Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1225-1577(Print)
ISSN : 2384-0900(Online)
The Korean Journal of Oral and Maxillofacial Pathology Vol.40 No.6 pp.899-909
DOI : https://doi.org/10.17779/KAOMP.2016.40.6.004

The Extract of Kochia scoparia Fruit Induces Programmed Necrosis in Oral Squamous Cell Carcinoma Cells

Hye-Yeon Han1)*, Bong-Soo Bark2)*, Hyung Joon Kim3), Seung-Hwa Jeong4), Jiyeon Kim5), Sung-Hee Jeong6), Gyoo Cheon Kim2), Dae-Seok Hwang7), Uk-Kyu Kim7), Hyungwoo Kim8), Mi Heon Ryu9)
1)Department of Oral Pathology, School of Dentistry, Research Institute for Oral Biotechnology
2)Department of Dental Anatomy, BK21 Plus project, School of Dentistry
3)Department of Oral Physiology, BK21 Plus project, School of Dentistry
4)Department of Preventive and Community Dentistry, BK21 Plus project, School of Dentistry
5)Department of Pediatric Dentistry, School of Dentistry, Dental Research Institute
6)Department of Oral Medicine, School of Dentistry, Dental Research Institute
7)Department of Oral and Maxillofacial Surgery, School of Dentistry
8)Division of Pharmacology, School of Korean Medicine
9)Department of Oral Pathology, BK21 Plus project, School of Dentistry, Pusan National University, Yangsan, South Korea

These authors contributing equally to this article

Corresponding: Mi Heon Ryu, Department of Oral Pathology, BK21 Plus project, School of Dentistry, Pusan National University, 49 Busandaehak-ro, Yangsan, Gyeongnam, 626-870, South Korea. +82-51-510-8251apollon@pusan.ac.kr
November 12, 2016 November 18, 2016 November 25, 2016

Abstract

The fruit of Kochia scoparia Scharder is traditionally used as a medicinal ingredient to treat allergic skin diseases and inflammatory diseases in China, Japan and Korea. Recently, several studies reported that K. scoparia had potential for the cytotoxicity of human cancer cells. To investigate the anti-cancer effect of K. scoparia on oral cancer and to determine the specific type of cell death induced by MEKS treatment. We investigated the anti-cancer effects of K. scoparia, methanol extract (MEKS) in HSC4 human oral cancer cells. We examined the effects of MEKS on the proliferation rate, cell cycle arrest, 7-AAD-ANNEXIN V double stain, reactive oxygen species (ROS) generation and activation of apoptosis and necroptosis-associated proteins in HSC4 cells. MTT assay results demonstrated that MEKS decreased the proliferation rates of HSC4 cells in a dose-dependent manner with an IC50 value of 45.3 μg/ml. MEKS at 50 μg/ml significantly increased the sub-G1 DNA contents of HSC4 cells to 84.8%, versus untreated cells. However, the activation of apoptosis-associated proteins such as cleaved caspase 3, cleaved caspase 8, cleaved caspase 9 and cleaved Poly (ADP-ribose) polymerase (PARP) did not detect. The level of Bax protein markedly increased in MEKS-treated HSC4 cells. In addition, the cell viability of the DPQ pre-treated HSC4 cells with MEKS treatment was significantly greater than that of MEKS treated-cells. These results suggest that MEKS inhibits cell proliferation and induces necroptosis in oral cancer cells and that MEKS may have potential chemotherapeutic value for the treatment of human oral cancer.


Kochia scoparia 열매 추출물이 구강 편평세포암종세포에서 예정된 괴사 유도

한 혜련1)*, 박 봉수2)*, 김 준형3), 정 승화4), 김 지연5), 정 성희6), 김 규천2), 황 대석7), 김 욱규7), 김 형우8), 류 미현9)
1)경구 생명 공학 연구소 치과 대학 구강 병리과
2)치과 해부학과, BK21 Plus 프로젝트, 치과 대학
3)구강 생리학 부, BK21 Plus 프로젝트, 치과 대학
4)학과 예방 및 지역 사회 치과, BK21 Plus 프로젝트, 치과 대학
5)학과 소아과 치과 치의학 연구소 치과 연구소
6)치과 연구소 구강 내과학 교실
7)학과 구강 악안면 외과학 교실, 치과 대학원
8)대한 약리학 교실
9)학과 부산 대학교 의과 대학 치과 대학 구강 병리학 교실, BK21 Plus 프로젝트

초록


    Pusan National University

    Ⅰ.INTRODUCTION

    Cell death is traditionally classified into 2 types; apoptosis and necrosis forms1,2). Apoptosis is considered programmed cell death, which is controlled by elaborated coordinated pathway3). Meanwhile, necrosis is well known as non-programmed cell death, which occurs in response to external shocks to the cells or tissue, such as trauma, infection, or toxin, which can result in the unregulated digestion of cell components. It is characterized by morphological features including the increase of cell volume, microorganelle swelling, the rupture of plasma membrane and subsequent cell death4).

    Recently, a new type of cell death, necroptosis, has been reported. Numerous reports identified necroptosis as a programmed cell death mechanism, not cell death caused by accidental chance. Necroptosis can be initiated when apoptosis is shut out. In addition, apoptosis is associated with caspase-dependent pathway while necroptosis is associated with kinase, especially receptor interacting protein (RIP), a sort of kinase activity containing protein, which is essential in the detection of cellular stress 5,6). Recently, multiple lines of evidence have demonstrated that necroptosis is associated with myocardial infarction7), cerebral infarction 8,9), atherosclerosis10), acute pancreatitis 11), and more importantly, cancer 12,13).

    The mature fruit of Kochia scoparia (L.) Schrad. (family Chenopodiaceae) is widely spread throughout the western and northern United States, Europe, Africa, South America, and the Far East14). The plant has been used in Chinese and Korean traditional medicine as a treatment for allergic skin diseases15), diabetes mellitus16), rheumatoid arthiritis17) and liver diseases18). In addition, the fruit is used as a food garnish in Japan19). Several recent studies indicated that the extract of K. scoparia fruit has anti-tumor activity against human hepatocellular carcinoma cells and immortal neuroblastoma cell lines20-22). Previously, the authors reported that the methanol extract of K. scoparia dried fruit (MEKS) has possible anti-cancer effects in human breast cancer cell lines and oral squamous cell carcinoma cell (OSCC) lines. The main mechanism of MEKS was reported to be apoptosis, which was controlled by the activation of caspases. However, no report has been issued on the other anti-cancer mechanism of K. scoparia in OSCC cell lines.

    Therefore, we evaluated the anti-cancer activity of MEKS by measuring proliferation rates and non-apoptotic cell death in HSC4 human oral cancer cells. In addition, we sought to identify the molecular mechanism responsible.

    Ⅱ.MATERIALS AND METHODS

    Reagents and antibodies

    Paclitaxel, MTT (3, 4, 5-dimethyl N-methylthiazol-2-yl-2, 5-d-phenyl tetrazolium bromide), 3, 4-Dihydro-5-[4-(1- piperidinyl)butoxy]-1(2H)-isoquinoline (DPQ) and propidium iodide (PI) solution were purchased from Sigma-Aldrich (St. Louis, MO, USA). 2, 7-Dichloro-fluorescein diacetate (DCFHDA) was obtained from Eastman Kodak (Rochester, NY, USA). The ANNEXIN V-FITC apoptosis detection kit, anti-rabbit IgG antibody and anti-mouse IgG antibody were purchased from Enzo Life Sciences (Farmingdale, NY, USA). The antibodies targeting Bax, cleaved caspase 3, cleaved caspase 8, cleaved caspase 9 and cleaved Poly (ADP-ribose) polymerase (PARP) were purchased from Cell Signaling Technology (Beverly, MA, USA), while anti-beta actin antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA).

    Preparation of MEKS

    The dried fruit of K. scoparia was purchased from Hwalim Medicinal Herbs (Pusan, Korea). Extraction was performed using a standard extraction process, as previously described. Briefly, 100 g of the dried fruit of K. scoparia were immersed in 1 L of methanol, sonicated for 30 min and then allowed to stand for 48 h. The obtained extract was filtered through No. 20 Whatman filter paper, evaporated under reduced pressure using a vacuum evaporator (Eyela, Japan) and lyophilized using a freeze dryer (Labconco, Kansas City, MO, USA). Finally, 4.46 g of lyophilized powder was obtained (yield, 4.46%). A sample of the lyophilized powder (MEKS, Voucher No. MH2013-006) was deposited at the Division of Pharmacology, School of Korean Medicine, Pusan National University.

    Cell culture

    HSC4 cell (a human oral cancer cell line) was purchased from the JCRB Cell Bank (Japan) and cultured in DMEM (Hyclone Laboratories, Logan, UT, USA) supplemented with 10% FBS (Hyclone Laboratories) and 1% penicillin/ streptomycin (Invitrogen, Carlsbad, CA, USA).

    MTT assay

    Proliferation rates of HSC4 cells were measured using a MTT proliferation assay. Briefly, cells were seeded in 24-well plates (5x104 per well) and cultured overnight to allow attachment. They were then treated with MEKS at the concentration of 0, 12.5, 25, 50, 75 and 100 μg/ml for 4 h, respectively, and then in fresh MEKS-free media for 20 h. Control cells were treated with vehicle (dimethyl sulfoxide; DMSO) for 4 h. MTT activities were measured in triplicate for control and experimental groups. MTT solution was added to each well, and then the cells were incubated at 37 ℃ for 4 h in a 5% CO2 atmosphere. Media were then removed and the formazan crystals produced were dissolved in 100 μl DMSO. Absorbances were read at 570 nm using a microplate reader (Bio-Rad Laboratories, Hercules, CA, USA).

    Cell morphology

    A phase contrast microscope (Olympus, Tokyo, Japan) was used to determine the cell morphologies of untreated and MEKS-treated HSC4 cells.

    ANNEXIN V and 7-AAD double staining

    Cells were seeded in 6-well plates (3x105 per well), incubated overnight and treated with MEKS in indicated concentrations for 4 h. MEKS was then removed and cells were cultured in fresh media for a further 20 h. Control cells were treated with vehicle (DMSO) for 4 h. Thirty nM of paclitaxel was used as positive control. After incubation, cells were trypsinized, harvested, washed with phosphatebuffered saline (PBS), resuspended in 500 μl of Binding Buffer (ANNEXIN V-FITC apoptosis detection kit, Enzo Life Sciences) and stained using the ANNEXIN V-FITC apoptosis detection kit (Enzo Life Sciences) at room temperature for 5 min in the dark, according to the manufacturer’s instructions. Stained cells were analyzed using a Flow cytometer (BD Biosciences, Heidelberg, Germany) and the data obtained was analyzed using the FACS (Fluorescence Activated Cell Sorting)-Canto II software.

    Cell cycle analysis

    After treatment with MEKS for 4 h, HSC4 cells were trypsinized, harvested and washed with PBS twice. After homogenisation, cells were fixed with 75% ethanol and washed with PBS. They were then re-suspended in PBS containing RNase (40 μg/ml) for 30 min and treated with PI solution (10 μg/ml). Cells were then transferred to FACS tubes and DNA contents were analysed using a FACS Scan flow cytometer. Data were analysed using FACS-Canto II software.

    Intracellular reactive oxygen species detection

    The levels of intracellular (ROS) were measured using DCFH-DA. Briefly, HSC4 cells (2.5x104) were cultured overnight in a 96-well plate. Then, 50 μM of DCFH-DA was added and the cells were cultured for a further 60 min. Cells were then treated at the concentration of 0, 15, 30, 45, or 60 μg/ml of MEKS and fluorescence intensities were measured using a TECAN Infinite M200 fluorometric plate reader (excitation at 485 nm, emission at 530 nm, Munnedorf, Switzerland).

    Western blot analysis

    HSC4 cells were treated with MEKS at the concentration of 0, 25, 50 or 75 μg/ml for 4 h, then cultured in fresh media for further 20 h. After treatment, cells were collected and lysed with RIPA buffer (Cell Signaling Technology) for 30 min. Then, the supernatant was taken by 20 min centrifugation at 12,000 rpm, diluted in sodium dodecyl sulfate (SDS) buffer, and boiled for 5 min. After quantification, 50 μg of protein extract were subjected to electrophoresis using 10% SDS-polyacrylamide gel, then transferred to polyvinylidene fluoride membrane for 2 h. Blocking was performed using TNE buffer [50 mm Tris/HCl (pH 7.4), 100 mm NaCl, 0.1 mm EDTA] containing 5% skim milk and 0.1% tween-20. Antibodies against Bax, cleaved PARP, cleaved caspase 3, cleaved caspase 8 and cleaved caspase 9 were incubated overnight for 4 ℃. Then, it was applied with horseradish-conjugated secondary antibody and detected using SuperSignal West-Femto reagent (Pierce, Rockford, IL, USA).

    Statistical analysis

    The Student’s-t test in Window PASW (Predictive Analytics SoftWare) version 21.0 (SPSS Inc, Chicago, IL, USA) was used to analyse the significance of differences between the control and MEKS-treated groups. Results are presented as means ± standard errors and statistical significance was accepted for P<0.05, as indicated.

    Ⅲ.RESULTS

    MEKS treatment inhibited cell proliferation and modified the morphology of oral cancer cells.

    MEKS inhibited cell proliferation in a dose-dependent manner (Figure 1a), with an IC50 value (50% growth inhibition) of 45.3 μg/ml. After treatment with MEKS, cells became smaller and rounder and lost their cellular processes (Figure 1b). MEKS-treated cells displayed the ruptured cell membrane without chromatin clumping or apoptotic body formation, distinguished from apoptotic characteristics. Taken together, these results demonstrated that MEKS inhibited cell proliferation and induced cell death.

    MEKS induced early and late apoptosis in HSC4 cells

    7-AAD and ANNEXIN V FITC double staining demonstrated that MEKS increased the number of apoptotic cells in a dose-dependent manner. In particular, treatment with of MEKS at 75 μg/ml caused the apoptosis of 78.5% of cells (Figure 2).

    MEKS induced sub-G1 arrest and increased the number of apoptotic cells

    Only 1.0% of control cells were in the sub-G1 phase. However, cells treated with 50 μg/ml MEKS showed a marked increase in the number of cells undergoing sub-G1 arrest. The proportions of sub-G1 arrested cells in the 50 and 75 μg/ml MEKS were 84.8% and 93.7%, respectively (Figure 3).

    MEKS elevated intracellular ROS levels

    Treatment with MEKS markedly increased intracellular ROS levels, as shown in Figure 4. ROS was found to accumulate in a time-dependent manner, with peak ROS production occurring following exposure to 25 and 100 μg/ml of MEKS. After 60 min of incubation with 25 μg/ml of MEKS, intracellular ROS levels were approximately 2.0 fold higher than in the untreated control group (Figure 4).

    MEKS did not induce the intrinsic and extrinsic apoptotic pathway

    Interestingly, treatment of HSC4 cells with MEKS did not induce intrinsic pathway-related proteins such as caspase 8 and extrinsic pathway-related proteins such as caspase 9, caspase 3 and cleaved PARP. In contrast, MEKS significantly increased the levels of Bax protein in a dose-dependent manner (Figure 5).

    DPQ pretreatment alleviated cell death induced by MEKS treatment in HSC4 cells

    To determine the specific type of cell death induced by MEKS treatment, we performed an MTT assay on cells pre-treated with DPQ (PARP1 inhibitor) for 1 h prior to MEKS treatment. It was found that the cell viability of the DPQ pre-treated HSC4 cells with MEKS treatment was significantly greater than that of MEKS treated-cells (Figure 6).

    Ⅳ.DISCUSSION

    Our results demonstrated that MEKS treatment caused the proliferation inhibition of HSC4 cells and the increase of late apoptotic cell numbers corresponding to increasing concentrations of MEKS. Moreover, the percentage of sub-G1 arrested cells and the production of ROS also increased. However, the cleavage of caspases including caspase 3, 8 and 9 were not detected in the MEKS-treated HSC4 cells. More importantly, the activation of Bax was detected and the application of DPQ effectively diminished the cell death in response to MEKS treatment. Taken together, these findings indicated that the cell death induced by MEKS occurs via apoptotic mechanisms.

    Apoptosis is characterized by typical morphological features, including chromatin condensation, nuclear fragmentation, apoptotic body formation, cellular shrinkage, and plasma membrane blebbing5). In contrast, MEKS-treated HSC4 cells showed morphological changes in a somewhat different form from characteristic apoptotic morphology. The MEKS-treated cells displayed the ruptured cell membrane without chromatin clumping or apoptotic body formation. Furthermore, the cleavage of caspases involved in the apoptotic pathway was not detected. DPQ pretreatment with MEKS abolished MEKS-induced cell death. Programmed necrosis is becoming increasingly realized as cell death that can take place through regulated mechanisms or pathways, termed necroptosis23). This type of cell death occurrs via the activation of tumor necrosis factor (TNF) alpha, Fas L, and TRAIL, which are the same proteins that can trigger an apoptotic pathway23). However, in the case of inactivation of caspases, the alternative cell death pathway, necroptosis, can be stimulated.

    RIP kinases are crucial regulators of cell survival and cell death, and required as key regulators of necroptosis. Our results demonstrated DPQ pretreatment prior to MEKS decreased cell death effectively. In addition, the cleavage of caspases which are essential in the apoptotic pathway did not occurr. Given the different cell morphology, the cell death induced by MEKS treatment in HSC4 cells seems to be necroptosis.

    ROS are produced as a natural byproduct of the normal metabolism of cells and play an important role in cell signaling and homeostasis24). It involves various cellular processes, physiological conditions as well as pathological conditions. Under pathological conditions like ionizing radiation, heat exposure, and UV, uncontrolled excessive ROS production begins and provokes various cellular responses. Mitochondrial ROS can intercede cell death mechanisms and it shows characteristic ultrastructural changes3,25). The necroptosis pathway is closely connected with ROS production and mitochondrial as well as non-mitochondrial3). Irrinki et al. reported that the over-expression of antioxidants diminished ROS production and protected cells from necroptosis26). Therefore, similar to apoptosis, ROS plays an essential role in the initiation of the necroptic pathway.

    Bax is a crucial regulator during apoptotic cell death, especially mitochondria-dependent apoptosis. It mediates not only the apoptosis by an intrinsic pathway, but also a necroptosis pathway through mitochondrial membrane permeabilization27). The activated Bax protein can induce the translocation in mitochondria, and trigger a cytochrome C release28,29). In the necroptotic pathway, MEK/ERK and JNK can be triggered by DNA damage and result in necrotic cell death30,31). However, the regulatory mechanism by the activation of Bax protein can be related to specific cell types and depends on cytotoxic agents.

    Between PARP1 activation and ROS production, there are bidirectional interactions32). ROS production can trigger PARP1 activation, and PARP1 activation may induce ROS production form mitochondria33). Chen et al. reported that ROS production and PARP activation eventually led to necroptosis in murine L929 fibrosarcoma cells34). Dysfunction of mitochondria plays an essential role in PARP-induced necroptosis, proven by the PARP1 inhibitor (DPQ) pretreatment25).

    Recently, several approaches demonstrated that necroptosis can be a novel strategy for cancer chemotherapy35-38). The research indicated that induction of necroptosis is a valuable cancer strategy in several cancer cells. More importantly, necroptosis seems to be a promising strategy under conditions where apoptosis fail or not be effective2,35). These days, it is promising to discover that new chemotherapeutic agents can implicate multiple cell death pathways. Understanding the mechanism of necroptosis and its application in new therapeutic agents for cancer can be an alternative strategy.

    In summary, the results of our study revealed the induction of necroptosis in HSC4 cells by MEKS treatment. Additionally, MEKS can be a promising adjuvant chemotherapeutic agent that induces multiple cell death pathways.

    Ⅴ.ACKNOWLEDGEMENT

    Ⅴ.

    This work was supported by a 2-Year Research Grant of Pusan National University.

    Figure

    KAOMP-40-899_F1.gif

    Effects of MEKS on HSC4 human oral cancer cells. After treatment with the vehicle or 12.5, 25, 50, 75, or 100 μg/ml of MEKS for 4 h, then we changed media and maintained for an additional 20 h. Cell viability was determined by a MTT assay. A: Viability of HSC4 cells after 4 h of MEKS treatment. B: Morphological change in MEKS-treated HSC4 cells after 4 h. (A) 0 μg/ml of MEKS; (B) 12.5 μg/ml of MEKS; (C) 25 μg/ml of MEKS; (D) 50 μg/ml of MEKS; (E) 75 μg/ml of MEKS; and (F) 100 μg/ml of MEKS. Cell morphology was observed using phase-contrast microscopy (Magnification, X200).

    KAOMP-40-899_F2.gif

    Effect of MEKS on the induction of apoptosis after 24 h of treatment with the vehicle or 12.5, 25, 50, 75, or 100 μg/ml of MEKS. After the MEKS treatment for 4 h, we changed media and maintained for an additional 20 h. After MEKS treatment, the cells were stained with Annexin-V and 7-AAD and analyzed by flow cytometry. Early apoptotic cells were stained with Annexin-V but not 7-AAD (lower right quadrant, Q4). Late apoptotic cells were stained by both Annexin-V and 7-AAD (The upper right quadrant, Q2).

    KAOMP-40-899_F3.gif

    The percentage of HSC4 cells undergoing sub-G1 arrest after 24 h of treatment with the vehicle or 12.5, 25, 50, 75, or 100 μg/ml of MEKS for 4 h, then changed media and maintained for additional 20 h, respectively. After MEKS treatment, propidium iodide staining was performed and the cells were analyzed by flow cytometry.

    KAOMP-40-899_F4.gif

    Effects of MEKS on the production of ROS in HSC4 cells. The cells were treated with vehicle or 12.5, 25, 50, or 100 μg/ml of MEKS for 60 min. Cells were then loaded with the ROS-specific probe, DCFH-DA and fluorescence were measured using a fluorometric plate reader. Values are means ± standard errors.

    KAOMP-40-899_F5.gif

    Effects of MEKS on the activations of apoptosis and necroptosis-related proteins. HSC4 cells were treated with 0, 25, 50 or 75 μg/ml of MEKS for 4 h or 30 nM paclitaxel for 24 h (as a positive control). Protein expressions of cleaved caspase 8 (A), cleaved PARP and cleaved caspase 9 (B) cleaved caspase 3 (C), Bax (D) and β-actin (the loading control) were detected by Western blotting.

    KAOMP-40-899_F6.gif

    Effects of MEKS on the viabilities of HSC4 cells pre-treated with DPQ. HSC4 cells were pretreated with or without DPQ (10μM) for 1 h and then with vehicle or 12.5, 25, 50, 75, or 100 μg/ml of MEKS for 4 h, then changed DPQ-contained media and maintained for additional 20 h. Cell viability was analyzed using an MTT assay.

    Table

    Reference

    1. He S , Wang L , Zhao L , Wang X (2009) Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha , Cell, Vol.137 ; pp.1100-1111
    2. Conrad M , Angeli JP , Vandenabeele P , Stockwell BR (2016) Regulated necrosis disease relevance and therapeutic opportunities , Nat Rev Drug Discov, Vol.15 ; pp.348- 366
    3. Vandenabeele P , Galluzzi L , Vanden Berghe T , Kroemer G (2010) Molecular mechanisms of necroptosis an ordered cellular explosion , Nat Rev Mol Cell Biol, Vol.11 ; pp.700-714
    4. Kroemer G , Galluzzi L , Zhivotovsky B , Melino G (2009) Nomenclature Committee on Cell D Classification of cell death recommendations of the Nomenclature Committee on Cell Death 2009, , Cell Death Differ, Vol.16 ; pp.3-11
    5. Galluzzi L , Kroemer G (2008) Necroptosis a specialized pathway of programmed necrosis , Cell, Vol.135 ; pp.1161-1163
    6. Hitomi J , Christofferson DE , Xavier RJ , Yuan J (2008) Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway , Cell, Vol.135 ; pp.1311-1323
    7. Smith CC , Davidson SM , Hothersall JS , Yellon DM (2007) Necrostatin a potentially novel cardioprotective agent? , Cardiovasc Drugs Ther, Vol.21 ; pp.227-233
    8. Newton K , Dugger DL , Webster JD , Vucic D (2016) RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury , Cell Death Diffe, Vol.23 ; pp.1565-1576
    9. Xuan M , Okazaki M , Iizuka H , Hibino Y (2015) Chronic Treatment with a Water-Soluble Extract from the Culture Medium of Ganoderma lucidum Mycelia Prevents Apoptosis and Necroptosis in Hypoxia/Ischemia-Induced Injury of Type 2 Diabetic Mouse Brain , Evid Based Complement Alternat Med, Vol.2015 ; pp.865986
    10. Grootaert MO , Schrijvers DM , De Meyer GR , Martinet W (2016) Caspase-3 Deletion Promotes Necrosis in Atherosclerotic Plaques of ApoE Knockout Mice , Oxid Med Cell Longev, Vol.2016 ; pp.3087469
    11. Wu J , Huang Z , Chen L , Han J (2013) Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis , Cell Res, Vol.23 ; pp.994-1006
    12. Sosna J , Philipp S , Schutze S , Adam D (2016) Differences and Similarities in TRAIL- and Tumor Necrosis Factor-Mediated Necroptotic Signaling in Cancer Cells , Mol Cell Biol, Vol.36 ; pp.2626-2644
    13. Jia LT , Zhang R , Shen L , Yang AG (2015) Regulators of carcinogenesis emerging roles beyond their primary functions , Cancer Lett, Vol.357 ; pp.75-82
    14. Friesen LF , Beckie HJ , Warwick SI , Van Acker RC (2009) The biology of Canadian weeds. 138. Kochia scoparia (L.) Schrad , Can J Plant Sci, Vol.89 ; pp.141-167
    15. Choi YY , Kim MH , Kim SH , Yang WM (2014) Topical application of Kochia scoparia inhibits the development of contact dermatitis in mice , J Ethnopharmacol, Vol.154 ; pp.380-385
    16. Matsuda H , Li Y , Yamahara J , Yoshikawa M (1999) Inhibition of gastric emptying by triterpene saponin, momordin Ic, in mice roles of blood glucose, capsaicin-sensitive sensory nerves, and central nervous system , J Pharmacol Exp Ther, Vol.289 ; pp.729-734
    17. Choi J , Lee KT , Park HS , Park HJ (2002) Anti-rheumatoid arthritis effect of the Kochia scoparia fruits and activity comparison of momordin lc, its prosapogenin and sapogenin , Arch Pharm Res, Vol.25 ; pp.336-342
    18. Kim NY , Lee MK , Cho SY , Lee JS (2005) Momordin Ic and oleanolic acid from Kochiae Fructus reduce carbon tetrachlorideinduced hepatotoxicity in rats , J Med Food, Vol.8 ; pp.177-183
    19. Yoshikawa M , Shimada H , Hori K , Yamahara J (1997) Medicinal foodstuffs. VII. On the saponin constituents with glucose and alcohol absorption-inhibitory activity from a food garnish "Tonburi", the fruit of Japanese Kochia scoparia (L.) Schrad.: structures of scoparianosides A, B, and C , Chem Pharm Bull(Tokyo), Vol.45 ; pp.1300-1305
    20. Mazzio EA , Soliman KFA (2009) In Vitro Screening for the Tumoricidal Properties of International Medicinal Herbs , Phytother Res, Vol.23 ; pp.385-398
    21. Wang J , Yuan L , Wang YT , Liu XB (2014) A novel mechanism for momordin Ic-induced HepG2 apoptosis involvement of PI3K-and MAPK-dependent PPAR gamma activation , Food Funct, Vol.5 ; pp.859-868
    22. Wang J , Yuan L , Wang YT , Liu XB (2013) Momordin Ic induces HepG2 cell apoptosis through MAPK and PI3K/Akt-mediated mitochondrial pathways , Apoptosis, Vol.18 ; pp.751-765
    23. Teng X , Degterev A , Yuan J , Cuny GD (2005) Structure-activity relationship study of novel necroptosis inhibitors , Bioorg Med Chem Lett, Vol.15 ; pp.5039-5044
    24. Devasagayam TP , Tilak JC , Ghaskadbi SS , Lele RD (2004) Free radicals and antioxidants in human health current status and future prospects , J Assoc Physicians India, Vol.52 ; pp.794-804
    25. Wu W , Liu P , Li J (2012) Necroptosis an emerging form of programmed cell death , Crit Rev Oncol Hematol, Vol.82 ; pp.249-258
    26. Irrinki KM , Mallilankaraman K , Balachandran S , Madesh M (2011) Requirement of FADD, NEMO, and BAX/BAK for aberrant mitochondrial function in tumor necrosis factor alpha-induced necrosis , Mol Cell Biol, Vol.31 ; pp.3745-3758
    27. Antignani A , Youle RJ (2006) How do Bax and Bak lead to permeabilization of the outer mitochondrial membrane? , Curr Opin Cell Biol, Vol.18 ; pp.685-689
    28. Holcakova J , Ceskova P , Palecek E , Vojtesek B (2008) The cell type-specific effect of TAp73 isoforms on the cell cycle and apoptosis , Cell Mol Biol Lett, Vol.13 ; pp.404-420
    29. Satoh S , Arai K , Watanabe S (2004) Identification of a novel splicing form of zebrafish p73 having a strong transcriptional activity , Biochem Biophys Res Commun, Vol.325 ; pp.835-842
    30. Bai L , Yoon SO , King PD , Merchant JL (2004) ZBP-89-induced apoptosis is p53-independent and requires JNK , Cell Death Differ, Vol.11 ; pp.663-673
    31. Prasad S , Yadav VR , Kannappan R , Aggarwal BB (2011) Ursolic acid, a pentacyclin triterpene, potentiates TRAIL-induced apoptosis through p53-independent up-regulation of death receptors evidence for the role of reactive oxygen species and JNK , J Biol Chem, Vol.286 ; pp.5546-5557
    32. Xu Y , Kim SO , Li Y , Han J (2006) Autophagy contributes to caspase-independent macrophage cell death , J Biol Chem, Vol.281 ; pp.19179-19187
    33. Xu Y , Huang S , Liu ZG , Han J (2006) Poly(ADP-ribose) polymerase-1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2-mediated JNK1 activation , J Biol Chem, Vol.281 ; pp.8788-8795
    34. Chen SY , Chiu LY , Chien CL , Lin WW (2011) zVAD-induced autophagic cell death requires c-Src-dependent ERK and JNK activation and reactive oxygen species generation , Autophagy, Vol.7 ; pp.217-228
    35. Moriwaki K , Bertin J , Orlowski GM , Chan FK (2015) Differential roles of RIPK1 and RIPK3 in TNF-induced necroptosis and chemotherapeutic agent-induced cell death , Cell Death Dis, Vol.6 ; pp.e1636
    36. Chromik J , Safferthal C , Serve H , Fulda S (2014) Smac mimetic primes apoptosis-resistant acute myeloid leukaemia cells for cytarabine-induced cell death by triggering necroptosis , Cancer Lett, Vol.344 ; pp.101-109
    37. Laukens B , Jennewein C , Vandenabeele P , Fulda S (2011) Smac mimetic bypasses apoptosis resistance in FADD- or caspase-8-deficient cells by priming for tumor necrosis factor alpha-induced necroptosis , Neoplasia, Vol.13 ; pp.971-979
    38. Steinhart L , Belz K , Fulda S (2013) Smac mimetic and demethylating agents synergistically trigger cell death in acute myeloid leukemia cells and overcome apoptosis resistance by inducing necroptosis , Cell Death Dis, Vol.4 ; pp.e802
    오늘하루 팝업창 안보기 닫기