Responsive image
博碩士論文 etd-0211111-023201 詳細資訊
Title page for etd-0211111-023201
論文名稱
Title
Manganese superoxide Dismutase (SOD2)過度表現對抑制肝癌細胞致癌性的研究
Overexpression of Manganese Superoxide Dismutase (SOD2) Inhibited the Tumorigenicity of Hepatoma Cells
系所名稱
Department
畢業學年期
Year, semester
語文別
Language
學位類別
Degree
頁數
Number of pages
61
研究生
Author
指導教授
Advisor
召集委員
Convenor
口試委員
Advisory Committee
口試日期
Date of Exam
2010-07-16
繳交日期
Date of Submission
2011-02-11
關鍵字
Keywords
活性氧化物、肝細胞癌、超氧陰離子、過氧化氫、錳超氧化物歧化酶
Hydrogen peroxide, reactive oxygen species, Mn superoxide dismutase, Hepatocellular carcinoma, superoxide anions
統計
Statistics
本論文已被瀏覽 5728 次,被下載 1006
The thesis/dissertation has been browsed 5728 times, has been downloaded 1006 times.
中文摘要
在台灣肝細胞癌是最常見的一種惡性腫瘤。 肝臟細胞癌化,會由於氧化壓力間的活性氧化物(ROS)和解毒作用不平衡而發生。當細胞受到自由基破壞時,超氧化物歧化酶(SODS)發揮關鍵性的解毒作用,保護細胞來至自由基導致的損傷。錳超氧化物歧化酶(MnSOD或SOD2)是超氧化物歧化酶(SODS)成員之ㄧ存在於線粒體內。SOD2生理作用是將線粒體內具毒性的超氧陰離子( O2- )轉變成過氧化氫( H2O2 )。在人類肝癌組織樣本中已經證實SOD2表現量減弱,而SOD2表現減弱與肝癌細胞形成的關聯性目前仍然不清楚。在本論文中,首先證明在人類惡性肝癌細胞株SK-Hep-1細胞中SOD2 的表現量較良性肝癌細胞株少。此外, 在141例手術切除之肝癌樣本中,其中97例( 68.8 % ) SOD2的表現明顯低於鄰近非腫瘤組織。運用腺病毒傳送SOD2基因在SK-Hep-1肝癌細胞中,會增加SOD2蛋白表現量和過氧化氫(H2O2)產量, 然而會減少超氧陰離子( O2- )含量。而且,在SOD2基因傳送明顯著抑制SK-Hep-1細胞的增殖,轉移和菌落生長。在體內動物實驗中, SOD2的過度表現會抑制腫瘤的生長且SOD2的增加能有效的抑制腫瘤細胞生長的速度及大小。在流式細胞儀實驗分析顯示,SOD2基因傳送誘導抑制肝癌細胞之細胞週期停滯在G2/M期,經由減少cdc2/cdk1和 cyclin B1的表現,及增加p21Cip1的生成有相互關聯性。然而,SOD2過度表現不影響matrix metalloproteinase-2 (MMP-2) and MMP-9的分泌。結論,本論文證明SOD2過度表達可抑制肝癌細胞的致癌性發生,有可能運用於肝癌治療。
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and devastating malignant tumors in Taiwan. Due to an imbalanced between reactive oxygen species (ROS) production and detoxification, oxidative stress, has been implicated in liver carcinogenesis. Superoxide dismutases (SODS) play a key role in the detoxification of superoxide radicals and thus protect cells from damage induced by free radicals. Manganese superoxide dismutase (MnSOD or SOD2) is a member of the superoxide dismutase family located in mitochondria. SOD2 transforms toxic superoxide, a byproduct of the mitochondrial electron transport chain, into hydrogen peroxide and diatomic oxygen. Though reduced SOD2 protein level and activities have been reported in hepatoma tissues, it remains unclear how SOD2 expression affected the tumorigenic processes of hepatoma cells. Expression analysis of an array of human HCC cell lines revealed that SOD2 were down-regulated in poorly differentiated SK-Hep-1 hepatoma cells. Moreover, SOD2 is downregulated in 68.8% of resected HCC samples (97 out of 141 cases). Adenovirus-mediated SOD2 gene delivery increased the cellular SOD2 protein level and H2O2 production, but reduced the superoxide anion level in SK-Hep-1 cells. Furthermore, SOD2 restoration significantly reduced the proliferation, motility, and colony formation of SK-Hep-1 cells. In vivo animal model, the finding of SOD2 overexpression inhibited the proliferation of Sk-Hep-1 hepatoma cells while reduced the tumor growth in mice. Flow cytometry analysis showed that SOD2 gene transfer inhibited the growth of hepatoma cells through induction of cell cycle arrest at G2/M phase. This was associated with declined cdc2/cdk1 and cyclin B1 expression and upregulation p21Cip1 by SOD2 gene delivery. However, SOD2 overexpression had no effect on the secretion of matrix metalloproteinase-2 (MMP-2) and MMP-9.In conclusion, SOD2 overexpression suppresses the tumorigenicity of hepatoma cells and may hold promise for HCC treatment.
目次 Table of Contents
CONTENTS
Page
Approval letter ........................................................... ii
Abstract in Chinese................................................... iii
Abstract in English…………………………………. iv
Abbreviation……………………………………….... vii
Introduction ....……………………………………… 1
Specific Aims ...…………………………………….. 8
Materials and Methods ...…………………………. 9
Results ....................................................................... 18
Discussion ............................................................... 25
Figures and Legends .............................................. 29
Table I ........................................................................ 46
Table II ....................................................................... 47
Appenedix ................................................................. 48
References................................................................. 49
參考文獻 References
1.Yuen MF, Hou JL, Chutaputti A. Asia Pacific Working Party on Prevention of Hepatocellular Carcinoma. Hepatocellular carcinoma in the Asia pacific region. J Gastroenterol Hepatol 2009;24:346-353.
2.Hu TH, Huang CC, Lin PR, Chang HW, Ger LP, Lin YW, Changchien CS, Lee CM, Tai MH.Expression and prognostic role of tumor suppressorgene
PTEN/MMAC1/TEP1 in hepatocellular arcinoma.Cancer 2003;97:1929-1940.
3.Chen CJ, Liang KY, Chang AS. Effects of hepatitis B virus, alcohol drinking, cigarette smoking and familial tendency on hepatocellular carcinoma. Hepatology 1991;13:398 - 406.
4.Chen CJ, Wang LY, Lu SN, et al. Elevated aflatoxin exposure and increased risk of hepatocellular carcinoma. Hepatology 1996;24:38-42.
5.Qian GS, Ross RK, Yu MC, et al. A follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, People’s Republic of China. Cancer Epidemiol Biomarkers Prev 1994;3:3 – 10.
6.Wu HC, Wang Q, Yang HI, Ahsan H, Tsai WY, Wang LY, Chen SY, Chen CJ, Santella RM. Aflatoxin B1 Exposure, Hepatitis B Virus Infection, and Hepatocellular
Carcinoma in Taiwan. Cancer Epidemiol Biomarkers Prev 2009;18:846-853.
7.Waris G, Ahsan H. Reactive oxygen species: role in the development of cancer and various chronic conditions. J Carcinog 2006;5:14.
8.Hagen TM, Huang S, Curnutte J, Fowler P, Martinez V, Wehr CM,
Ames BN, Chisari FV: Extensive oxidative DNA damage in hepatocytes of transgenic mice with chronic active hepatitis destined to develop hepatocellular carcinoma. Proc
Natl Acad Sci U S A 1994; 91:12808-12812.
9.Waris G, Siddiqui A: Regulatory mechanisms of viral hepatitis B and. J Biosci 2003; 28:311-321.

10.Bag A, Bag N. Target Sequence Polymorphism of Human Manganese Superoxide Dismutase Gene and Its Association with Cancer Risk: A Review.Cancer Epidemiol Biomarkers Prev 2008;17:3298-3305.
11.Klaunig JE, Kamendulis LM.The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol 2004;44:239-267.
12.Karihtala P, Soini Y. Reactive oxygen species and antioxidant mechanisms in human tissues and their relation to malignancies. APMIS 2007;115:81-103.
13.Tsanou E, Ioachim E, Briasoulis E, Damala K, Charchanti1 A, Karavasilis V, Pavlidis N and Agnantis N.J. Immunohistochemical expression of superoxide dismutase (MnSOD) anti-oxidant enzyme in invasive breast carcinoma. Histol Histopathol 2004 ;19: 807-813
14.Waris G, AlamK: Immunogenicity of superoxide radical modified-DNA: studies on induced antibodies and SLE anti-DNA autoantibodies. Life Sci 2004; 75:2633-42.
15.Waris G, Huh KW, Siddiqui A: Mitochondrially associated hepatitis B virus X protein constitutively activates transcription factors STAT-3 and NF-kappa B via oxidative stress. Mol Cell Biol 2001; 21:7721-7730.
16.Waris G, Livolsi A, Imbert V, Peyron JF, Siddiqui A: Hepatitis C
virus NS5A and subgenomic replicon activate NF-kappaB via tyrosine
phosphorylation of IkappaBalpha and its degradation by calpain protease. J Biol Chem 2003; 278:40778-40787.
17.Karihtala P, Soini Y. Reactive oxygen species and antioxidant mechanisms in human tissues and their relation to malignancies. APMIS. 2007;115:81-103.
18.Betteridge DJ. 2000. What is oxidative stress? Metabolism 2000;49:3–8.
19.Abuja PM, Albertini R. Methods for monitoring oxidative stress, lipid peroxidation, and oxidation resistance of lipoproteins. Clin Chim Acta 2001; 306:1–17.
20.Clarkson PM, Thompson HS. Antioxidants: what role do they play in physical activity and health? Am J Clin Nutr 2000;72:637–646.
21.Ziegler DM. Role of reversible oxidation-reduction of enzyme thiolsdisulfides in metabolic regulation. Annu Rev Biochem1985; 54:305–329.
22.Nakamura H, Nakamura K,Yodoi J. Redox regulation of cellular activation. Annu Rev Immunol 1997; 15:351–369.
23.Activity and protein level of CuZnSOD and MnSOD in benign and malignant liver tumors. Skrzycki M , Ścibior D, Podsiad M, Czeczot H. Clinical Biochemistry 2008;41:91-96.
24.Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact 2006;160:1-40.
25.Crystal structure of nickel-containing superoxide dismutase reveals another type of active site. Wuerges J, Lee JW, Yim YI, Yim HS, Kang SO, Carugo KD. Proce Natl Acad Sci USA 2004;101:8569-8574.
26.Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the Cu,ZnSOD (sod1), Mn-SOD (sod2), and EC-SOD (sod3) gene structures, evolution, and expression. Free Radic Biol Med 2002;33:337–349.
27.Sarsour EH, Venkataraman S, Kalen AL, Oberley LW, Goswami1 PC. Manganese superoxide dismutase activity regulates transitions between quiescent and proliferative growth. Aging Cell 2008;7:405-417.
28.Bag A, Niladri Bag N. Target sequence polymorphism of human manganese superoxide dismutase gene and its association with cancer risk: a review. Cancer Epidemiol Biomarkers Prev 2008;17:3298-3305.
29.Xu Y, Fang F, Dhar SK, Bosch A, St Clair WH, Kasarskis FJ, St Clair DK.Mutations in the SOD2 Promoter Reveal a Molecular Basis for an Activating Protein 2-Dependent Dysregulation of Manganese Superoxide Dismutase Expression in Cancer Cells. Mol Cancer Res 2008;6:1881-1893.
30.Menon SG, Goswami PC. A redox cycle within the cell cycle: ring in the old with the new. Oncogene 2007;26:1101-1109.
31. Sarsour EH, Kumar MG, Chaudhuri L, Kalen AL, Goswami PC. Redox Control of the Cell Cycle in Health and Disease. Antioxid Redox Signaling 2009 Dec;11:2985-3011.
32.Havens CG, Ho A, Yoshioka N, Dowdy SF. Regulation of Late G1/S Phase Transition and APCCdh1 by Reactive Oxygen Species. Mol Cell Biol 2006;26: 4701–4711.
33.Shackelford RE, Kaufmann WK, Paules RS. Cell Cycle Control, Checkpoint Mechanisms, and Genotoxic Stress. Environ Health Perspect 1999;107 Suppl 1:5-24.
34.Okuda, Tl. PCTAIRE-1 and PCTAIRE-2: two members of a novel
cdc2/CDC28-related protein kinase gene family. Oncogene 1992;7: 2249-2258.
35.Serrano, M. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 1993; 366: 704-707.
36.Pines, J. The cell cycle kinases. Sem Cancer Biol 1994; 5: 305-313.
37.Kato, JY. Regulation of cyclin D-dependent kinase (Cdk4) by
Cdk4-activating kinase. Mol Cell Biol 1994; 14: 2713-2721.
38.Matsuoka, M. Activation of cyclin-dependent kinase 4 (Cdk4) by mouse MO15-associated kinase. Mol Cell Biol 1994; 14: 7265-7275.
39.MacLachlan, T.K. Cyclins, cyclin-dependent kinases and Cdk inhibitors: implications in cell cycle control and cancer. Crit Rev Euk Gene Expr 1995; 5: 127-156.

40.Siebert, R.. Role of the cyclin-dependent kinase 4 and 6 inhibitor gene family p15, p16, p18 and p19 in leukemia and lymphoma. Leuk. Lymphoma 1996; 23: 505-520.
41.Alao JP.The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention. Mol Cancer 2007; 2;6:24.
42.SCHMITT E, PAQUET C, BEAUCHEMIN M, BERTRAND R. DNA-damage response network at the crossroads of cell-cycle checkpoints, cellular senescence and apoptosis. J Zhejiang Univ Sci B 2007;8:377-397.
43.Macip S, Igarashi M, Fang L, Chen A, Pan ZQ, Lee SW, Aaronson SA. Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO J 2002 ;21:2180-2188.
44.Li JJ, Oberlery LW, St Clair DK, Ridnour LA, Oberley TD. Phenotypic changes induced in human breast cancer cells by overexpression of manganese-containing superoxide dismutase. Oncogen 1995;10:1989-2000.
45.Kahlos K, Soini Y, Paakko P, Saily M, Linnainmaa K, Kinnula VL. Proliferation, apoptosis, and manganese superoxide dismutase in malignant mesothelioma. Int J Cancer 2000;88:37-43.
46.Fisher CJ, Goswami PC. Mitochondria-targeted antioxidant enzyme activity regulates radioresistance in human pancreatic cancer cells. Cancer Biol Ther 2008;7:1271-1279.
47.Alexandre J, Nicco C , Chereau C, Laurent A , Weill B ,Goldwasser F, Batteux F. Improvement of the Therapeutic Index of Anticancer Drugs by the Superoxide Dismutase Mimic Mangafodipir. J Natl Cancer 2006;98:236-244.
48.Cullen JJ, Weydert C, Hinkhouse MM, Ritchie J, Domann FE, Spitz D. The role of
manganese superoxide dismutase in the growth of pancreatic adenocarcinoma. Cancer Res 2003;63:1297-1303.

49.Zhang Y, Zhao W, Zhang HJ, Domann FE, Oberley LW. Overexpression of copper zinc superoxide dismutase suppresses human glioma cell growth. Cancer Res 2002;62: 1205-1212.
50.Sarsour EH, Venkataraman S, Kalen AL, Oberley LW, Goswami PC. Manganese superoxide dismutase activity regulates transitions between quiescent and proliferative growth. Aging Cell 2008;7:405-417.
51.Nita ME, Alves VA, Carrilho FJ, Ono-Nita SK, Mello ES, Gama-Rodrigues JJ.Molecular Aspects of Hepatic Carcinogenesis. Rev Inst Med Trop Sao Paulo 2002; 44:39-48.
52.Swaminathan K, Yang Y, Grotz N, Campisi L, Jack T. An enhancer trap line associated with a D-class cyclin gene in Arabidopsis. Plant Physiology, December 2000; 124:1658-1667.
電子全文 Fulltext
本電子全文僅授權使用者為學術研究之目的,進行個人非營利性質之檢索、閱讀、列印。請遵守中華民國著作權法之相關規定,切勿任意重製、散佈、改作、轉貼、播送,以免觸法。
論文使用權限 Thesis access permission:校內立即公開,校外一年後公開 off campus withheld
開放時間 Available:
校內 Campus: 已公開 available
校外 Off-campus: 已公開 available


紙本論文 Printed copies
紙本論文的公開資訊在102學年度以後相對較為完整。如果需要查詢101學年度以前的紙本論文公開資訊,請聯繫圖資處紙本論文服務櫃台。如有不便之處敬請見諒。
開放時間 available 已公開 available

QR Code