簡易檢索 / 詳目顯示

研究生: 馬格帝
Ahmed Magdy Abu El-Fotouh
論文名稱: 砷對A549肺腺癌細胞自噬作用與一氧化氮之探討
The role of autophagy and nitric oxide in arsenite-induced cytotoxicity and genotoxicity in lung adenocarcinoma A549 cells
指導教授: 楊嘉鈴
Yang, Jia-Ling
口試委員: 王翊青
Wang, I-Ching
鄒粹軍
Tsou, Tsui-Chun
學位類別: 碩士
Master
系所名稱: 生命科學暨醫學院 - 生物科技研究所
Biotechnology
論文出版年: 2016
畢業學年度: 104
語文別: 英文
論文頁數: 63
中文關鍵詞: 肺癌自噬一氧化氮
外文關鍵詞: Lung cancer, Arsenic, Autophagy, Nitric oxide
相關次數: 點閱:2下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 哺乳類細胞接觸到類金屬砷導致細胞毒性以及基因上的損害,但在砷誘導後產生的自噬作用機轉尚未清楚。自噬作用在演化上具有高度保守性,其自噬細胞代謝過程中會將受損的胞器或是異常的蛋白降解,適當的自噬作用是為了讓細胞免於傷害,過多現象產生反而容易造成細胞死亡。另外也發現一氧化氮在調節癌細胞存活與死亡扮演很重要之角色。因此,進而希望能夠釐清砷在A549肺腺癌引起的自噬作用以及氧化作用。為了觀察到自噬作用是否存在A549肺腺癌,使用吖啶橙染色 (acridine orange staining, AO staining)判定到自噬泡的生成,並以西方點墨法(western blot)觀察微管結合蛋白第三型輕鍊-I(LC3-I)的轉換,形成 LC3-II的堆積。藉由螢光顯微鏡證實以砷誘導細胞產生自噬作用,更進一步驗證先前的實驗。此外,以細胞自噬抑制劑如spautin-1, chloroquine, or 3-methyladenine進行細胞毒殺試驗(MTT assay)以及癌細胞群落形成能力試驗(colony forming ability assay),發現加入細胞自噬抑制劑反而導致了癌細胞的死亡,推測細胞自噬作用在含有砷的環境壓力下為保護細胞的機制。另一方面,為了更深入了解砷刺激細胞後,一氧化氮是如何影響細胞存活,以不同的調節劑觀察一氧化氮。從實驗的結果,我們認為一氧化氮的生成是為了提高細胞存活對抗砷毒性。最後藉由細胞形成微核(micronuclei)的能力探討自噬作用與對基因的潛在影響,也進一步驗證自噬作用與一氧化氮是避免基因受到砷毒害而產生的機制以維持基因之穩定性。


    It is well known that arsenic induces cytotoxicity and genotoxicity in mammalian cells, and arsenic-induced autophagy was clearly demonstrated. Autophagy is an evolutionarily conserved, catabolic process that maintains cellular homeostasis by regulating organelles and proteins turnover and, meantime, induces autophagic cell
    death. In line with this, nitric oxide is also known for its dual role in cancer cells by regulating cell survival and cell death. We have recently identified that arsenite can induce nitric oxide progression in A549 lung adenocarcinoma cell line. So, I focused my thesis studies on understanding the role of autophagy and nitric oxide mediated by arsenite in cytotoxicity and genotoxicity in A549 non-small cell lung cancer cells. To accomplish the previously stated goal, I have performed the acridine orange staining assay to detect the activation of autophagosome formation, and used western blot technique to monitor the conversion of LC3B-I to LC3B-II, which is a key marker protein for autophagy induction. The preliminary results from fluorescent microscopy have been further validated by western blot analysis, which confirmed the role of arsenite in inducing autophagy. Moreover, the results obtained from MTT assay and colony forming ability assay showed that treating the cells with autophagy inhibitors, such as spautin-1, chloroquine, or 3-methyladenine, enhanced arsenite-induced cell death suggesting that autophagy has a protective role under arsenite stress. In addition, to study the role of nitric oxide in regulating cell viability under arsenite treatment; I treated the cells with nitric oxide modulators, such as PTIO, 1400W, and GSNO, and the results obtained from MTT assay suggested that NO has a prosurvival role against arsenite toxicity. Moreover, investigating the potential function of autophagy and nitric oxide in genome stability via assessing the ability of the cells to form micronuclei was also conducted, and the results suggested that arsenite-induced
    autophagy is required for suppressing genotoxicity following arsenite exposure.
    Interestingly, the results obtained from micronucleus assay suggested that nitric oxide is critically required for genome stability in normal and stressful conditions.

    DEDICATION …………………...…………………………………..………………. I ACKNOWLEDGEMENT ……………..……………………………………………. II ABBREVIATIONS…………………………………………………………………. III ABSTRACT ……………………………………………………………...………… IV CHAPTER 1. Chapter 1. Introduction ………………………………………………………. 1 1.1. Lung cancer …………..…………………………………………………… 1 1.1.1. Lung cancer definition and statistics ……………………...…………. 1 1.1.2. Lung cancer classification …...………………………………………. 1 1.1.3. Smoking and lung cancer incidence …………………………………. 2 1.1.4. Pathogenesis of lung cancer: molecular mechanisms and genetic mutations ……………..…………………………………………………………. 2 1.1.5. Etiology of lung cancer ……………………………………………… 3 1.1.6. Signs and symptoms of lung cancer ……………….………………… 3 1.1.7. Lung cancer tests and diagnosis ………….………………………….. 3 1.1.8. Lung cancer staging (TNM) …………………………………………. 4 1.2. Arsenic ….………………………………………………………………… 4 1.2.1. Definition ……………………………………………………………. 4 1.2.2. Arsenic sources ……………………..……………………………….. 5 1.2.3. Arsenic and arsenic compounds …..…………………………………. 6 1.2.4. Arsenic toxicity …………..………………………………………….. 7 1.2.5. Arsenic effects on laboratory animals ……………………………….. 7 1.2.6. Human exposure to arsenic and health effects ...…………………….. 8 1.2.7. Arsenic and cancer development …….………………………………. 9 1.2.8. Therapeutic effects of arsenic ..………………………………………. 9 1.3. Autophagy .………………………………………………………………. 10 1.3.1. Overview of autophagy .……………………………………………. 10 1.3.2. The autophagy machinery ………………………………………….. 10 1.3.3. Autophagy regulation and cancer ...………………………………… 12 1.3.4. Autophagy inhibitors ………..……………………………………… 12 1.4. Nitric oxide ……….……………………………………………………… 14 1.4.1. Overview of nitric oxide ….………………………………………… 14 1.4.2. Nitric oxide synthases ………..…………………………………….. 14 1.4.3. Nitric oxide and lung carcinogenesis …….………………………… 15 1.5. Aims of the study ……...………………………………………………… 15 2. Chapter 2. Material and methods ...………………………………………… 16 2.1. Reagent and antibodies ...………………………………………………… 16 2.2. Reagent preparation and storage …...……………………………………. 16 2.3. Cell culture …………….………………………………………………… 16 2.4. Arsenite treatment …..…………………………………………………… 17 2.5. Cell viability assays ……………………………………………………… 17 2.6. Clonogenic assay …...……………………………………………………. 18 2.7. Cytokinesis-block micronucleus assay ...………………………………… 19 2.8. Flow cytometry analysis of nitric oxide production ….………………….. 19 2.9. Cell lysate extraction and preparation …………………………………… 19 2.10. Western blot analysis .……………………………………………………. 20 2.11. Acridine orange staining analysis by fluorescence microscope …...…….. 21 2.12. Statistical analysis ….……………………………………………………. 21 3. Chapter 3. Results …………………………………………………………… 22 3.1. Arsenite induces autophagy in lung adenocarcinoma A549 cells …………… 22 3.2. Autophagy inhibitors enhance cell death in arsenite-treated A549 cells ......… 22 3.3. Autophagy inhibitors reduce colony-forming ability in arsenite-treated A549 cells ……………...…………………………………………………………… 23 3.4. Arsenite induces nitric oxide (NO) generation in A549 cells ……………..…. 23 3.5. NO inhibitors increase arsenite-induced cell death in A549 cells ...….……… 24 3.6. Autophagy inhibitors increase arsenite-induced genotoxicity ………….……. 24 3.7. Nitric oxide counteracts genome instability induced by arsenite stress in A549 cells ...………………………………………………………………………… 25 4. Chapter 4. Discussion .………..……………………………………………… 26 5. Chapter 5. Reference .………...……………………………………………… 31 6. Figures …………...…………………………………………………………… 43 6.1. Figure 1.Arsenite induces autophagy in A549 lung adenocarcinoma cells ..… 43 6.2. Figure 2. Autophagy inhibitors enhance arsenite-induced cell death …….….. 45 6.3. Figure 3. Autophagy inhibitors enhance arsenite-induced cytotoxicity …..…. 47 6.4. Figure 4. Validation of nitric oxide (NO) induction following arsenite exposure …………………………………...…………………………………. 49 6.5. Figure 5. Effect of NO modulators on arsenite-induced cell death …..……… 50 6.6. Figure 6. Autophagy inhibitors increase genotoxicity induced by arsenite in A549 cells ...……….…………………………………………………………. 52 6.7. Figure 7. Effect of NO modulators on arsenite-induced genotoxicity .………. 54

    Amaravadi, R. K., and Winkler, J. D. (2012). Lys05: a new lysosomal autophagy inhibitor. Autophagy 8, 1383-1384.
    Arnold, A., Davis, S., and Jordan, A. L. (1969). The determination of arsenic in organic and inorganic arsenic compounds: a radioisotope-dilution substoicheiometric application. The Analyst 94, 664-669.
    Barth, S., Glick, D., and Macleod, K. F. (2010). Autophagy: assays and artifacts. The Journal of pathology 221, 117-124.
    Bath, P. M., Willmot, M., Leonardi-Bee, J., and Bath, F. J. (2002). Nitric oxide donors (nitrates), L-arginine, or nitric oxide synthase inhibitors for acute stroke. The Cochrane database of systematic reviews, CD000398.
    Beckles, M. A., Spiro, S. G., Colice, G. L., and Rudd, R. M. (2003). Initial evaluation of the patient with lung cancer: symptoms, signs, laboratory tests, and paraneoplastic syndromes. Chest 123, 97S-104S.
    Benbrahim-Tallaa, L., and Waalkes, M. P. (2008). Inorganic arsenic and human prostate cancer. Environmental health perspectives 116, 158-164.
    Bilello, K. S., Murin, S., and Matthay, R. A. (2002). Epidemiology, etiology, and prevention of lung cancer. Clinics in chest medicine 23, 1-25.
    Biswas, S., Talukder, G., and Sharma, A. (1999). Prevention of cytotoxic effects of arsenic by short-term dietary supplementation with selenium in mice in vivo. Mutation research 441, 155-160.
    Blanco, R., Iwakawa, R., Tang, M., Kohno, T., Angulo, B., Pio, R., Montuenga, L. M., Minna, J. D., Yokota, J., and Sanchez-Cespedes, M. (2009). A gene-alteration profile of human lung cancer cell lines. Human mutation 30, 1199-1206.
    Boucher, J. L., Moali, C., and Tenu, J. P. (1999). Nitric oxide biosynthesis, nitric oxide synthase inhibitors and arginase competition for L-arginine utilization. Cellular and molecular life sciences : CMLS 55, 1015-1028.
    Brero, A., Ramella, R., Fitou, A., Dati, C., Alloatti, G., Gallo, M. P., and Levi, R. (2010). Neuregulin-1beta1 rapidly modulates nitric oxide synthesis and calcium handling in rat cardiomyocytes. Cardiovascular research 88, 443-452.
    Bryan, N. S., Bian, K., and Murad, F. (2009). Discovery of the nitric oxide signaling pathway and targets for drug development. Frontiers in bioscience 14, 1-18.
    Cai, X., Jia, P., and Shi, X. (1998). [In vitro study on arsenic trioxide-induced apoptosis of retinoic acid resistant acute promyelocytic leukemia cell line(MR-2)]. Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi 19, 339-341.
    Cancer Genome Atlas Research, N. (2014). Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543-550.
    Celik, I., Gallicchio, L., Boyd, K., Lam, T. K., Matanoski, G., Tao, X., Shiels, M., Hammond, E., Chen, L., Robinson, K. A., et al. (2008). Arsenic in drinking water and lung cancer: a systematic review. Environmental research 108, 48-55.
    Chang, T. K., Shravage, B. V., Hayes, S. D., Powers, C. M., Simin, R. T., Wade Harper, J., and Baehrecke, E. H. (2013). Uba1 functions in Atg7- and Atg3-independent autophagy. Nature cell biology 15, 1067-1078.
    Chen, G. G., Lee, T. W., Xu, H., Yip, J. H., Li, M., Mok, T. S., and Yim, A. P. (2008). Increased inducible nitric oxide synthase in lung carcinoma of smokers. Cancer 112, 372-381.
    Chen, G. Q., Shi, X. G., Tang, W., Xiong, S. M., Zhu, J., Cai, X., Han, Z. G., Ni, J. H., Shi, G. Y., Jia, P. M., et al. (1997). Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells. Blood 89, 3345-3353.
    Cheng, J., Wei, H. L., Chen, J., and Xie, B. (2012). Antitumor effect of arsenic trioxide in human K562 and K562/ADM cells by autophagy. Toxicology mechanisms and methods 22, 512-519.
    Chiang, C. E., Luk, H. N., Wang, T. M., and Ding, P. Y. (2002). Prolongation of cardiac repolarization by arsenic trioxide. Blood 100, 2249-2252.
    Chiarelli, R., and Roccheri, M. C. (2012). Heavy metals and metalloids as autophagy inducing agents: focus on cadmium and arsenic. Cells 1, 597-616.
    Chiu, H. W., Ho, Y. S., and Wang, Y. J. (2011a). Arsenic trioxide induces autophagy and apoptosis in human glioma cells in vitro and in vivo through downregulation of survivin. Journal of molecular medicine 89, 927-941.
    Chiu, H. W., Lin, J. H., Chen, Y. A., Ho, S. Y., and Wang, Y. J. (2010). Combination treatment with arsenic trioxide and irradiation enhances cell-killing effects in human fibrosarcoma cells in vitro and in vivo through induction of both autophagy and apoptosis. Autophagy 6, 353-365.
    Chiu, H. W., Lin, W., Ho, S. Y., and Wang, Y. J. (2011b). Synergistic effects of arsenic trioxide and radiation in osteosarcoma cells through the induction of both autophagy and apoptosis. Radiation research 175, 547-560.
    Clarke, P. G. (1990). Developmental cell death: morphological diversity and multiple mechanisms. Anatomy and embryology 181, 195-213.
    Cobbs, C. S., Brenman, J. E., Aldape, K. D., Bredt, D. S., and Israel, M. A. (1995). Expression of nitric oxide synthase in human central nervous system tumors. Cancer research 55, 727-730.
    Conrad, M. E. (1999). Treatment of acute promyelocytic leukemia with arsenic trioxide. The New England journal of medicine 340, 1043-1045.
    Dahal, B. M., Fuerhacker, M., Mentler, A., Karki, K. B., Shrestha, R. R., and Blum, W. E. (2008). Arsenic contamination of soils and agricultural plants through irrigation water in Nepal. Environmental pollution 155, 157-163.
    De Villaine, S., Mesguich, P., Fabien, N., Isaac, S., Rochet, M., and Paulin, C. (1996). [Evaluation of the role of cytology in the diagnosis of cancer of the lung. Comparison between cytology and pathological anatomy in 330 cases of proximal cancers]. Rev Mal Respir 13, 295-299.
    Degenhardt, K., Mathew, R., Beaudoin, B., Bray, K., Anderson, D., Chen, G., Mukherjee, C., Shi, Y., Gelinas, C., Fan, Y., et al. (2006). Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer cell 10, 51-64.
    Deknudt, G., Leonard, A., Arany, J., Jenar-Du Buisson, G., and Delavignette, E. (1986). In vivo studies in male mice on the mutagenic effects of inorganic arsenic. Mutagenesis 1, 33-34.
    Delgado, J., Martinez, L. M., Sanchez, T. T., Ramirez, A., Iturria, C., and Gonzalez-Avila, G. (2005). Lung cancer pathogenesis associated with wood smoke exposure. Chest 128, 124-131.
    Dilda, P. J., and Hogg, P. J. (2007). Arsenical-based cancer drugs. Cancer treatment reviews 33, 542-564.
    Ettinger, D. S., Wood, D. E., Akerley, W., Bazhenova, L. A., Borghaei, H., Camidge, D. R., Cheney, R. T., Chirieac, L. R., D'Amico, T. A., Demmy, T. L., et al. (2014). Non-small cell lung cancer, version 1.2015. J Natl Compr Canc Netw 12, 1738-1761.
    Fenech, M. (2007). Cytokinesis-block micronucleus cytome assay. Nature protocols 2, 1084-1104.
    Florea, A. M., Yamoah, E. N., and Dopp, E. (2005). Intracellular calcium disturbances induced by arsenic and its methylated derivatives in relation to genomic damage and apoptosis induction. Environmental health perspectives 113, 659-664.
    Forstermann, U., Closs, E. I., Pollock, J. S., Nakane, M., Schwarz, P., Gath, I., and Kleinert, H. (1994). Nitric oxide synthase isozymes. Characterization, purification, molecular cloning, and functions. Hypertension 23, 1121-1131.
    Fulda, S., and Kogel, D. (2015). Cell death by autophagy: emerging molecular mechanisms and implications for cancer therapy. Oncogene 34, 5105-5113.
    Ge, F., Zhang, Y., Cao, F., Li, J., Hou, J., Wang, P., Li, H., Xu, M., Liu, S., Li, L., et al. (2016). Arsenic trioxide-based therapy is suitable for patients with psoriasis-associated acute promyelocytic leukemia - A retrospective clinical study. Hematology 21, 287-294.
    Gebel, T. (1998). Suppression of arsenic-induced chromosome mutagenicity by antimony. Mutation research 412, 213-218.
    Ghavamzadeh, A., Alimoghaddam, K., Ghaffari, S. H., Rostami, S., Jahani, M., Hosseini, R., Mossavi, A., Baybordi, E., Khodabadeh, A., Iravani, M., et al. (2006). Treatment of acute promyelocytic leukemia with arsenic trioxide without ATRA and/or chemotherapy. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO 17, 131-134.
    Gomez, S. E., del Razo, L. M., and Munoz Sanchez, J. L. (2005). Induction of DNA damage by free radicals generated either by organic or inorganic arsenic (AsIII, MMAIII, and DMAIII) in cultures of B and T lymphocytes. Biological trace element research 108, 115-126.
    Goussetis, D. J., Altman, J. K., Glaser, H., McNeer, J. L., Tallman, M. S., and Platanias, L. C. (2010). Autophagy is a critical mechanism for the induction of the antileukemic effects of arsenic trioxide. The Journal of biological chemistry 285, 29989-29997.
    Guha Mazumder, D. N., Chakraborty, A. K., Ghose, A., Gupta, J. D., Chakraborty, D. P., Dey, S. B., and Chattopadhyay, N. (1988). Chronic arsenic toxicity from drinking tubewell water in rural West Bengal. Bulletin of the World Health Organization 66, 499-506.
    Gurr, J. R., Yih, L. H., Samikkannu, T., Bau, D. T., Lin, S. Y., and Jan, K. Y. (2003). Nitric oxide production by arsenite. Mutation research 533, 173-182.
    Haddad, I. Y., Pataki, G., Hu, P., Galliani, C., Beckman, J. S., and Matalon, S. (1994). Quantitation of nitrotyrosine levels in lung sections of patients and animals with acute lung injury. The Journal of clinical investigation 94, 2407-2413.
    Hammond, E. C. (1966). Smoking in relation to the death rates of one million men and women. Natl Cancer Inst Monogr 19, 127-204.
    Harada, K., Supriatno, Kawaguchi, S., Tomitaro, O., Yoshida, H., and Sato, M. (2004). Overexpression of iNOS gene suppresses the tumorigenicity and metastasis of oral cancer cells. In vivo 18, 449-455.
    Hecht, S. S. (1999). Tobacco smoke carcinogens and lung cancer. J Natl Cancer Inst 91, 1194-1210.
    Hikita, E., Arai, M., Tanaka, S., Onda, K., Utsumi, H., Yuan, B., Toyoda, H., and Hirano, T. (2011). Effects of inorganic and organic arsenic compounds on growth and apoptosis of human T-lymphoblastoid leukemia cells. Anticancer research 31, 4169-4178.
    Hou, Y. C., Janczuk, A., and Wang, P. G. (1999). Current trends in the development of nitric oxide donors. Current pharmaceutical design 5, 417-441.
    Hughes, M. F. (2002). Arsenic toxicity and potential mechanisms of action. Toxicology letters 133, 1-16.
    Jagirdar, J. (2008). Application of immunohistochemistry to the diagnosis of primary and metastatic carcinoma to the lung. Arch Pathol Lab Med 132, 384-396.
    Jemal, A., Siegel, R., Xu, J., and Ward, E. (2010). Cancer statistics, 2010. CA Cancer J Clin 60, 277-300.
    Jia, P. M., Zhu, Q., Yu, Y., Chen, G. Q., Chen, S. J., Chen, Z., Wang, Z. Y., and Tong, J. H. (2002). [Experimental study of low dose arsenic trioxide in treatment of patients with acute promyelocytic leukemia]. Ai zheng = Aizheng = Chinese journal of cancer 21, 337-340.
    Jiao, G., Ren, T., Guo, W., Ren, C., and Yang, K. (2015). Arsenic trioxide inhibits growth of human chondrosarcoma cells through G2/M arrest and apoptosis as well as autophagy. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 36, 3969-3977.
    Jin, M., and Klionsky, D. J. (2014). Regulation of autophagy: modulation of the size and number of autophagosomes. FEBS letters 588, 2457-2463.
    Johnson, J. L., Pillai, S., and Chellappan, S. P. (2012). Genetic and biochemical alterations in non-small cell lung cancer. Biochem Res Int 2012, 940405.
    Jung, H. J., Yang, M. Z., Kwon, K. H., Yenari, M. A., Choi, Y. J., Lee, W. T., Park, K. A., and Lee, J. E. (2010). Endogenous agmatine inhibits cerebral vascular matrix metalloproteinases expression by regulating activating transcription factor 3 and endothelial nitric oxide synthesis. Current neurovascular research 7, 201-212.
    Kang, M. R., Kim, M. S., Oh, J. E., Kim, Y. R., Song, S. Y., Kim, S. S., Ahn, C. H., Yoo, N. J., and Lee, S. H. (2009). Frameshift mutations of autophagy-related genes ATG2B, ATG5, ATG9B and ATG12 in gastric and colorectal cancers with microsatellite instability. The Journal of pathology 217, 702-706.
    Kanzawa, T., Zhang, L., Xiao, L., Germano, I. M., Kondo, Y., and Kondo, S. (2005). Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene 24, 980-991.
    Kao, Y. H., Yu, C. L., Chang, L. W., and Yu, H. S. (2003). Low concentrations of arsenic induce vascular endothelial growth factor and nitric oxide release and stimulate angiogenesis in vitro. Chemical research in toxicology 16, 460-468.
    Karantza-Wadsworth, V., Patel, S., Kravchuk, O., Chen, G., Mathew, R., Jin, S., and White, E. (2007). Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes & development 21, 1621-1635.
    Klionsky, D. J., Abeliovich, H., Agostinis, P., Agrawal, D. K., Aliev, G., Askew, D. S., Baba, M., Baehrecke, E. H., Bahr, B. A., Ballabio, A., et al. (2008a). Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 4, 151-175.
    Klionsky, D. J., Elazar, Z., Seglen, P. O., and Rubinsztein, D. C. (2008b). Does bafilomycin A1 block the fusion of autophagosomes with lysosomes? Autophagy 4, 849-850.
    Kroemer, G., Galluzzi, L., Vandenabeele, P., Abrams, J., Alnemri, E. S., Baehrecke, E. H., Blagosklonny, M. V., El-Deiry, W. S., Golstein, P., Green, D. R., et al. (2009). Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell death and differentiation 16, 3-11.
    Kumar, A., Singh, U. K., and Chaudhary, A. (2015). Targeting autophagy to overcome drug resistance in cancer therapy. Future medicinal chemistry 7, 1535-1542.
    Kusunoki, Y., Imamura, F., Uda, H., Mano, M., and Horai, T. (2000). Early detection of lung cancer with laser-induced fluorescence endoscopy and spectrofluorometry. Chest 118, 1776-1782.
    Langer, C. J., Besse, B., Gualberto, A., Brambilla, E., and Soria, J. C. (2010). The evolving role of histology in the management of advanced non-small-cell lung cancer. J Clin Oncol 28, 5311-5320.
    Lee-Chen, S. F., Yu, C. T., and Jan, K. Y. (1992). Effect of arsenite on the DNA repair of UV-irradiated Chinese hamster ovary cells. Mutagenesis 7, 51-55.
    Lee, T. C., Huang, R. Y., and Jan, K. Y. (1985a). Sodium arsenite enhances the cytotoxicity, clastogenicity, and 6-thioguanine-resistant mutagenicity of ultraviolet light in Chinese hamster ovary cells. Mutation research 148, 83-89.
    Lee, T. C., Lee, K. C., Tzeng, Y. J., Huang, R. Y., and Jan, K. Y. (1986a). Sodium arsenite potentiates the clastogenicity and mutagenicity of DNA crosslinking agents. Environmental mutagenesis 8, 119-128.
    Lee, T. C., Oshimura, M., and Barrett, J. C. (1985b). Comparison of arsenic-induced cell transformation, cytotoxicity, mutation and cytogenetic effects in Syrian hamster embryo cells in culture. Carcinogenesis 6, 1421-1426.
    Lee, T. C., Wang-Wuu, S., Huang, R. Y., Lee, K. C., and Jan, K. Y. (1986b). Differential effects of pre- and posttreatment of sodium arsenite on the genotoxicity of methyl methanesulfonate in Chinese hamster ovary cells. Cancer research 46, 1854-1857.
    Levine, B., and Klionsky, D. J. (2004). Development by self-digestion: molecular mechanisms and biological functions of autophagy. Developmental cell 6, 463-477.
    Levine, B., and Kroemer, G. (2008). Autophagy in the pathogenesis of disease. Cell 132, 27-42.
    Li, C. L., Wei, H. L., Chen, J., Wang, B., Xie, B., Fan, L. L., and Li, L. J. (2014). Arsenic trioxide induces autophagy and antitumor effects in Burkitt's lymphoma Raji cells. Oncology reports 32, 1557-1563.
    Li, N., Li, X., Li, S., Zhou, S., and Zhou, Q. (2013). Cisplatin-induced downregulation of SOX1 increases drug resistance by activating autophagy in non-small cell lung cancer cell. Biochemical and biophysical research communications 439, 187-190.
    Li, Y., Sun, X., Wang, L., Zhou, Z., and Kang, Y. J. (2002). Myocardial toxicity of arsenic trioxide in a mouse model. Cardiovascular toxicology 2, 63-73.
    Liang, X. H., Jackson, S., Seaman, M., Brown, K., Kempkes, B., Hibshoosh, H., and Levine, B. (1999). Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402, 672-676.
    Liu, J., Xia, H., Kim, M., Xu, L., Li, Y., Zhang, L., Cai, Y., Norberg, H. V., Zhang, T., Furuya, T., et al. (2011). Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell 147, 223-234.
    Liu, S. X., Davidson, M. M., Tang, X., Walker, W. F., Athar, M., Ivanov, V., and Hei, T. K. (2005). Mitochondrial damage mediates genotoxicity of arsenic in mammalian cells. Cancer research 65, 3236-3242.
    Mao, Y., Yang, D., He, J., and Krasna, M. J. (2016). Epidemiology of Lung Cancer. Surg Oncol Clin N Am 25, 439-445.
    Marino, G., Niso-Santano, M., Baehrecke, E. H., and Kroemer, G. (2014). Self-consumption: the interplay of autophagy and apoptosis. Nature reviews Molecular cell biology 15, 81-94.
    Masri, F. A., Comhair, S. A., Koeck, T., Xu, W., Janocha, A., Ghosh, S., Dweik, R. A., Golish, J., Kinter, M., Stuehr, D. J., et al. (2005). Abnormalities in nitric oxide and its derivatives in lung cancer. American journal of respiratory and critical care medicine 172, 597-605.
    Matsui, A., Kamada, Y., and Matsuura, A. (2013). The role of autophagy in genome stability through suppression of abnormal mitosis under starvation. PLoS genetics 9, e1003245.
    Mehrpour, M., Esclatine, A., Beau, I., and Codogno, P. (2010). Autophagy in health and disease. 1. Regulation and significance of autophagy: an overview. American journal of physiology Cell physiology 298, C776-785.
    Michel, T., and Feron, O. (1997). Nitric oxide synthases: which, where, how, and why? The Journal of clinical investigation 100, 2146-2152.
    Miller, Y. E. (2005). Pathogenesis of lung cancer: 100 year report. Am J Respir Cell Mol Biol 33, 216-223.
    Mizushima, N., Yoshimori, T., and Levine, B. (2010). Methods in mammalian autophagy research. Cell 140, 313-326.
    Mizushima, N., Yoshimori, T., and Ohsumi, Y. (2003). Role of the Apg12 conjugation system in mammalian autophagy. The international journal of biochemistry & cell biology 35, 553-561.
    Mountain, C. F. (1997). Revisions in the International System for Staging Lung Cancer. Chest 111, 1710-1717.
    Mukhopadhyay, S., and Katzenstein, A. L. (2011). Subclassification of non-small cell lung carcinomas lacking morphologic differentiation on biopsy specimens: Utility of an immunohistochemical panel containing TTF-1, napsin A, p63, and CK5/6. Am J Surg Pathol 35, 15-25.
    Mulshine, J. L., Cuttitta, F., Tockman, M. S., and De Luca, L. M. (2002). Lung cancer evolution to preinvasive management. Clinics in chest medicine 23, 37-48.
    Naseem, K. M. (2005). The role of nitric oxide in cardiovascular diseases. Molecular aspects of medicine 26, 33-65.
    National Toxicology, P. (2002). Arsenic compounds, inorganic. Report on carcinogens : carcinogen profiles / US Dept of Health and Human Services, Public Health Service, National Toxicology Program 10, 17-19.
    Navarro-Yepes, J., Burns, M., Anandhan, A., Khalimonchuk, O., del Razo, L. M., Quintanilla-Vega, B., Pappa, A., Panayiotidis, M. I., and Franco, R. (2014). Oxidative stress, redox signaling, and autophagy: cell death versus survival. Antioxidants & redox signaling 21, 66-85.
    Nishida, Y., Arakawa, S., Fujitani, K., Yamaguchi, H., Mizuta, T., Kanaseki, T., Komatsu, M., Otsu, K., Tsujimoto, Y., and Shimizu, S. (2009). Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 461, 654-658.
    Ogata, M., Hino, S., Saito, A., Morikawa, K., Kondo, S., Kanemoto, S., Murakami, T., Taniguchi, M., Tanii, I., Yoshinaga, K., et al. (2006). Autophagy is activated for cell survival after endoplasmic reticulum stress. Molecular and cellular biology 26, 9220-9231.
    Paglin, S., Hollister, T., Delohery, T., Hackett, N., McMahill, M., Sphicas, E., Domingo, D., and Yahalom, J. (2001). A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer research 61, 439-444.
    Park, J. H., Kim, E. J., Jang, H. Y., Shim, H., Lee, K. K., Jo, H. J., Kim, H. J., Yang, S. H., Jeong, E. T., and Kim, H. R. (2008). Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncology reports 20, 379-384.
    Puhakka, A. R., Harju, T. H., Paakko, P. K., Soini, Y. M., and Kinnula, V. L. (2006). Nitric oxide synthases are associated with bronchial dysplasia. Lung cancer 51, 275-282.
    Qi, Y., Li, H., Zhang, M., Zhang, T., Frank, J., and Chen, G. (2014). Autophagy in arsenic carcinogenesis. Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie 66, 163-168.
    Qian, J., Qin, S., He, Z., Wang, L., Chen, Y., Shao, Z., and Liu, X. (2002). [Arsenic trioxide for the treatment of medium and advanced primary liver cancer]. Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese journal of hepatology 10, 63.
    Qu, X., Yu, J., Bhagat, G., Furuya, N., Hibshoosh, H., Troxel, A., Rosen, J., Eskelinen, E. L., Mizushima, N., Ohsumi, Y., et al. (2003). Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. The Journal of clinical investigation 112, 1809-1820.
    Quatrehomme, G., Ricq, O., Lapalus, P., Jacomet, Y., and Ollier, A. (1992). Acute arsenic intoxication: forensic and toxicologic aspects (an observation). Journal of forensic sciences 37, 1163-1171.
    Radosavljevic, V., and Jakovljevic, B. (2008). Arsenic and bladder cancer: observations and suggestions. Journal of environmental health 71, 40-42.
    Ratnaike, R. N. (2003). Acute and chronic arsenic toxicity. Postgraduate medical journal 79, 391-396.
    Reveneau, S., Arnould, L., Jolimoy, G., Hilpert, S., Lejeune, P., Saint-Giorgio, V., Belichard, C., and Jeannin, J. F. (1999). Nitric oxide synthase in human breast cancer is associated with tumor grade, proliferation rate, and expression of progesterone receptors. Laboratory investigation; a journal of technical methods and pathology 79, 1215-1225.
    Rosbe, K. W., Prazma, J., Petrusz, P., Mims, W., Ball, S. S., and Weissler, M. C. (1995). Immunohistochemical characterization of nitric oxide synthase activity in squamous cell carcinoma of the head and neck. Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery 113, 541-549.
    RoyChoudhury, A., Das, T., Sharma, A., and Talukder, G. (1996). Dietary garlic extract in modifying clastogenic effects of inorganic arsenic in mice: two-generation studies. Mutation research 359, 165-170.
    Sancar, A., Lindsey-Boltz, L. A., Unsal-Kacmaz, K., and Linn, S. (2004). Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annual review of biochemistry 73, 39-85.
    Sarkar, S., Korolchuk, V. I., Renna, M., Imarisio, S., Fleming, A., Williams, A., Garcia-Arencibia, M., Rose, C., Luo, S., Underwood, B. R., et al. (2011). Complex inhibitory effects of nitric oxide on autophagy. Molecular cell 43, 19-32.
    Schairer, D. O., Chouake, J. S., Nosanchuk, J. D., and Friedman, A. J. (2012). The potential of nitric oxide releasing therapies as antimicrobial agents. Virulence 3, 271-279.
    Schoolmeester, W. L., and White, D. R. (1980). Arsenic poisoning. Southern medical journal 73, 198-208.
    Sellers, A. H. (1971). The clinical classification of malignant tumours: the TNM system. Can Med Assoc J 105, 836 passim.
    Shang, Z. J., Li, J. R., and Li, Z. B. (2002). Effects of exogenous nitric oxide on oral squamous cell carcinoma: an in vitro study. Journal of oral and maxillofacial surgery : official journal of the American Association of Oral and Maxillofacial Surgeons 60, 905-910; discussion 910-901.
    Shao, S., Li, S., Qin, Y., Wang, X., Yang, Y., Bai, H., Zhou, L., Zhao, C., and Wang, C. (2014). Spautin-1, a novel autophagy inhibitor, enhances imatinib-induced apoptosis in chronic myeloid leukemia. International journal of oncology 44, 1661-1668.
    Shen, Z. Y., Tan, L. J., Cai, W. J., Shen, J., Chen, C., Tang, X. M., and Zheng, M. H. (1999). Arsenic trioxide induces apoptosis of oesophageal carcinoma in vitro. International journal of molecular medicine 4, 33-37.
    Simeonova, P. P., Wang, S., Toriuma, W., Kommineni, V., Matheson, J., Unimye, N., Kayama, F., Harki, D., Ding, M., Vallyathan, V., and Luster, M. I. (2000). Arsenic mediates cell proliferation and gene expression in the bladder epithelium: association with activating protein-1 transactivation. Cancer research 60, 3445-3453.
    Sokolov, M. V., Panyutin, I. G., and Neumann, R. D. (2012). Whole-genome gene expression profiling reveals the major role of nitric oxide in mediating the cellular transcriptional response to ionizing radiation in normal human fibroblasts. Genomics 100, 277-281.
    Spiro, S. G., and Porter, J. C. (2002). Lung cancer--where are we today? Current advances in staging and nonsurgical treatment. American journal of respiratory and critical care medicine 166, 1166-1196.
    Stella, G. M., Scabini, R., Inghilleri, S., Cemmi, F., Corso, S., Pozzi, E., Morbini, P., Valentini, A., Dore, R., Ferrari, S., et al. (2013). EGFR and KRAS mutational profiling in fresh non-small cell lung cancer (NSCLC) cells. J Cancer Res Clin Oncol 139, 1327-1335.
    Stewart, A. G., Phan, L. H., and Grigoriadis, G. (1994). Physiological and pathophysiological roles of nitric oxide. Microsurgery 15, 693-702.
    Suda, K., Tomizawa, K., Yatabe, Y., and Mitsudomi, T. (2011). Lung cancers unrelated to smoking: characterized by single oncogene addiction? Int J Clin Oncol 16, 294-305.
    Sun, S., Schiller, J. H., and Gazdar, A. F. (2007). Lung cancer in never smokers--a different disease. Nat Rev Cancer 7, 778-790.
    Surdu, S. (2014). Non-melanoma skin cancer: occupational risk from UV light and arsenic exposure. Reviews on environmental health 29, 255-264.
    Tartarone, A., Rossi, E., Lerose, R., Mambella, G., Calderone, G., Zamarchi, R., and Aieta, M. (2016). Possible applications of circulating tumor cells in patients with non small cell lung cancer. Lung cancer.
    Toh, C. K., and Lim, W. T. (2007). Lung cancer in never-smokers. J Clin Pathol 60, 337-340.
    Tomida, S., Yatabe, Y., Yanagisawa, K., Mitsudomi, T., and Takahashi, T. (2005). Throwing new light on lung cancer pathogenesis: updates on three recent topics. Cancer Sci 96, 63-68.
    Torre, L. A., Bray, F., Siegel, R. L., Ferlay, J., Lortet-Tieulent, J., and Jemal, A. (2015). Global cancer statistics, 2012. CA Cancer J Clin 65, 87-108.
    Travis, W. D., Brambilla, E., Noguchi, M., Nicholson, A. G., Geisinger, K., Yatabe, Y., Powell, C. A., Beer, D., Riely, G., Garg, K., et al. (2011). International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society: international multidisciplinary classification of lung adenocarcinoma: executive summary. Proc Am Thorac Soc 8, 381-385.
    Tseng, W. P. (1977). Effects and dose--response relationships of skin cancer and blackfoot disease with arsenic. Environmental health perspectives 19, 109-119.
    Uslu, R., Sanli, U. A., Sezgin, C., Karabulut, B., Terzioglu, E., Omay, S. B., and Goker, E. (2000). Arsenic trioxide-mediated cytotoxicity and apoptosis in prostate and ovarian carcinoma cell lines. Clinical cancer research : an official journal of the American Association for Cancer Research 6, 4957-4964.
    Walter, F. M., Rubin, G., Bankhead, C., Morris, H. C., Hall, N., Mills, K., Dobson, C., Rintoul, R. C., Hamilton, W., and Emery, J. (2015). Symptoms and other factors associated with time to diagnosis and stage of lung cancer: a prospective cohort study. Br J Cancer 112 Suppl 1, S6-13.
    Wang, J. P., Qi, L., Moore, M. R., and Ng, J. C. (2002). A review of animal models for the study of arsenic carcinogenesis. Toxicology letters 133, 17-31.
    Wang, W., Cheng, S., and Zhang, D. (2014). Association of inorganic arsenic exposure with liver cancer mortality: A meta-analysis. Environmental research 135, 120-125.
    Wang, Z. G., Rivi, R., Delva, L., Konig, A., Scheinberg, D. A., Gambacorti-Passerini, C., Gabrilove, J. L., Warrell, R. P., Jr., and Pandolfi, P. P. (1998). Arsenic trioxide and melarsoprol induce programmed cell death in myeloid leukemia cell lines and function in a PML and PML-RARalpha independent manner. Blood 92, 1497-1504.
    Wei, X. M., Wang, Q., Gao, S. J., and Sui, L. (2011). [Relationship between nitric oxide in cervical microenvironment and different HPV types and effect on cervical cancer cells]. Zhonghua fu chan ke za zhi 46, 260-265.
    World Health, O. (2000). Air quality guidelines for Europe. WHO regional publications European series, V-X, 1-273.
    Wu, Y. T., Tan, H. L., Shui, G., Bauvy, C., Huang, Q., Wenk, M. R., Ong, C. N., Codogno, P., and Shen, H. M. (2010). Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. The Journal of biological chemistry 285, 10850-10861.
    Xie, D., Yin, S., Ou, Y., Bai, H., Ding, F., Wang, X., Liu, Z., Zhou, C., and Wu, M. (2002). Arsenic trioxide (As(2)O(3)) induced apoptosis and its mechanisms in a human esophageal squamous carcinoma cell line. Chinese medical journal 115, 280-285.
    Xie, R., Wang, F., McKeehan, W. L., and Liu, L. (2011). Autophagy enhanced by microtubule- and mitochondrion-associated MAP1S suppresses genome instability and hepatocarcinogenesis. Cancer research 71, 7537-7546.
    Xie, Z., and Klionsky, D. J. (2007). Autophagosome formation: core machinery and adaptations. Nature cell biology 9, 1102-1109.
    Yamamoto, A., Tagawa, Y., Yoshimori, T., Moriyama, Y., Masaki, R., and Tashiro, Y. (1998). Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell structure and function 23, 33-42.
    Yang, Y. P., Liang, Z. Q., Gao, B., Jia, Y. L., and Qin, Z. H. (2008). Dynamic effects of autophagy on arsenic trioxide-induced death of human leukemia cell line HL60 cells. Acta pharmacologica Sinica 29, 123-134.
    Yang, Z., and Klionsky, D. J. (2010). Eaten alive: a history of macroautophagy. Nature cell biology 12, 814-822.
    Yano, T., Okamoto, T., Fukuyama, S., and Maehara, Y. (2014). Therapeutic strategy for postoperative recurrence in patients with non-small cell lung cancer. World J Clin Oncol 5, 1048-1054.
    Youlden, D. R., Cramb, S. M., and Baade, P. D. (2008). The International Epidemiology of Lung Cancer: geographical distribution and secular trends. J Thorac Oncol 3, 819-831.
    Yu, L., Gu, C., Zhong, D., Shi, L., Kong, Y., Zhou, Z., and Liu, S. (2014). Induction of autophagy counteracts the anticancer effect of cisplatin in human esophageal cancer cells with acquired drug resistance. Cancer letters 355, 34-45.
    Yue, Z., Jin, S., Yang, C., Levine, A. J., and Heintz, N. (2003). Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proceedings of the National Academy of Sciences of the United States of America 100, 15077-15082.
    Zarazua, S., Perez-Severiano, F., Delgado, J. M., Martinez, L. M., Ortiz-Perez, D., and Jimenez-Capdeville, M. E. (2006). Decreased nitric oxide production in the rat brain after chronic arsenic exposure. Neurochemical research 31, 1069-1077.
    Zhang, T. C., Cao, E. H., Li, J. F., Ma, W., and Qin, J. F. (1999). Induction of apoptosis and inhibition of human gastric cancer MGC-803 cell growth by arsenic trioxide. European journal of cancer 35, 1258-1263.

    無法下載圖示 全文公開日期 本全文未授權公開 (校內網路)
    全文公開日期 本全文未授權公開 (校外網路)

    QR CODE