNA was then precipitated overnight at -20°C in 50% isopropanol and RNase A-treated for 1 h at 3 7°C. The DNA from 106 cells (15 μl) was equally loaded on each lane of 2% agarose gels in Tris-acetic acid/EDTA buffer containing 0.5 μg/ml ethidium bromide at 50 mA for 1.5 h.

    Western blot analysis

    Cells (2 x 106) were washed twice in ice-cold PBS, resuspended in 200 μl ice-cold solubilizing buffer [300 mM NaCl, 50 mM Tris-Cl (pH 7.6), 0.5% Triton X-100, 2 mM PMSF, 2 μg/ml aprotinin and 2 μg/ml leupeptin] and incubated at 4°C for 30 min. The lysates were centrifuged at 14,000 rpm for 15 min at 4°C. Protein concentrations of cell lysates were determined with Bradford protein assay (Bio-Rad, Richmond, CA, USA) and 50 μg of proteins were loaded onto 7.5-15% SDS/PAGE. The gels were transferred to nitrocellulose membrane (Amersham GE Healthcare, Little Chalfont, UK) and reacted with each antibody. Immunostaining with antibodies was performed using SuperSignal West Femto enhanced chemiluminescence substrate and detected with Alpha Imager HP (Alpha Innotech, Santa Clara, USA).

    Immunofluorescence staining

    Cells were cytocentrifuged and fixed for 10 min in 4% paraformaldehyde, incubated with each primary antibody for 1 h, washed 3 times for 5 min, and then incubated with FITC-conjugated secondary antibody for 1 h at room temperature. Cells were mounted with PBS. Fluorescent images were observed and analyzed under Zeiss LSM 700 laser-scanning confocal microscope (Göettingen, GER).

    Assay of mitochondrial membrane potential (MMP)

    JC-1 was added directly to the cell culture medium (1 μM final concentration) and incubated for 15 min. The medium was then replaced with PBS. Flow cytometry to measure MMP was performed on a CYTOMICS FC500 flow cytometry (Beckman Coulter, Brea, CA, USA). Data were acquired and analyzed using CXP software version 2.2.

    Quantification of DNA hypoploidy by flow cytometry

    After treatment for 24 h, cells were harvested by trypsinization and ice cold 95% ethanol with 0.5% Tween 20 was added to the cell suspensions to a final concentration of 70% ethanol. Fixed cells were pelleted, and washed in 1% BSA-PBS solution. Cells were resuspended in 1 ml PBS containing 20 μg/ml RNase A, incubated at 4°C for 30 min, washed once with BSA-PBS, and resuspended in PI solution (10 μg/ml). After cells were incubated at 4°C for 5 min in the dark, DNA content were measured on a CYTOMICS FC500 flow cytometry system (Beckman Coulter, FL, CA, USA) and data was analyzed using the Multicycle software which allowed a simultaneous estimation of cell-cycle parameters and apoptosis.

    Results

    Co-treatment with curcumin and cisplatin augmented the reduction in viability of SCC25 cells

    To investigate whether co-treatment with curcumin and cisplatin reduced the viability of the SCC25 cells, an MTT assay was conducted. Single treatment of curcumin (5 μM) or cisplatin (4 μg/ml) for 24 h slightly reduced the viability of the SCC25 cells (curcumin, 94.6%; cisplatin, 93.4%). Co-treatment with curcumin and cisplatin significantly reduced the cell viability (45.2%) compared to the single treatment (Fig. 1). The concentration of DMSO used in this study had no effect on SCC25 cell proliferation in our preliminary studies.

    Co-treatment with curcumin and cisplatin augmented the nuclear condensation and fragmantation in SCC25 cells

    To explore whether nuclear condensation and fragmentation were induced by the co-treatment, Hoechst staining, a hallmark of apoptosis, was conducted. Hoechst staining showed slight nuclear condensation in response to the single treatment of curcumin and cisplatin. However, co-treatment with curcumin and cisplatin resulted in a variety of condensed and fragmented nuclei (Fig. 2A and 2B). To assess DNA fragmentation induced by the co-treatment, DNA electrophoresis was conducted. DNA electrophoresis did not show a ladder pattern of DNA fragments in the cells exposed to the single treatment of curcumin and cisplatin, whereas those exposed to the co-treatment exhibited a ladder pattern of DNA fragments (Fig. 2C).

    Augmentation of apoptosis by co-treatment with curcumin and cisplatin in SCC25 cells

    The flow cytometry results showed that the co-treatment with curcumin and cisplatin significantly increased the number of apoptotic cells with DNA hypoploidy compared to the single treatment (Fig. 3A). The co-treatment with curcumin and cisplatin resulted in the induction of the cleavage of caspase-3, caspase-7, and PARP and the production of DFF45/ICAD 11 kDa, PARP 85 kDa, and caspase-3 17 kDa cleaved products (Fig. 3B and Fig. 4A). The results of the confocal microscopy showed that DFF40/CAD was located in the cytosol following the single treatment with either curcumin or cisplatin, whereas it was clearly translocated into the nuclei following the co-treatment (Fig. 4B).

    Augmentation of apoptosis by the co-treatment with curcumin and cisplatin was demonstrated by reduction of Bcl-2 and mitochondrial membrane potential (MMP) in SCC25 cells

    The expression level of the antiapoptotic protein Bcl-2 decreased and the expression level of the apoptotic protein Bax increased in response to the co-treatment (Fig. 5A). The single treatment with either curcumin or cisplatin slightly reduced the MMP compared to the control group. In contrast, the co-treatment with curcumin and cisplatin resulted in a remarkable loss of MMP (Fig. 5B).

    Co-treatment with curcumin and cisplatin caused the translocation of AIF from mitochondria into the nucleus

    The results of confocal microscopy showed that AIF was located in the mitochondria in the cells that received the single treatment of curcumin and cisplatin, whereas it was evidently translocated into the nuclei in those that received the co-treatment (Fig. 6).

    Co-treatment with curcumin and cisplatin caused the release of cytochrome c from mitochondria into the cytosol

    The confocal microscopy results showed that cytochrome c was located in the mitochondria following the single treatment with either curcumin or cisplatin, whereas it was evidently released into the cytosol following the co-treatment (Fig. 7).

    Discussion

    Recently, the pharmacological mechanisms underlying the activities of many herbal remedies have been elucidated, and herbal medicine is becoming increasingly popular among health care professionals and the public. Curcumin is a widely used flavoring agent in food, and it has been reported to inhibit cell growth and induce apoptosis and to have antitumor activity in many cancers [19-21].

    Since the 1970s, the popularity of the neutral, square planar, coordination complex cis-diamminedichloroplatinum (II) (cisplatin) in the treatment of cancer has increased. Cisplatin, which is a well-known DNA-damaging agent and one of the most active cancer treatment agents available, is widely used for the treatment of many malignancies, including testicular, ovarian, bladder, cervical, head and neck, and small-cell and nonsmall-cell lung cancers. Unfortunately, cisplatin produces many unwanted side effects [22,23]. Studies have reported that cisplatin-induced cell death is involved in apoptotic- and cell cycle- regulating pathways [24-28]. Moreover, the synergistic antitumor effect of combination treatment with cisplatin and antitumor agents or natural products has been demonstrated [29-32].

    The mitochondria plays an important role in the induction of the mitochondrial permeability transition and also plays a key part in the regulation of apoptosis [3,33,34]. The outer mitochondrial membrane becomes permeable to intermembrane space proteins, such as cytochrome c and apoptosis-inducing factor (AIF) during apoptosis [35]. The release of cytochrome c and the disruption of the MMP are known features of apoptosis triggered by proteasome inhibition [36-38]. On induction of apoptosis, AIF translocates to the nucleus, resulting in chromatin condensation and large-scale DNA fragmentation [39,40]. This study clearly showed that co-treatment of the SCC25 cells with curcumin and cisplatin results in a remarkable decrease in the MMP, an increase in Bax, and a decrease in Bcl-2, as well as the release of cytochrome c into the cytosol and the translocation of AIF to the nuclei. In contrast, these patterns were not observed in the SCC25 cells in response to the single treatment.

    A common final event in apoptosis is nuclear condensation, which is controlled by caspases, DFF, and PARP. Caspases, cysteinyl aspartate-specific intracellular proteinases, play an essential role during apoptotic death [41]. Once activated, the effector caspases (caspase-3, caspase-6, or caspase-7) result in proteolytic cleavage of a broad spectrum of cellular targets, ultimately leading to cell death. The known cellular substrates include structural components (such as actin and nuclear lamin), inhibitors of deoxyribonuclease (such as DFF45/ICAD), and DNA repair proteins (such as PARP) [42-44]. In apoptotic cells, the activation of DFF40/CAD, also a substrate of caspase-3, occurs with the cleavage of DFF45/ICAD. Once DFF40/CAD is activated and released from the complex of DFF45/ICAD and DFF40/CAD, it can translocate to the nucleus and then degrade chromosomal DNA and produce DNA fragmentation. This study demonstrated that co-treatment of SCC25 cells with curcumin and cisplatin results in the degradation and the cleavage of caspase-3, caspase-7, PARP, and DFF45/ICAD, in addition to the translocation of DFF40/CAD to nuclei, whereas the single treatment does not.

    Taken collectively, the combination therapy with curcumin and cisplatin could be considered in the future as an alternative therapeutic strategy for human oral squamous carcinoma. Further extensive studies are needed prior to its clinical application.

    Conflict of interest

    The authors declare that they have no competing interest.

    Figure

    IJOB-39-159_F1.gif

    Co-treatment with curcumin and cisplatin significantly reduced the cell viability in SCC25 cells. Cell viability was determined by MTT assay. Three independent assays were performed. Values are means ± SD of triplicates of each experiment. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μM curcumin plus 4 μg/ml cisplatin for 24 h)

    IJOB-39-159_F2.gif

    Co-treatment with curcumin and cisplatin showed numerous condensed and fragmented nuclei in SCC25 cells compared to the single treatment. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μM curcumin plus 4 μg/ml cisplatin for 24 h) (A) Immunofluorescence micrographs showing nuclear morphology after Hoechst staining. Scale bar, 10 μm. (B) The values are represented as the mean ± SD of the number of apoptotic cells as determined by Hoechst staining. The results are from three independent experiments. (C) DNA fragmentation analysis was determined by the agarose gel electrophoresis. Whereas cells treated with either curcumin or cisplatin did not show DNA fragmentation, co-treated cells showed characteristic apoptotic figures with a ladder pattern of DNA fragments.

    IJOB-39-159_F3.gif

    The kinetic analysis of the effect of co-treatment on the SCC25 cell cycle progression and the induction of apoptosis. (A) Co-treatment showed the remarkable increase of apoptotic cells with DNA hypoploidy compared to the single treatment with either curcumin or cisplatin. (B) Western blot analysis showing that the co-treatment with curcumin and cisplatin in SCC25 cells remarkably induced caspase-3, caspase-7 and PARP degradations and produced the processed caspase-3 17 kDa, and PARP 85 kDa cleaved products. GAPDH, a loading control. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μM curcumin plus 4 μg/ml cisplatin for 24 h)

    IJOB-39-159_F4.gif

    Western blot analysis and confocal microscopy showing the efficient apoptotic effects in SCC25 cells co-treated with curcumin and cisplatin. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μM curcumin plus 4 μg/ml cisplatin for 24 h) (A) The co-treatment remarkably induced DFF45 (ICAD) degradation and produced the processed DFF45 11 kDa cleaved product. GAPDH, a loading control (B) The confocal microscopy showed that DFF40 (CAD) was translocated onto the nuclei in the co-treatment of curcumin and cisplatin. Scale bar, 10 μm.

    IJOB-39-159_F5.gif

    Co-treatment with curcumin and cisplatin showed remarkably (A) down-regulation of Bcl-2, up-regulation of the Bax and (B) the loss of MMP (Δψm) compared to the single treatmemt. MMP was measured by JC-1 with flow cytometry. The levels of GAPDH were used as an internal standard for quantifying Bcl-2 and Bax expression (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μM curcumin plus 4 μ g/ml cisplatin for 24 h)

    IJOB-39-159_F6.gif

    The confocal microscopy showed that AIF was evidently translocated onto nuclei in SCC25 cells when co-treated with curcumin and cisplatin. Scale bar, 10 μm. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μ M curcumin plus 4 μg/ml cisplatin for 24 h)

    IJOB-39-159_F7.gif

    The confocal microscopy showed that cytochrome c was evidently released to the cytosol in SCC25 cells that were co-treated with curcumin and cisplatin Scale bar, 10 μm. (Cur, cells treated with 5 μM curcumin for 24 h; Cis, cells treated with 4 μg/ml cisplatin for 24 h; Cur+Cis, cells treated with 5 μ M curcumin plus 4 μg/ml cisplatin for 24 h)

    Table

    Reference

    1. Williams GT (1991) Programmed cell death apoptosis and oncogenesis , Cell, Vol.65; pp.1097-1098
    2. Yuan J (1996) Evolutionary conservation of a genetic pathway of programmed cell death , J Cell Biochem, Vol.60; pp.4-11
    3. Susin SA , Lorenzo HK , Zamzami N , Marzo I , Snow BE , Brothers GM , Mangion J , Jacotot E , Costantini P , Loeffler M , Larochette N , Goodlett DR , Aebersold R , Siderovski DP , Penninger JM , Kroemer G (1999) Molecular characterization of mitochondrial apoptosis-inducing factor , Nature, Vol.397; pp.441-446
    4. Orlowski RZ (1999) The role of the ubiquitin-proteasome pathway in apoptosis , Cell Death Differ, Vol.6; pp.303-313
    5. Shen J , Huang C , Jiang L , Gao F , Wang Z , Zhang Y , Bai J , Zhou H , Chen Q (2007) Enhancement of cisplatin induced apoptosis by suberoylanilide hydroxamic acid in human oral squamous cell carcinoma cell lines , Biochem Pharmacol, Vol.73; pp.1901-1909
    6. Henrotin Y , Clutterbuck AL , Allaway D , Lodwig EM , Harris P , Mathy-Hartert M , Shakibaei M , Mobasheri A (2010) Biological actions of curcumin on articular chondrocytes , Osteoarthritis Cartilage, Vol.18; pp.141-149
    7. Chuang SE , Yeh PY , Lu YS , Lai GM , Liao CM , Gao M , Cheng AL (2002) Basal levels and patterns of anticancer drug-induced activation of nuclear factor-kappaB (NFkappaB), and its attenuation by tamoxifen, dexamethasone, and curcumin in carcinoma cells , Biochem Pharmacol, Vol.63; pp.1709-1716
    8. Nakamura K , Yasunaga Y , Segawa T , Ko D , Moul JW , Srivastava S , Rhim JS (2002) Curcumin down-regulates AR gene expression and activation in prostate cancer cell lines , Int J Oncol, Vol.21; pp.825-830
    9. Han SS , Keum YS , Seo HJ , Surh YJ (2002) Curcumin suppresses activation of NF-kappaB and AP-1 induced by phorbol ester in cultured human promyelocytic leukemia cells , J Biochem Mol Biol, Vol.35; pp.337-342
    10. Bachmeier BE , Mohrenz IV , Mirisola V , Schleicher E , Romeo F , Hohneke C , Jochum M , Nerlich AG , Pfeffer U (2008) Curcumin downregulates the inflammatory cytokines CXCL1 and -2 in breast cancer cells via NFkappaB , Carcinogenesis, Vol.29; pp.779-789
    11. Aravindan N , Madhusoodhanan R , Ahmad S , Johnson D , Herman TS (2008) Curcumin inhibits NFkappaB mediated radioprotection and modulate apoptosis related genes in human neuroblastoma cells , Cancer Biol Ther, Vol.7; pp.569-576
    12. Wang D , Veena MS , Stevenson K , Tang C , Ho B , Suh JD , Duarte VM , Faull KF , Mehta K , Srivatsan ES , Wang MB (2008) Liposome-encapsulated curcumin suppresses growth of head and neck squamous cell carcinoma in vitro and in xenografts through the inhibition of nuclear factor kappaB by an AKT-independent pathway , Clin Cancer Res, Vol.14; pp.6228-6236
    13. Wang G , Reed E , Li QQ (2004) Molecular basis of cellular 166 Hyeon-Jin Sohn, In-Ryoung Kim, Yong-Ho Kim, Gyoo-Cheon Kim, Hyun-Ho Kwak, and Bong-Soo Park response to cisplatin chemotherapy in non-small cell lung cancer (Review) , Oncol Rep, Vol.12; pp.955-965
    14. Adhami VM , Malik A , Zaman N , Sarfaraz S , Siddiqui IA , Syed DN , Afaq F , Pasha FS , Saleem M , Mukhtar H (2007) Combined inhibitory effects of green tea polyphenols and selective cyclooxygenase-2 inhibitors on the growth of human prostate cancer cells both in vitro and in vivo , Clin Cancer Res, Vol.13; pp.1611-1619
    15. Mai Z , Blackburn GL , Zhou JR (2007) Genistein sensitizes inhibitory effect of tamoxifen on the growth of estrogen receptor-positive and HER2-overexpressing human breast cancer cells , Mol Carcinog, Vol.46; pp.534-542
    16. Song MQ , Zhu JS , Chen JL , Wang L , Da W , Zhu L , Zhang WP (2007) Synergistic effect of oxymatrine and angiogenesis inhibitor NM-3 on modulating apoptosis in human gastric cancer cells , World J Gastroenterol, Vol.13; pp.1788-1793
    17. Vinall RL , Hwa K , Ghosh P , Pan CX , Lara PN Jr, de Vere White RW (2007) Combination treatment of prostate cancer cell lines with bioactive soy isoflavones and perifosine causes increased growth arrest and/or apoptosis , Clin Cancer Res, Vol.13; pp.6204-6216
    18. Lee SH , Ryu JK , Lee KY , Woo SM , Park JK , Yoo JW , Kim YT , Yoon YB (2008) Enhanced anti-tumor effect of combination therapy with gemcitabine and apigenin in pancreatic cancer , Cancer Lett, Vol.259; pp.39-49
    19. Ravindran J , Prasad S , Aggarwal BB (2009) Curcumin and cancer cells how many ways can curry kill tumor cells selectively? , AAPS. J, Vol.11; pp.495-510
    20. Wang Z , Zhang Y , Banerjee S , Li Y , Sarkar FH (2006) Notch-1 down-regulation by curcumin is associated with the inhibition of cell growth and the induction of apoptosis in pancreatic cancer cells , Cancer, Vol.106; pp.2503-2513
    21. Sarkar FH , Li Y , Wang Z , Padhye S (2010) Lesson learned from nature for the development of novel anti-cancer agents implication of isoflavone, curcumin, and their synthetic analogs , Curr Pharm Des, Vol.16; pp.1801-1812
    22. Cooley ME , Davis LE , DeStefano M , Abrahm J (1994) Cisplatin a clinical review: Part I--Current uses of cisplatin and administration guidelines , Cancer Nurs, Vol.17; pp.173-184
    23. Gonzalez VM , Fuertes MA , Alonso C , Perez JM (2001) Is cisplatin-induced cell death always produced by apoptosis? , Mol Pharmacol, Vol.59; pp.657-663
    24. Zhou H , Kato A , Yasuda H , Miyaji T , Fujigaki Y , Yamamoto T , Yonemura K , Hishida A (2004) The induction of cell cycle regulatory and DNA repair proteins in cisplatin-induced acute renal failure , Toxicol Appl Pharmacol, Vol.200; pp.111-120
    25. Fox SA , Kusmiaty Loh SS , Dharmarajan AM , Garlepp MJ (2005) Cisplatin and TNF-alpha downregulate transcription of Bcl-xL in murine malignant mesothelioma cells , Biochem Biophys Res Commun, Vol.337; pp.983-991
    26. Garcia-Berrocal JR , Nevado J , Ramirez-Camacho R , Sanz R , Gonzalez-Garcia JA , Sanchez-Rodriguez C , Cantos B , Espana P , Verdaguer JM , Trinidad Cabezas A (2007) The anticancer drug cisplatin induces an intrinsic apoptotic pathway inside the inner ear , Br J Pharmacol, Vol.152; pp.1012-1020
    27. Gagnon V , Van Themsche C , Turner S , Leblanc V , Asselin E (2008) Akt and XIAP regulate the sensitivity of human uterine cancer cells to cisplatin, doxorubicin and taxol , Apoptosis, Vol.13; pp.259-271
    28. Kondo K , Yamasaki S , Inoue N , Sugie T , Teratani N , Kan T , Shimada Y (2006) Prospective antitumor effects of the combination of tumor necrosis factor-related apoptosis -inducing ligand (TRAIL) and cisplatin against esophageal squamous cell carcinoma , Surg Today, Vol.36; pp.966-974
    29. Mohammad RM , Banerjee S , Li Y , Aboukameel A , Kucuk O , Sarkar FH (2006) Cisplatin-induced antitumor activity is potentiated by the soy isoflavone genistein in BxPC-3 pancreatic tumor xenografts , Cancer, Vol.106; pp.1260-1268
    30. Iwase M , Yoshiba S , Uchid M , Takaoka S , Kurihara Y , Ito D , Hatori M , Shintani S (2007) Enhanced susceptibility to apoptosis of oral squamous cell carcinoma cells subjected to combined treatment with anticancer drugs and phosphatidylinositol 3-kinase inhibitors , Int J Oncol, Vol.31; pp.1141-1147
    31. Pisano C , Vesci L , Fodera R , Ferrara FF , Rossi C , De Cesare M , Zuco V , Pratesi G , Supino R , Zunino F (2007) Antitumor activity of the combination of synthetic retinoid ST1926, and cisplatin in ovarian carcinoma models , Ann Oncol, Vol.18; pp.1500-1505
    32. Kroemer G , Zamzami N , Susin SA (1997) Mitochondrial control of apoptosis , Immunol Today, Vol.18; pp.44-51
    33. Green DR , Reed JC (1998) Mitochondria and apoptosis , Science, Vol.281; pp.1309-1312
    34. Golab J , Stoklosa T , Czajka A , Dabrowska A , Jakobisiak M , Zagozdzon R , Wojcik C , Marczak M , Wilk S (2000) Synergistic antitumor effects of a selective proteasome inhibitor and TNF in mice , Anticancer Res, Vol.20; pp.1717-1721
    35. Huffman HA , Sadeghi M , Seemuller E , Baumeister W , Dunn MF (2003) Proteasome-cytochrome c interactions a model system for investigation of proteasome host-guest interactions , Biochemistry, Vol.42; pp.8679-8686
    36. MacFarlane M , Merrison W , Bratton SB , Cohen GM (2002) Proteasome-mediated degradation of Smac during apoptosis XIAP promotes Smac ubiquitination in vitro , J Biol Chem, Vol.277; pp.36611-36616
    37. Marshansky V , Wang X , Bertrand R , Luo H , Duguid W , Chinnadurai G , Kanaan N , Vu MD , Wu J (2001) Proteasomes modulate balance among proapoptotic and antiapoptotic Bcl-2 family members and compromise functioning of the electron transport chain in leukemic cells , J Immunol, Vol.166; pp.3130-142
    38. Daugas E , Susin SA , Zamzami N , Ferri KF , Irinopoulou T , Larochette N , Prevost MC , Leber B , Andrews D , Penninger J , Kroemer G (2000) Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis , FASEB. J, Vol.14; pp.729-739
    39. Sevrioukova IF (2011) Apoptosis-inducing factor structure, function, and redox regulation , Antioxid Redox Signal, Vol.14; pp.2545-2579
    40. Gross A , McDonnell JM , Korsmeyer SJ (1999) BCL-2 family members and the mitochondria in apoptosis , Genes Dev, Vol.13; pp.1899-1911,
    41. Widlak P , Garrard WT (2006) Unique features of the apoptotic endonuclease DFF40/CAD relative to micrococcal nuclease as a structural probe for chromatin , Biochem Cell Biol, Vol.84; pp.405-410
    42. Magalska A , Brzezinska A , Bielak-Zmijewska A , Piwocka K , Mosieniak G , Sikora E (2006) Curcumin induces cell death without oligonucleosomal DNA fragmentation in quiescent and proliferating human CD8+ cells , Acta Biochim Pol, Vol.53; pp.531-538
    43. Widlak P , Garrard WT (2005) Discovery, regulation, and action of the major apoptotic nucleases DFF40/CAD and endonuclease, G , J Cell Biochem, Vol.94; pp.1078-1087
    44. Cheng AC , Jian CB , Huang YT , Lai CS , Hsu PC , Pan MH (2007) Induction of apoptosis by Uncaria tomentosa through reactive oxygen species production, cytochrome c release, and caspases activation in human leukemia cells , Food Chem Toxicol, Vol.45; pp.2206-2218