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.4 pp.193-199
DOI : https://doi.org/10.11620/IJOB.2014.39.4.193

NaF-induced Autophagy on SCC25 Human Tongue Squamous Cell Carcinoma Cells

Jin-Mo Kang1, Bo-Young Lee1, In-Ryoung Kim1, Yong-Ho Kim1, Su-Bin Yu1, Hae-Ryoun Park2, Bong-Soo Park1*
1Department of Oral Anatomy, School of Dentistry, Pusan National University
2Department of Oral Pathology, 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
October 15, 2014 November 10, 2014 November 24, 2014

Abstract

Fluoride has been accepted as an important material for oral health and is widely used to prevent dental caries in dentistry. However, its safety is still questioned by some. Autophagy has been implicated in cancer cell survival and death, and may play an important role in oral cancer. This study was undertaken to examine whether sodium fluoride (NaF) modulates autophagy in SCC25 human tongue squamous cell carcinoma cells. NaF demonstrated anticancer activity via autophagic and apoptotic cell death. Autophagic vacuoles were detectable using observed to form by monodansylcadaverine (MDC) and acridine orange (AO). Analysis of NaF-treated SCC25 cells for the presence of biochemical markers revealed direct effects on the conversion of LC-3II, degradation of p62/SQSTM1, cleavage formation of ATG5 and Beclin-1, and caspase activation. NaF-induced cell death was suppressed by the autophagy inhibitor 3-methyladenine (3-MA). NaF-induced autophagy was confirmed as a pro-death signal in SCC25 cells. These results implicate NaF as a novel anticancer compound for oral cancer therapy.


초록


    Pusan National University

    Introduction

    Fluoride is extensively distributed in water, soils, and the atmosphere, causing constant exposure of human beings and animals to fluoride compounds [1]. The principal sources of ingested fluoride are drinking water or beverages produced with fluoride-containing dental products, and food [1,2]. Fluoride is widely used as a remedy to dental caries, although controversy remains about the safety. While long-term fluoride ingestion for the prevention of dental caries may lead to side effects asserious acute toxicity [3,4]. Nevertheless, fluoride has been considered as an important material for oral health care.

    Autophagy is an evolutionary phenomenon, long-lived or damaged proteins and cellular organelles by internal and external stimuli are digested in lysosomes and it is reuse as energy or lead to death within cell [5,6]. However, autophagy also promotes cancer cell survival under conditions of stress, functioning as a defense mechanism in response to various anticancer drugs [7,8]. Therefore, the induction of autophagic cell death by anti-cancer reagents has been recognized to play an important role in cancer therapy [9-11].

    Oral squamous cell carcinoma (OSCC) is the most common and persistent malignant tumor of the oral cavity, with nearly 500,000 patients suffering from this disease each year world-wide [12,13]. Three major treatment of oral cancer including surgery, radiation and chemotherapy are currently applied, but it is difficult to cure because successful treatment is not always achieved in patients [14]. Although the OSCC patients are treated with classical treatment modalities, noticeably high mortality rates are still observed [15-17]. Therefore, new therapeutic approaches are being investigated, with the use of natural agents being one of the most promising anti-cancer treatment options.

    Treatment with sodium fluoride (NaF) has also been examined in the course of OSCC research, and was shown to cause the death of OSCC cells via apoptosis. However, no reports have yet examined the effects of NaF on autophagic cell death in the SCC25 human tongue squamous carcinoma cells. In the present study, we examined whether NaF can induces autophagic and apoptotic cell death in SCC25 cells, and established the molecular mechanism of association between autophagy and apoptosis.

    Materials and Methods

    Reagents

    Acridine orange (AO), monodansylcadaverine (MDC), and 3-[4,5-dimethylthiazol-2-yl]2,5-diphenyl tetrazolium bromide (MTT) were purchased from Sigma (St. Louis, MO, USA). 3-methyladenine (3-MA) was obtained from Calbiochem (La Jolla, CA, USA). Antibodies against Beclin-1, caspase-3, caspase-8, and PARP were purchased from Cell Signaling Technology (Beverly, MA, USA). Antibodies against microtubule-associated protein 1 light chain 3 (LC3) (Sigma, MO, USA) were also used. The autophagy protein 5 (ATG5), caspase-9, GAPDH, and p62/SQSTM1 were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The mouse anti-rabbit IgG antibody, and rabbit anti-mouse IgG secondary antibodies were purchased from Enzo Life Sciences (Farmingdale, NY, USA). All other chemicals and reagents were purchased from Sigma, unless otherwise specified.

    Cell culture

    The SCC25 human oral squamous cell carcinoma cell line was purchased from ATCC (Rockville, MD, USA) and maintained as monolayers in Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM⁄F-12) with supplemented with 10% FBS and 100 μg/ml streptomycin at 37°C in a humidified atmosphere containing 5% CO2.

    Treatment of NaF

    The stock solutions of NaF (100 mM) were dissolved in distilled water, and were prepared as needed. NaF stock solution was diluted to the working concentration with media where needed. Cells were cultured about 80% confluence in wells or plates, and then treated to various concentrations of NaF (0-2 mM) for 24 h. Cells grown in medium without NaF served as the control and Earle’s Balanced Salt Solution (EBSS) used as an autophagy-positive control.

    MTT assay

    SCC25 cells (1 × 104) were seeded and incubated for 24 h in tissue culture-coated 96-well plate. Then, cells were treated with various concentrations of NaF (0-2 mM) for 24-72 h. After treatment, thiazolyl blue tetrazolium bromide (MTT) solution (500 μg/ml) was added to the plates, and they were incubated at 37°C for 4 h. The medium was aspirated and 100 μl DMSO was added for dissolving the formazan crystals. Cell viability was measured by an ELISA reader (Tecan, Männedorf, Switzerland) at the excitatory emission wavelength of 570 nm.

    Flow cytometer analysis

    After drug treatment, cells (1 × 106) were harvested by trypsinization. For fix the cells, ice cold 95% ethanol and 0.5% Tween 20 were added until a final ethanol concentration of 70%. The fixed cells were pelleted and then washed PBS in 1% BSA. Cells were resuspended in 1 ml PBS containing 20 μg/ml RNase A, incubated on ice for 30 min, and finally added 10 μl propidium iodide (PI) stock solution (1 mg/ml). After incubation on ice for 2 min, DNA content was measured and analyzed on a CYTOMICS FC500 flow cytometry system (Beckman Coulter, FL, CA, USA) to allow simultaneous estimation of apoptosis. Induction of autophagy in cells, shows acidic vesicular organelles (AVOs), it can be orescentstained red and detected in FL3 red-fluorescence by acridine orange (AO). To find a ratio of NaF induced autophagy, cells were stained with AO (0.1 μg/mL) for 5 min, analyzed using a FACScan flow cytometer. For inhibition of autophagy, cells were pretreated with 1mM 3-MA for 1 h before incubation with NaF for 24 h.

    Fluorescence microscopy

    5 × 104 cells were grown on coverslips in 24-well plate and treated with NaF. After treatment, cells were stained with monodansylcadaverine (MDC) (0.05 mM) for 30 min and acridine orange (AO) (0.1 μg/mL) for 15 min at 37°C. Acidic vesicular organelles (AVOs) stained with both MDC and AO fluorescent, and cells were observed using a fluorescence microscope (Axioskop, Carl Zeiss, Germany). EBSS was using as a positive control of autophagy.

    Western blot analysis

    2 x 106 cells were harvested using a cell scraper, washed twice with ice-cold PBS, and lysed ice-cold RIPA lysis buffer (Invitrogen, Carlsbad, CA, USA) and incubated on ice for 1 h. The lysates were then centrifuged at 14,000 rpm for 15 min at 4°C. Protein quantitation was measured using a Bradford protein assay (Bio-Rad, Richmond, CA, USA), and aliquots of the lysates (20 μg protein) were resolved by 10% SDS/PAGE. The gels were transferred on a to polyvinylidene fluoride (PVDF) membranes (Millipore, Billerica, MA, USA). The membranes were kept with 5% non fat dry milk blocking buffer, and incubated with primary antibodies and secondary antibodies for 1 h. Immunoblotting of potein was accomplished via the SuperSignal West Femto (Pierce, Rockford, IL, USA) improved chemiluminescence substrate, detected with an Alpha Imager HP (Alpha Innotech, Santa Clara, USA).

    Statistic analysis

    All results were representative of at least 3 independent experiments and are expressed as the mean ± SEM (standard error of the mean). Statistical analyses were performed by using the SPSS version 14.0 statistical software program (SPSS Inc., IL, USA). A one-way ANOVA for multiple comparison was used for statistical analysis.

    Results

    NaF induces apoptosis in SCC25

    The cytotoxicity of NaF was investigated over a wide concentration range in SCC25 cells. Cells were treated with NaF (0-2 mM) for 24 h, after which viability was assessed using the MTT assay. NaF concentrations from 0 to 2 mM potently induced cell death in SCC25 cells (Fig. 1A). Treatment with 1 mM NaF resulted in decrease of cell viability in a time-dependent manner (Fig. 1B). Thus, the viability of SCC25 cells was decreased in both a dose- and time-dependent manner by NaF treatment. A flow cytometry assay was undertaken to test whether the induction of cell death was mediated via apoptosis. The percentage of sub diploid cells, indicative of apoptotic cells, was found to increase in a dose-dependent fashion by NaF treatment (Fig. 2).

    NaF induced autophagy in SCC25 cells

    It was next investigated whether autophagy occurs in the SCC25 cells treated with NaF. When cells were stained with monodansylcadaverine (MDC), a selective marker of autophagic vesicles, the NaF-treated SCC25 cells exhibited staining autophagic vesicles as punctate compared to the control (Fig. 3A). The formation of autophagic vacuoles due to NaF was also confirmed by staining the acidic vesicular organelles (AVOs) with acridine orange (AO). The AVOs, which represent autophagic vacuoles, were clearly formed in the SCC25 cells (red fluorescence). As shown in Fig. 3B, red-stained AVOs were observed following treatment with 1 mM NaF for 24 h. These findings indicate that treatment of NaF is sufficient to promote autophagic response in SCC25 cells, as observed the autophagic vacuoles by MDC and AO staining.

    NaF promotes the formation of autophagic and apoptotic molecules in SCC25 cells

    Herein, NaF was also examined for the ability to induce autophagy in SCC25 cells through observation of various autophagy markers, including ATG5, Beclin-1, p62/SQSTM1, and LC3. LC3, the only reliable marker of autophagosomes, was increased in the NaF-treated SCC25 cells in a dose-dependent manner. In contrast, the level of p62/SQSTM1, a protein degraded by autophagy, and Beclin-1 were reduced in the NaF-treated SCC25 cells (Fig. 4A). The mechanism of NaF-induced cell death were further investigated by examining the levels of PARP cleavage and the cleavage of procaspase to active caspase-8, caspase-9 and caspase-3, markers of apoptotic activity in SCC25 cells. As shown in Fig. 4B, NaF induced the activation of caspase-8, caspase-9, and caspase-3, also producing 85 kDa processed PARP fragment.

    3-MA inhibits NaF-induced apoptosis and autophagy

    The role of NaF-induced autophagy in SCC25 was further clarified by investigating the effects of treatment with 3-methyladenine (3-MA) as known autophagy inhibitors, on the NaF-treated SCC25 cells. To confirm whether 3-MA can inhibit autophagy, we determined the cell viability of NaF-treated cell lines after pretreatment with 1 mM 3-MA for 1 h. Pretreatment with 3-MA slightly increased the viability of the NaF-treated cells (Fig. 5A). 3-MA also inhibited the induction of AVOs by NaF. This was confirmed by AO staining by measurement of the red-to-green fluorescence ratio by FACS (Fig. 5B). Therefore, NaF induced autophagy and 3-MA inhibited this process, resulting in reduced cell death. In conclusion, these results suggest that NaF induces autophagy and regards death signals leading to apoptosis in SCC25 cells.

    Discussion

    Human beings and animals are constantly exposed to fluoride compounds, at greater levels than ever due to the increased use of fluorides in industry, medicine, and dentistry [1,18]. Sodium fluoride (NaF), the most widely used material of fluorides, showed no significant cytotoxicity in OSCC cells at a low dose in the present study. However, NaF has been reported to induce apoptosis via the intrinsic pathway in HL-60 cells at relatively high doses [19-21]. The possible connection between NaF-induced apoptotic cell death and glycolysis [22] or autophagy [23] has also been reported.

    Autophagy is widely known as an important process in cell physiology, for both cell survival and death [24]. It starts with the elimination of cytoplasmic organelles in a double-membrane vacuole, an autophagosome, after which they are delivered to a degradative organelle, the vacuole/ lysosome, for breakdown and eventual recycling of the resulting macromolecules. Because numerous recent studies have shown increased autophagic activity to be associated with cell death [10,25], autophagy is now considered as a type of cell death [26]. One of the recent studies reported that autophagy may play a critical role as a protective response in ameloblasts as a result of NaF-induced cell stress. Therefore, the necessity of performed more studies emerged, as the molecular mechanism between NaF-induced apoptosis and autophagy has not been clearly identified.

    Previous studies in our laboratory also showed that NaF induced cell death in SCC25 cells via apoptosis. The effect of NaF in the autophagic process of SCC25 has not yet been determined. The present data demonstrated that the NaF-treated SCC25 cells had decreased viability, and underwent cell death via apoptosis and autophagy (Fig. 1, 2). The autophagic effects of NaF in SCC25 cells were confirmed via AO and MDC staining. NaF induced the formation of cytoplasmic vacuoles and acidic vesicular organelles (AVOs) in the SCC25 cells (Fig. 3A, B).

    The NaF-treated SCC25 cells were also analyzed for the presence of biochemical markers of autophagy, including p62/SQSTM1, LC3, ATG5-ATG12 complex and Beclin-1. NaF treatment directly affected the conversion of LC-3II, degradation of p62/SQSTM1 and full-length Beclin-1, and cleavage formation of the ATG5-ATG12 complex (Fig. 4A). Several previous studies reported that a relationship may exist between LC3 and p62/SQSTM1, which is selectively degraded in cells undergoing autophagy [27-30]. Autophagy mediates a nonspecific bulk degradation pathway responsible for the degradation of the majority of long-lived proteins and some organelles. ATG5-ATG12 conjugation systems are necessary for the formation of the autophagosome [31]. Beclin-1 (Bcl-2-interacting protein-1) is a key protein in autophagy signaling, functioning with Vps34, UVRAG, AMBRA-1, and Barkor to assemble the PI3KC3 complex during initiation of autophagosome formation [32-34]. Several studies using different cell types and stimuli recently described that caspase-mediated cleavage of Beclin-1 and ATG proteins enhances apoptosis [35-38]. The results obtained herein showed that NaF led to the degradation of caspase-8, caspase-9, and caspase-3, and assumed a decisive role on Beclin-1 (Fig. 4B). The role of autophagy in SCC25 was further clarified by demonstration that the NaF-induced cell death was suppressed by 3-MA, an inhibitor of autophagy. This result strongly implies that NaF-induced autophagy is a pro-death rather than a pro-survival signal (Fig. 5A, B).

    In the present study, we reported for the first time that treatment with NaF induced autophagic cell death in SCC25 cells. NaF induced autophagy as a pro-death signal prior to apoptosis in SCC25, hence, treatment of oral cancer with NaF could be a potentially useful therapeutic strategy. Finding the molecular mechanisms behind autophagic cell death induced by NaF is expected to be useful information for the development possibility as a new therapeutic potential against malignant oral cancer.

    Conflict of interest

    The authors declare that they have no competing interest.

    Figure

    IJOB-39-193_F1.gif

    Cytotoxic effect of NaF on SCC25 cells. NaF treated cell viability was determined by MTT assay. (A) Cells were treated with various concentrations of NaF (0-2 mM) for 24 h. (B) To investigate cell viability according to the time variation, cells were treated with NaF (1 mM) for 24-72 hours. Data show mean±SEM of three independent experiments. *p < 0.05, **p < 0.01

    IJOB-39-193_F2.gif

    NaF induced apoptotic cell death in SCC25 cells. Cells were treated with NaF (0-2 mM) for 24 h, and the percent of apoptotic cells was determined by FACS.

    IJOB-39-193_F3.gif

    NaF showed induction of autophagy in SCC25 cells. Cells were seeded in 24-well plates and NaF (1 mM) was treatment 24 h later. NaF induced autophagy was using MDC and acridine orange staining, it was analyzed by fluorescence microscopy. EBSS was used as a positive control of autophagy.

    IJOB-39-193_F4.gif

    Autophagy and apoptosis-related proteins were activated by NaF treatement. NaF treatment were performed at various concentrations (0-2 mM) in SCC25 cells. Autophagy-related proteins such as p62/SQSTM1, LC3, ATG5, Beclin-1 and apoptosis–related proteins such as caspase-8, caspase-9, caspase-3, PARP were measured by western blot analysis.

    IJOB-39-193_F5.gif

    NaF-induced autophagy was usually inhibited by 3-MA in SCC25 cells. (A) Cells were pretreated with 3-MA (1 mM) for 1 h, and then exposed to NaF (1 mM ) for 24 h. Cell viability were analyzed by the MTT assay. (B) Cells were stained with MDC (a) and acridine orange (b), observed with a fluorescence microscope. The ratio of red fluorescence was quantified using FACS flow cytometry (c). Data show mean±SEM of three independent experiments. *p < 0.05.

    Table

    Reference

    1. Ozsvath D (2009) Fluoride and environmental health a review , Rev Environ Sci Biotechnol, Vol.8; pp.59-79
    2. Petersen P.E , Lennon M.A (2004) Effective use of fluorides for the prevention of dental caries in the 21st century the WHO approach , Community Dent Oral Epidemiol, Vol.32; pp.319-21
    3. Li Y (1993) Fluoride safety issues , J Indiana Dent Assoc, Vol.72; pp.22-26
    4. Lee J.H , Jung J.Y , Jeong Y.J , Park J.H , Yang K.H , Choi N.K , Kim S.H , Kim W.J (2008) Involvement of both mitochondrial- and death receptor-dependent apoptotic pathways regulated by Bcl-2 family in sodium fluoride-induced apoptosis of the human gingival fibroblasts , Toxicology, Vol.243; pp.340-347
    5. Yorimitsu T , Klionsky D.J (2005) Autophagy molecular machinery for self-eating , Cell Death Differ, Vol.12 (Suppl 2); pp.1542-1552
    6. Codogno P (2005) Autophagy in cell survival and death , J Soc Biol, Vol.199; pp.233-241
    7. Zhang J.Q , Li Y.M , Chen X.H , Liu T , Chen Y.T , He W.T , Zhang Q.B , Liu S.Y (2011) Autophagy is involved in anticancer effects of matrine on SGC-7901 human gastric cancer cells , Oncol Rep, Vol.26; pp.115-124
    8. Michaud M , Martins I , Sukkurwala A.Q , Adjemian S , Ma Y , Pellegatti P , Shen S , Kepp O , Scoazec M , Mignot G , Rello-Varona S , Tailler M , Menger L , Vacchelli E , Galluzzi L , Ghiringhelli F , di Virgilio F , Zitvogel L , roemer G (2011) Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice , Science, Vol.334; pp.1573-1577
    9. Giannopoulou E , Antonacopoulou A , Matsouka P , Kalofonos H.P (2009) Autophagy Novel Action of Panitumumab in Colon Cancer , Anticancer Res, Vol.29; pp.5077-5082
    10. Gozuacik D , Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism , Oncogene, Vol.23; pp.2891-2906
    11. Guo X.L , Li D , Hu F , Song J.R , Zhang S.S , Deng W.J , Sun K , Zhao Q.D , Xie X.Q , Song Y.J , Wu M.C , Wei L.X (2012) Targeting autophagy potentiates chemotherapy-induced apoptosis and proliferation inhibition in hepatocarcinoma cells , Cancer Lett, Vol.320; pp.171-179
    12. Clayman G.L , Ebihara S , Terada M , Mukai K , Goepfert H (1997) Report of the Tenth International Symposium of the Foundation for Promotion of Cancer Research Basic and clinical research in head and neck cancer , Jpn J Clin Oncol, Vol.27; pp.361-368
    13. Shen J , Huang C.H , Jiang L , Gao F , Wang Z , Zhang Y.Y , Bai J.P , Zhou H.M , Chen Q.M (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
    14. Suzuki M , Endo M , Shinohara F , Echigo S , Rikiishi H (2009) Enhancement of cisplatin cytotoxicity by SAHA involves endoplasmic reticulum stress-mediated apoptosis in oral squamous cell carcinoma cells , Cancer Chemother Pharmacol, Vol.64; pp.1115-1122
    15. Bell R.B , Kademani D , Homer L , Dierks E.J , Potter B.E (2007) Tongue cancer Is there a difference in survival compared with other subsites in the oral cavity , J Oral Maxil Surg, Vol.65; pp.229-236
    16. Lo W.L , Kao S.Y , Chi L.Y , Wong Y.K , Chang R.C.S (2003) Outcomes of oral squamous cell carcinoma in Taiwan after surgical therapy Factors affecting survival , J Oral Maxil Surg, Vol.61; pp.751-758
    17. Shintani S , Li C.N , Mihara M , Klosek S.K , Terakado N , Hino S , Hamakawa H (2006) Anti-tumor effect of radiation response by combined treatment with angiogenesis inhibitor, TNP-470, in oral squamous cell carcinoma , Oral Oncol, Vol.42; pp.66-72
    18. Hansen K.J , Clemen L.A , Ellefson M.E , Johnson H.O (2001) Compound-specific, quantitative characterization of organic fluorochemicals in biological matrices , Environ Sci Technol, Vol.35; pp.766-770
    19. Song J.S , Lee H.Y , Lee E , Hwang H.J , Kim J.H (2002) Cytotoxicity and apoptosis induction of sodium fluoride in human promyelocytic leukemia (HL-60):cells , Environ Toxicol Phar, Vol.11; pp.85-91
    20. Anuradha C.D , Kanno S , Hirano S (2000) Fluoride induces apoptosis by caspase-3 activation in human leukemia HL-60 cells , Arch Toxicol, Vol.74; pp.226-230
    21. Anuradha C.D , Kanno S , Hirano S (2001) Oxidative damage to mitochondria is a preliminary step to caspase-3 activation in fluoride-induced apoptosis in HL-60 cells , Free Radical Bio Med, Vol.31; pp.367-373
    22. Otsuki S , Morshed S , Chowdhury S , Takayama F , Satoh T , Hashimoto K , Sugiyama K , Amano O , Yasui T , Yokote Y (2005) Possible link between glycolysis and apoptosis induced by sodium fluoride , J Dent Res, Vol.84; pp.919-923
    23. Suzuki M , Bartlett J.D (2014) Sirtuin1 and autophagy protect cells from fluoride-induced cell stress , Biochim Biophys Acta, Vol.1842; pp.245-255
    24. Lum J.J , Bauer D.E , Kong M , Harris M.H , Li C , Lindsten T , Thompson C.B (2005) Growth factor regulation of autophagy andcell survival in the absence of apoptosis , Cell, Vol.120; pp.237-248
    25. Tsujimoto Y , Shimizu S (2005) Another way to die autophagic programmed cell death , Cell Death Differ, Vol.12; pp.1528-1534
    26. Levine B , Yuan J (2005) Autophagy in cell death an innocent convict? , J Clin Invest, Vol.115; pp.2679-2688
    27. Chen L.H , Loong C.C , Su T.L , Lee Y.J , Chu P.M , Tsai M.L , Tsai P.H , Tu P.H , Chi C.W , Lee H.C , Chiou S.H (2011) Autophagy inhibition enhances apoptosis triggered by BO-1051, an N-mustard derivative, and involves the ATM signaling pathway , Biochem Pharmacol, Vol.81; pp.594-605
    28. Ichimura Y , Komatsu M (2010) Selective degradation of p62 by autophagy , Semin Immunopathol, Vol.32; pp.431-436
    29. Ichimura Y , Kominami E , Tanaka K , Komatsu M (2008) Selective turnover of p62/A170/SQSTM1 by autophagy , Autophagy, Vol.4; pp.1063-1066
    30. Pankiv S , Clausen T.H , Lamark T , Brech A , Bruun J.A , Outzen H , Overvatn A , Bjorkoy G , Johansen T (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy , J Biol Chem, Vol.282; pp.24131-24145
    31. Jounai N , Takeshita F , Kobiyama K , Sawano A , Miyawaki A , Xin K.Q , Ishii K.J , Kawai T , Akira S , Suzuki K , Okuda K (2007) The Atg5 Atg12 conjugate associates with innate antiviral immune responses , Proc Natl Acad Sci USA, Vol.104; pp.14050-14055
    32. Fimia G.M , Stoykova A , Romagnoli A , Giunta L , Di Bartolomeo S , Nardacci R , Corazzari M , Fuoco C , Ucar A , Schwartz P , Gruss P , Piacentini M , Chowdhury K , Cecconi F (2007) Ambra1 regulates autophagy and development of the nervous system , Nature, Vol.447; pp.1121-1125
    33. Kihara A , Kabeya Y , Ohsumi Y , Yoshimori T (2001) Beclinphosphatidylinositol 3-kinase complex functions at the trans-Golgi network , Embo Rep, Vol.2; pp.330-335
    34. Sun Q.M , Fan W.L , Zhong Q (2009) Regulation of Beclin 1 in autophagy , Autophagy, Vol.5; pp.713-716
    35. Betin V.M.S , Lane J.D (2009) Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis , J Cell Sci, Vol.122; pp.2554-2566
    36. Cho D.H , Jo Y.K , Hwang J.J , Lee Y.M , Roh S.A , Kim J.C (2009) Caspase-mediated cleavage of ATG6/Beclin-1 links apoptosis to autophagy in HeLa cells , Cancer Lett, Vol.274; pp.95-100
    37. Djavaheri-Mergny M , Maiuri M.C , Kroemer G (2010) Cross talk between apoptosis and autophagy by caspase-mediated cleavage of Beclin 1 , Oncogene, Vol.29; pp.1717-1719
    38. Wirawan E , Vande Walle L , Kersse K , Cornelis S , Claerhout S , Vanoverberghe I , Roelandt R , De Rycke R , Verspurten J , Declercq W , Agostinis P , Vanden Berghe T , Lippens S , Vandenabeele P (2010) Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria , Cell Death Dis, Vol.1; pp.e18