Skip navigation

DSpace

機構典藏 DSpace 系統致力於保存各式數位資料(如:文字、圖片、PDF)並使其易於取用。

點此認識 DSpace
DSpace logo
English
中文
  • 瀏覽論文
    • 校院系所
    • 出版年
    • 作者
    • 標題
    • 關鍵字
    • 指導教授
  • 搜尋 TDR
  • 授權 Q&A
    • 我的頁面
    • 接受 E-mail 通知
    • 編輯個人資料
  1. NTU Theses and Dissertations Repository
  2. 生命科學院
  3. 生命科學系
請用此 Handle URI 來引用此文件: http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/79335
完整後設資料紀錄
DC 欄位值語言
dc.contributor.advisor吳金洌(Jen-Leih Wu)
dc.contributor.authorKeng-Yu Chiangen
dc.contributor.author江耕宇zh_TW
dc.date.accessioned2022-11-23T08:58:21Z-
dc.date.available2021-11-03
dc.date.available2022-11-23T08:58:21Z-
dc.date.copyright2021-11-03
dc.date.issued2021
dc.date.submitted2021-10-28
dc.identifier.citation1. D.H. Wasserman, Four grams of glucose. Am J Physiol Endocrinol Metab, 2009. 296(1): p. E11-21. 2. D.M. Grant, Detoxification pathways in the liver. J Inherit Metab Dis, 1991. 14(4): p. 421-30. 3. J.L. Boyer, Bile formation and secretion. Compr Physiol, 2013. 3(3): p. 1035-78. 4. K.S. Zaret, Regulatory phases of early liver development: paradigms of organogenesis. Nat Rev Genet, 2002. 3(7): p. 499-512. 5. R. Taub, Liver regeneration: from myth to mechanism. Nat Rev Mol Cell Biol, 2004. 5(10): p. 836-47. 6. N. Fausto, J.S. Campbell, and K.J. Riehle, Liver regeneration. Hepatology, 2006. 43(2 Suppl 1): p. S45-53. 7. J.W. Kung, I.S. Currie, S.J. Forbes, and J.A. Ross, Liver development, regeneration, and carcinogenesis. J Biomed Biotechnol, 2010. 2010: p. 984248. 8. H.H. Otu, K. Naxerova, K. Ho, H. Can, N. Nesbitt, T.A. Libermann, and S.J. Karp, Restoration of liver mass after injury requires proliferative and not embryonic transcriptional patterns. J Biol Chem, 2007. 282(15): p. 11197-204. 9. F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, and A. Jemal, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018. 68(6): p. 394-424. 10. C.Y. Liu, K.F. Chen, and P.J. Chen, Treatment of Liver Cancer. Cold Spring Harb Perspect Med, 2015. 5(9): p. a021535. 11. D. Schuppan and N.H. Afdhal, Liver cirrhosis. Lancet, 2008. 371(9615): p. 838-51. 12. F. Bohm, U.A. Kohler, T. Speicher, and S. Werner, Regulation of liver regeneration by growth factors and cytokines. EMBO Mol Med, 2010. 2(8): p. 294-305. 13. M. Borowiak, A.N. Garratt, T. Wustefeld, M. Strehle, C. Trautwein, and C. Birchmeier, Met provides essential signals for liver regeneration. Proc Natl Acad Sci U S A, 2004. 101(29): p. 10608-13. 14. V.M. Factor, D. Seo, T. Ishikawa, P. Kaposi-Novak, J.U. Marquardt, J.B. Andersen, E.A. Conner, and S.S. Thorgeirsson, Loss of c-Met disrupts gene expression program required for G2/M progression during liver regeneration in mice. PLoS One, 2010. 5(9). 15. S. Benvenuti and P.M. Comoglio, The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol, 2007. 213(2): p. 316-25. 16. C.G. Huh, V.M. Factor, A. Sanchez, K. Uchida, E.A. Conner, and S.S. Thorgeirsson, Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proc Natl Acad Sci U S A, 2004. 101(13): p. 4477-82. 17. D.B. Stolz, W.M. Mars, B.E. Petersen, T.H. Kim, and G.K. Michalopoulos, Growth factor signal transduction immediately after two-thirds partial hepatectomy in the rat. Cancer Res, 1999. 59(16): p. 3954-60. 18. A. Bell, Q. Chen, M.C. DeFrances, G.K. Michalopoulos, and R. Zarnegar, The five amino acid-deleted isoform of hepatocyte growth factor promotes carcinogenesis in transgenic mice. Oncogene, 1999. 18(4): p. 887-95. 19. G.K. Michalopoulos, Liver regeneration. J Cell Physiol, 2007. 213(2): p. 286-300. 20. Z. He and A. Bateman, Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med (Berl), 2003. 81(10): p. 600-12. 21. C.J. Liu and X. Bosch, Progranulin: a growth factor, a novel TNFR ligand and a drug target. Pharmacol Ther, 2012. 133(1): p. 124-32. 22. Y.H. Li, M.H. Chen, H.Y. Gong, S.Y. Hu, Y.W. Li, G.H. Lin, C.C. Lin, W. Liu, and J.L. Wu, Progranulin A-mediated MET signaling is essential for liver morphogenesis in zebrafish. J Biol Chem, 2010. 285(52): p. 41001-9. 23. G. Frampton, P. Invernizzi, F. Bernuzzi, H.Y. Pae, M. Quinn, D. Horvat, C. Galindo, L. Huang, M. McMillin, B. Cooper, L. Rimassa, and S. DeMorrow, Interleukin-6-driven progranulin expression increases cholangiocarcinoma growth by an Akt-dependent mechanism. Gut, 2012. 61(2): p. 268-77. 24. D. Alvaro, Progranulin and cholangiocarcinoma: another bad boy on the block! Gut, 2012. 61(2): p. 170-1. 25. C. Liu, J. Li, W. Shi, L. Zhang, S. Liu, Y. Lian, S. Liang, and H. Wang, Progranulin Regulates Inflammation and Tumor. Antiinflamm Antiallergy Agents Med Chem, 2020. 19(2): p. 88-102. 26. F. Liu, W. Zhang, F. Yang, T. Feng, M. Zhou, Y. Yu, X. Yu, W. Zhao, F. Yi, W. Tang, and Y. Lu, Interleukin-6-stimulated progranulin expression contributes to the malignancy of hepatocellular carcinoma cells by activating mTOR signaling. Sci Rep, 2016. 6: p. 21260. 27. A. Raya, C.M. Koth, D. Buscher, Y. Kawakami, T. Itoh, R.M. Raya, G. Sternik, H.J. Tsai, C. Rodriguez-Esteban, and J.C. Izpisua-Belmonte, Activation of Notch signaling pathway precedes heart regeneration in zebrafish. Proc Natl Acad Sci U S A, 2003. 100 Suppl 1: p. 11889-95. 28. E.J. Thatcher, I. Paydar, K.K. Anderson, and J.G. Patton, Regulation of zebrafish fin regeneration by microRNAs. Proc Natl Acad Sci U S A, 2008. 105(47): p. 18384-9. 29. A. Rojas-Munoz, S. Rajadhyksha, D. Gilmour, F. van Bebber, C. Antos, C. Rodriguez Esteban, C. Nusslein-Volhard, and J.C. Izpisua Belmonte, ErbB2 and ErbB3 regulate amputation-induced proliferation and migration during vertebrate regeneration. Dev Biol, 2009. 327(1): p. 177-90. 30. N. Kishimoto, K. Shimizu, and K. Sawamoto, Neuronal regeneration in a zebrafish model of adult brain injury. Dis Model Mech, 2012. 5(2): p. 200-9. 31. K.C. Sadler, K.N. Krahn, N.A. Gaur, and C. Ukomadu, Liver growth in the embryo and during liver regeneration in zebrafish requires the cell cycle regulator, uhrf1. Proc Natl Acad Sci U S A, 2007. 104(5): p. 1570-5. 32. P. Britz, Tilapia: Biology, Culture and Nutrition. African Journal of Aquatic Science, 2008. 33(1): p. 103. 33. W.G. Hlady and K.C. Klontz, The epidemiology of Vibrio infections in Florida, 1981-1993. J Infect Dis, 1996. 173(5): p. 1176-83. 34. W.W. Zhang, K. Sun, S. Cheng, and L. Sun, Characterization of DegQVh, a serine protease and a protective immunogen from a pathogenic Vibrio harveyi strain. Appl Environ Microbiol, 2008. 74(20): p. 6254-62. 35. L.I. Hor, T.T. Chang, and S.T. Wang, Survival of Vibrio vulnificus in whole blood from patients with chronic liver diseases: association with phagocytosis by neutrophils and serum ferritin levels. J Infect Dis, 1999. 179(1): p. 275-8. 36. D.B. Jump, N-3 polyunsaturated fatty acid regulation of hepatic gene transcription. Curr Opin Lipidol, 2008. 19(3): p. 242-7. 37. P. Lefebvre, G. Chinetti, J.C. Fruchart, and B. Staels, Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest, 2006. 116(3): p. 571-80. 38. B.P. Kota, T.H. Huang, and B.D. Roufogalis, An overview on biological mechanisms of PPARs. Pharmacol Res, 2005. 51(2): p. 85-94. 39. G.C. Burdge and P.C. Calder, Conversion of alpha-linolenic acid to longer-chain polyunsaturated fatty acids in human adults. Reprod Nutr Dev, 2005. 45(5): p. 581-97. 40. J.M. Lee, H. Lee, S. Kang, and W.J. Park, Fatty Acid Desaturases, Polyunsaturated Fatty Acid Regulation, and Biotechnological Advances. Nutrients, 2016. 8(1). 41. M. Geiger, B.S. Mohammed, S. Sankarappa, and H. Sprecher, Studies to determine if rat liver contains chain-length-specific acyl-CoA 6-desaturases. Biochim Biophys Acta, 1993. 1170(2): p. 137-42. 42. S.Y. Shin, V.K. Bajpai, H.R. Kim, and S.C. Kang, Antibacterial activity of bioconverted eicosapentaenoic (EPA) and docosahexaenoic acid (DHA) against foodborne pathogenic bacteria. Int J Food Microbiol, 2007. 113(2): p. 233-6. 43. A.P. Desbois and K.C. Lawlor, Antibacterial activity of long-chain polyunsaturated fatty acids against Propionibacterium acnes and Staphylococcus aureus. Mar Drugs, 2013. 11(11): p. 4544-57. 44. G. Schmitz and J. Ecker, The opposing effects of n-3 and n-6 fatty acids. Prog Lipid Res, 2008. 47(2): p. 147-55. 45. C.E. Loscher, E. Draper, O. Leavy, D. Kelleher, K.H. Mills, and H.M. Roche, Conjugated linoleic acid suppresses NF-kappa B activation and IL-12 production in dendritic cells through ERK-mediated IL-10 induction. J Immunol, 2005. 175(8): p. 4990-8. 46. H. Li, X.Z. Ruan, S.H. Powis, R. Fernando, W.Y. Mon, D.C. Wheeler, J.F. Moorhead, and Z. Varghese, EPA and DHA reduce LPS-induced inflammation responses in HK-2 cells: evidence for a PPAR-gamma-dependent mechanism. Kidney Int, 2005. 67(3): p. 867-74. 47. L. Martinez-Rubio, S. Morais, O. Evensen, S. Wadsworth, K. Ruohonen, J.L. Vecino, J.G. Bell, and D.R. Tocher, Functional feeds reduce heart inflammation and pathology in Atlantic Salmon (Salmo salar L.) following experimental challenge with Atlantic salmon reovirus (ASRV). PLoS One, 2012. 7(11): p. e40266. 48. B.S. Noriega, M.A. Sanchez-Gonzalez, D. Salyakina, and J. Coffman, Understanding the Impact of Omega-3 Rich Diet on the Gut Microbiota. Case Rep Med, 2016. 2016: p. 3089303. 49. H.N. Yu, J. Zhu, W.S. Pan, S.R. Shen, W.G. Shan, and U.N. Das, Effects of fish oil with a high content of n-3 polyunsaturated fatty acids on mouse gut microbiota. Arch Med Res, 2014. 45(3): p. 195-202. 50. S.E. Power, P.W. O'Toole, C. Stanton, R.P. Ross, and G.F. Fitzgerald, Intestinal microbiota, diet and health. Br J Nutr, 2014. 111(3): p. 387-402. 51. H. Watson, S. Mitra, F.C. Croden, M. Taylor, H.M. Wood, S.L. Perry, J.A. Spencer, P. Quirke, G.J. Toogood, C.L. Lawton, L. Dye, P.M. Loadman, and M.A. Hull, A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut, 2018. 67(11): p. 1974-1983. 52. A.D. Andersen, L. Molbak, K.F. Michaelsen, and L. Lauritzen, Molecular fingerprints of the human fecal microbiota from 9 to 18 months old and the effect of fish oil supplementation. J Pediatr Gastroenterol Nutr, 2011. 53(3): p. 303-9. 53. C. Bidu, Q. Escoula, S. Bellenger, A. Spor, M. Galan, A. Geissler, A. Bouchot, D. Dardevet, B. Morio, P.D. Cani, L. Lagrost, M. Narce, and J. Bellenger, The Transplantation of omega3 PUFA-Altered Gut Microbiota of fat-1 Mice to Wild-Type Littermates Prevents Obesity and Associated Metabolic Disorders. Diabetes, 2018. 67(8): p. 1512-1523. 54. C. Strobel, G. Jahreis, and K. Kuhnt, Survey of n-3 and n-6 polyunsaturated fatty acids in fish and fish products. Lipids Health Dis, 2012. 11: p. 144. 55. C.L. Cheng, S.J. Huang, C.L. Wu, H.Y. Gong, C.F. Ken, S.Y. Hu, and J.L. Wu, Transgenic expression of omega-3 PUFA synthesis genes improves zebrafish survival during Vibrio vulnificus infection. J Biomed Sci, 2015. 22: p. 103. 56. Y.D. Wang, K.C. Peng, J.L. Wu, and J.Y. Chen, Transgenic expression of salmon delta-5 and delta-6 desaturase in zebrafish muscle inhibits the growth of Vibrio alginolyticus and affects fish immunomodulatory activity. Fish Shellfish Immunol, 2014. 39(2): p. 223-30. 57. C.B. Kimmel, W.W. Ballard, S.R. Kimmel, B. Ullmann, and T.F. Schilling, Stages of embryonic development of the zebrafish. Dev Dyn, 1995. 203(3): p. 253-310. 58. W. Liu, J.R. Chen, C.H. Hsu, Y.H. Li, Y.M. Chen, C.Y. Lin, S.J. Huang, Z.K. Chang, Y.C. Chen, C.H. Lin, H.Y. Gong, C.C. Lin, K. Kawakami, and J.L. Wu, A zebrafish model of intrahepatic cholangiocarcinoma by dual expression of hepatitis B virus X and hepatitis C virus core protein in liver. Hepatology, 2012. 56(6): p. 2268-76. 59. C.-M.L. Thomas T. Chen, Maria J. Chen, Jay H. Lo, Pinwen P. Chiou, Hong-Yi Gong, Jen-Leih Wu, Mark H.-C. Chen, Charles Yarish, Chapter 13. Transgenic Technology in Marine Organisms. 2015, Springer Handbook of Marine Biotechnology. 60. M.D. Kinkel, S.C. Eames, L.H. Philipson, and V.E. Prince, Intraperitoneal injection into adult zebrafish. J Vis Exp, 2010(42). 61. M.G. Ormerod, Flow cytometric analysis of cells using an antibody to a surface antigen. Methods Mol Biol, 1992. 80: p. 373-9. 62. J. Folch, M. Lees, and G.H. Sloane Stanley, A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem, 1957. 226(1): p. 497-509. 63. E.O. Abu and I. Oluwatowoju, Omega-3 index determined by gas chromatography with electron impact mass spectrometry. Prostaglandins Leukot Essent Fatty Acids, 2009. 80(4): p. 189-94. 64. M.L. Hart, A. Meyer, P.J. Johnson, and A.C. Ericsson, Comparative Evaluation of DNA Extraction Methods from Feces of Multiple Host Species for Downstream Next-Generation Sequencing. PLoS One, 2015. 10(11): p. e0143334. 65. Y.T. Chen, N.S. Yang, Y.C. Lin, S.T. Ho, K.Y. Li, J.S. Lin, J.R. Liu, and M.J. Chen, A combination of Lactobacillus mali APS1 and dieting improved the efficacy of obesity treatment via manipulating gut microbiome in mice. Sci Rep, 2018. 8(1): p. 6153. 66. G.H. Fu, Z.Y. Wan, J.H. Xia, F. Liu, X.J. Liu, and G.H. Yue, The MCP-8 gene and its possible association with resistance to Streptococcus agalactiae in tilapia. Fish Shellfish Immunol, 2014. 40(1): p. 331-6. 67. V. Dhople, A. Krukemeyer, and A. Ramamoorthy, The human beta-defensin-3, an antibacterial peptide with multiple biological functions. Biochim Biophys Acta, 2006. 1758(9): p. 1499-512. 68. J.J. Dong, F. Wu, X. Ye, C.F. Sun, Y.Y. Tian, M.X. Lu, R. Zhang, and Z.H. Chen, Beta-defensin in Nile tilapia (Oreochromis niloticus): Sequence, tissue expression, and anti-bacterial activity of synthetic peptides. Gene, 2015. 566(1): p. 23-31. 69. J.M. Larsen, The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology, 2017. 151(4): p. 363-374. 70. Y. Kuse, K. Tsuruma, S. Sugitani, H. Izawa, Y. Ohno, M. Shimazawa, and H. Hara, Progranulin promotes the retinal precursor cell proliferation and the photoreceptor differentiation in the mouse retina. Sci Rep, 2016. 6: p. 23811. 71. M. Daya, W. Loilome, A. Techasen, M. Thanee, P. Sa-Ngiamwibool, A. Titapun, P. Yongvanit, and N. Namwat, Progranulin modulates cholangiocarcinoma cell proliferation, apoptosis, and motility via the PI3K/pAkt pathway. Oncotargets and Therapy, 2018. 11: p. 395-408. 72. S.A. Khan, B.R. Davidson, R.D. Goldin, N. Heaton, J. Karani, S.P. Pereira, W.M.C. Rosenberg, P. Tait, S.D. Taylor-Robinson, A.V. Thillainayagam, H.C. Thomas, and H. Wasan, Guidelines for the diagnosis and treatment of cholangiocarcinoma: an update. Gut, 2012. 61(12): p. 1657-1669. 73. C.D. Anderson, C.W. Pinson, J. Berlin, and R.S. Chari, Diagnosis and treatment of cholangiocarcinoma. Oncologist, 2004. 9(1): p. 43-57. 74. S. Kim, U.P. Radhakrishnan, S.K. Rajpurohit, V. Kulkarni, and P. Jagadeeswaran, Vivo-Morpholino knockdown of alphaIIb: A novel approach to inhibit thrombocyte function in adult zebrafish. Blood Cells Mol Dis, 2010. 44(3): p. 169-74. 75. M.E. Robu, J.D. Larson, A. Nasevicius, S. Beiraghi, C. Brenner, S.A. Farber, and S.C. Ekker, p53 activation by knockdown technologies. Plos Genetics, 2007. 3(5): p. 787-801. 76. S.Y. Hu, C.C. Tai, Y.H. Li, and J.L. Wu, Progranulin compensates for blocked IGF-1 signaling to promote myotube hypertrophy in C2C12 myoblasts via the PI3K/Akt/mTOR pathway. FEBS Lett, 2012. 586(19): p. 3485-92. 77. T.A. Beyer, W. Xu, D. Teupser, U.A.D. Keller, P. Bugnon, E. Hildt, J. Thiery, Y.W. Kan, and S. Werner, Impaired liver regeneration in Nrf2 knockout mice: role of ROS-mediated insulin/IGF-1 resistance. Embo Journal, 2008. 27(1): p. 212-223. 78. C. Schmidt, F. Bladt, S. Goedecke, V. Brinkmann, W. Zschiesche, M. Sharpe, E. Gherardi, and C. Birchmeier, Scatter Factor/Hepatocyte Growth-Factor Is Essential for Liver Development. Nature, 1995. 373(6516): p. 699-702. 79. R.M. Anderson, M. Delous, J.A. Bosch, L. Ye, M.A. Robertson, D. Hesselson, and D.Y. Stainier, Hepatocyte growth factor signaling in intrapancreatic ductal cells drives pancreatic morphogenesis. PLoS Genet, 2013. 9(7): p. e1003650. 80. W.E. Kim, B. Yue, and G. Serrero, Signaling Pathway of GP88 (Progranulin) in Breast Cancer Cells: Upregulation and Phosphorylation of c-myc by GP88/Progranulin in Her2-Overexpressing Breast Cancer Cells. Breast Cancer (Auckl), 2015. 9(Suppl 2): p. 71-7. 81. D.W. Seol, Q. Chen, and R. Zarnegar, Transcriptional activation of the hepatocyte growth factor receptor (c-met) gene by its ligand (hepatocyte growth factor) is mediated through AP-1. Oncogene, 2000. 19(9): p. 1132-7. 82. J. Garnett, V. Chumbalkar, B. Vaillant, A.E. Gururaj, K.S. Hill, K. Latha, J. Yao, W. Priebe, H. Colman, L.A. Elferink, and O. Bogler, Regulation of HGF Expression by ΔEGFR-Mediated c-Met Activation in Glioblastoma Cells. Neoplasia, 2013. 15(1): p. 73-IN21. 83. Y.H. Li, H.Y. Chen, Y.W. Li, S.Y. Wu, Wangta-Liu, G.H. Lin, S.Y. Hu, Z.K. Chang, H.Y. Gong, C.H. Liao, K.Y. Chiang, C.W. Huang, and J.L. Wu, Progranulin regulates zebrafish muscle growth and regeneration through maintaining the pool of myogenic progenitor cells. Scientific Reports, 2013. 3. 84. J. Sun, W. Fang, B. Ke, D. He, Y. Liang, D. Ning, H. Tan, H. Peng, Y. Wang, Y. Ma, C. Ke, and X. Deng, Inapparent Streptococcus agalactiae infection in adult/commercial tilapia. Sci Rep, 2016. 6: p. 26319. 85. K.N. Frayn, P. Arner, and H. Yki-Jarvinen, Fatty acid metabolism in adipose tissue, muscle and liver in health and disease. Essays Biochem, 2006. 42: p. 89-103. 86. J.A. Emery, K. Hermon, N.K. Hamid, J.A. Donald, and G.M. Turchini, Delta-6 Desaturase substrate competition: dietary linoleic acid (18:2n-6) has only trivial effects on alpha-linolenic acid (18:3n-3) bioconversion in the teleost rainbow trout. PLoS One, 2013. 8(2): p. e57463. 87. B. Lands, Consequences of essential fatty acids. Nutrients, 2012. 4(9): p. 1338-57. 88. H.Q. Liu, Y. Qiu, Y. Mu, X.J. Zhang, L. Liu, X.H. Hou, L. Zhang, X.N. Xu, A.L. Ji, R. Cao, R.H. Yang, and F. Wang, A high ratio of dietary n-3/n-6 polyunsaturated fatty acids improves obesity-linked inflammation and insulin resistance through suppressing activation of TLR4 in SD rats. Nutr Res, 2013. 33(10): p. 849-58. 89. J.K. Innes and P.C. Calder, Omega-6 fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids, 2018. 132: p. 41-48. 90. J.S. Bae, A.R. Oh, and J.Y. Cha, Regulation of Cholesterol Metabolism in Liver: Link to NAFLD and Impact of n-3 PUFAs. J Lifestyle Med, 2013. 3(1): p. 19-25. 91. S. Bilal, O. Haworth, L. Wu, K.H. Weylandt, B.D. Levy, and J.X. Kang, Fat-1 transgenic mice with elevated omega-3 fatty acids are protected from allergic airway responses. Biochim Biophys Acta, 2011. 1812(9): p. 1164-9. 92. D. Mil-Homens, N. Bernardes, and A.M. Fialho, The antibacterial properties of docosahexaenoic omega-3 fatty acid against the cystic fibrosis multiresistant pathogen Burkholderia cenocepacia. FEMS Microbiol Lett, 2012. 328(1): p. 61-9. 93. P. Solanki, A. Aminoshariae, G. Jin, T.A. Montagnese, and A. Mickel, The effect of docosahexaenoic acid (DHA) on expression of IL-1ss, IL-6, IL-8, and TNF-alpha in normal and lipopolysaccharide (LPS)-stimulated macrophages. Quintessence Int, 2013. 44(6): p. 393. 94. Y. Williams-Bey, C. Boularan, A. Vural, N.N. Huang, I.Y. Hwang, C. Shan-Shi, and J.H. Kehrl, Omega-3 free fatty acids suppress macrophage inflammasome activation by inhibiting NF-kappaB activation and enhancing autophagy. PLoS One, 2014. 9(6): p. e97957. 95. K.D. Kohl, J. Amaya, C.A. Passement, M.D. Dearing, and M.D. McCue, Unique and shared responses of the gut microbiota to prolonged fasting: a comparative study across five classes of vertebrate hosts. FEMS Microbiol Ecol, 2014. 90(3): p. 883-94. 96. B.T. Standen, A. Rodiles, D.L. Peggs, S.J. Davies, G.A. Santos, and D.L. Merrifield, Modulation of the intestinal microbiota and morphology of tilapia, Oreochromis niloticus, following the application of a multi-species probiotic. Appl Microbiol Biotechnol, 2015. 99(20): p. 8403-17. 97. C. Giatsis, D. Sipkema, H. Smidt, J. Verreth, and M. Verdegem, The colonization dynamics of the gut microbiota in tilapia larvae. PLoS One, 2014. 9(7): p. e103641. 98. C. Menni, J. Zierer, T. Pallister, M.A. Jackson, T. Long, R.P. Mohney, C.J. Steves, T.D. Spector, and A.M. Valdes, Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women. Sci Rep, 2017. 7(1): p. 11079. 99. A.M. Tarnecki, F.A. Burgos, C.L. Ray, and C.R. Arias, Fish intestinal microbiome: diversity and symbiosis unravelled by metagenomics. J Appl Microbiol, 2017. 123(1): p. 2-17. 100. C.S. Chung, P.F. Chang, C.H. Liao, T.H. Lee, Y. Chen, Y.C. Lee, M.S. Wu, H.P. Wang, and Y.H. Ni, Differences of microbiota in small bowel and faeces between irritable bowel syndrome patients and healthy subjects. Scand J Gastroenterol, 2016. 51(4): p. 410-9. 101. G.P. Schincaglia, B.Y. Hong, A. Rosania, J. Barasz, A. Thompson, T. Sobue, F. Panagakos, J.A. Burleson, A. Dongari-Bagtzoglou, and P.I. Diaz, Clinical, Immune, and Microbiome Traits of Gingivitis and Peri-implant Mucositis. J Dent Res, 2017. 96(1): p. 47-55. 102. S. Ji, Y. Kim, B.M. Min, S.H. Han, and Y. Choi, Innate immune responses of gingival epithelial cells to nonperiodontopathic and periodontopathic bacteria. J Periodontal Res, 2007. 42(6): p. 503-10. 103. M. Tabbaa, M. Golubic, M.F. Roizen, and A.M. Bernstein, Docosahexaenoic acid, inflammation, and bacterial dysbiosis in relation to periodontal disease, inflammatory bowel disease, and the metabolic syndrome. Nutrients, 2013. 5(8): p. 3299-310.
dc.identifier.urihttp://tdr.lib.ntu.edu.tw/jspui/handle/123456789/79335-
dc.description.abstract肝臟是人體必需的最大器官。當肝臟受到病原體、毒素、自身免疫性疾病、部分肝切除術或肝癌的損害時,它具有快速有效的修復能力。除了肝臟再生,抗發炎過程對於肝臟修復也很重要。在本論文中,我們分別利用斑馬魚及吳郭魚動物模式探討肝臟再生和免疫反應之機制。論文的前半部講述我們利用肝臟專一性表達螢光基因斑馬魚建立部分肝切除再生模式。我們發現在部分肝切除後,腹腔注射活體型反義寡核苷酸會抑制顆粒蛋白前體A型表現並延遲肝再生。此外,轉錄體學分析和即時聚合酶鏈鎖反應結果顯示比較對照組後抑制顆粒蛋白前體A型在部分肝切除三十六小時後可能藉由抑制肝細胞生長因子訊息傳遞路徑使細胞週期和細胞增生不活化。此外,肝臟專一性大量表現顆粒蛋白前體A型轉基因斑馬魚在部分肝切除後會促進細胞增生。因此,我們認為顆粒蛋白前體A型可以正向調節肝細胞生長因子訊息傳遞路徑以促進肝臟再生機制中的肝臟細胞增生。論文的後半部講述我們建立了具有單一或雙重表現鮭魚delta-5脂肪酸去飽和酶和delta-6脂肪酸去飽和酶之轉基因吳郭魚,接著利用創傷弧菌進行細菌感染。以亞麻籽油配方餵食的肝臟和肌肉專一性單一或雙重表現delta-5脂肪酸去飽和酶和delta-6脂肪酸去飽和酶轉基因吳郭魚均顯示N-3多元不飽和脂肪酸含量更高。其中,N-3多元不飽和脂肪酸含量高的雙重表達轉基因吳郭魚在創傷弧菌感染後存活率提高及抑制發炎和免疫相關基因表現。腸道微生物組分析進一步顯示雙重表達轉基因吳郭魚具有較高的腸道微生物群多樣性,而發炎相關微生物群中的普雷沃氏菌科較低。因此,我們的研究結果顯示雙重表現鮭魚delta-5脂肪酸去飽和酶和delta-6脂肪酸去飽和酶轉基因吳郭魚增強抗病力可能與N-3多元不飽和脂肪酸含量增加和腸道微生物群組成改變有關。總結上述,本論文提出了抗發炎生長因子顆粒蛋白前體及N-3多元不飽和脂肪酸分別對於當肝臟受損時促進肝細胞增殖和提高對病原體的抗發炎作用的有正向調節之作用。zh_TW
dc.description.provenanceMade available in DSpace on 2022-11-23T08:58:21Z (GMT). No. of bitstreams: 1
U0001-2710202102574500.pdf: 36456531 bytes, checksum: 4ad0270779b68ff56ba9309090609919 (MD5)
Previous issue date: 2021
en
dc.description.tableofcontents論文口試委員審定書 I 謝辭 II 摘要 III Abstract V Table of Contents VII List of Figures XI List of Tables XIII 1. Introduction 1 1.1 Liver 1 1.2 Liver embryonic development and regeneration 1 1.3 Liver regeneration 2 1.4 Pending issue of liver regeneration 2 1.5 HGF/c-Met signaling in liver regeneration 3 1.6 Progranulin in hepatic growth 4 1.7 The elusive regulatory role of progranulin in liver regeneration 4 1.8 Liver damage caused by pathogens infection 5 1.9 Biosynthesis of omega-3 polyunsaturated fatty acids 6 1.10 N-3 PUFAs and immune responses 7 1. 11 N-3 PUFAs and gut microbiota 7 1. 12 Prospection of n-3 PUFAs enriched tilapia against infection by pathogens 8 2. Materials and Methods 10 2.1 Fish 10 2.2 Establishment of transgenic fish 11 2.3 Morpholino design and Intraperitoneal injection 12 2.4 Partial Hepatectomy 13 2.5 RNA Sequencing and Analysis 13 2.6 Cell cycle analysis by flow cytometry 14 2.7 Quantitative real-time PCR 15 2.8 Western blots and Immunohistochemistry 16 2.9 Imaging 17 2.10 Fatty acid extraction and analysis 17 2.11 Vibrio vulnificus culture 18 2.12 Vibrio vulnificus challenge 18 2.13 DNA extraction and sequencing of gut microbiota 19 2.14 Statistical analysis 20 3. Results 21 3.1 Section I: The regulatory role of progranulin involved in liver regeneration after partial hepatectomy 21 3.1.1 Zebrafish progranulin A is involved in liver regeneration of hepatectomized zebrafish 21 3.1.2 Knockdown of GrnA delays liver regeneration after partial hepatectomy 22 3.1.3 Transcriptome analysis 24 3.1.4 GrnA morphants exhibit impaired liver regeneration with suppressed cell cycle and cell proliferation 26 3.1.5 Knockdown of GrnA prevents signaling through the HGF/c-met axis after partial hepatectomy 28 3.1.6 Liver-specific expression of GrnA in transgenic zebrafish promotes hepatocyte proliferation after partial hepatectomy 29 3.2 Section II: Dual expression of transgenic delta-5 and delta-6 desaturase in tilapia enhances resistance to Vibrio vulnificus infection 31 3.2.1 Tissue-specific expression of desaturases in tilapia 31 3.2.2 Fatty acid compositions of basal diet and supplemented linseed oil 33 3.2.3 LO basal diet increased the DHA content in dual-transgenic tilapia 33 3.2.4 Dual-transgenic tilapia had improved survival after Vibrio vulnificus challenge 34 3.2.5 Inflammatory gene expression after Vibrio vulnificus infection was attenuated in transgenic tilapia 36 3.2.6 High bacterial diversity and decrease of inflammation-associated gut microbiota in transgenic tilapia 38 4. Discussion 40 4.1 Section I: The regulatory role of progranulin involved in liver regeneration after partial hepatectomy 40 4.2 Section II: Dual expression of transgenic delta-5 and delta-6 desaturase in tilapia enhances resistance to Vibrio vulnificus infection 45 References 50 Figures 68 Tables 96 Appendix 98
dc.language.isoen
dc.title魚類肝臟的再生及免疫反應之分子調控機制zh_TW
dc.titleFish Liver Molecular Functions Involved in Regeneration and Immune Responsesen
dc.date.schoolyear109-2
dc.description.degree博士
dc.contributor.oralexamcommittee洪健睿(Hsin-Tsai Liu),陳志毅(Chih-Yang Tseng),龔紘毅,呂明偉,耿全福
dc.subject.keyword顆粒蛋白前體,部分肝切除手術,肝細胞生長因子,肝臟再生,脂肪酸去飽和酶,多元不飽和脂肪酸,普雷沃氏菌科,zh_TW
dc.subject.keywordProgranulin,Partial hepatectomy,Hepatocyte growth factor,Liver regeneration,Desaturase,Omega-3 polyunsaturated fatty acids,Prevotellaceae,en
dc.relation.page134
dc.identifier.doi10.6342/NTU202104295
dc.rights.note同意授權(全球公開)
dc.date.accepted2021-10-28
dc.contributor.author-college生命科學院zh_TW
dc.contributor.author-dept生命科學系zh_TW
顯示於系所單位:生命科學系

文件中的檔案:
檔案 大小格式 
U0001-2710202102574500.pdf35.6 MBAdobe PDF檢視/開啟
顯示文件簡單紀錄


系統中的文件,除了特別指名其著作權條款之外,均受到著作權保護,並且保留所有的權利。

社群連結
聯絡資訊
10617臺北市大安區羅斯福路四段1號
No.1 Sec.4, Roosevelt Rd., Taipei, Taiwan, R.O.C. 106
Tel: (02)33662353
Email: ntuetds@ntu.edu.tw
意見箱
相關連結
館藏目錄
國內圖書館整合查詢 MetaCat
臺大學術典藏 NTU Scholars
臺大圖書館數位典藏館
本站聲明
© NTU Library All Rights Reserved