請用此 Handle URI 來引用此文件:
http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/66009
完整後設資料紀錄
DC 欄位 | 值 | 語言 |
---|---|---|
dc.contributor.advisor | 陶秘華(Mi-Hua Tao) | |
dc.contributor.author | Tzu-wei Hsu | en |
dc.contributor.author | 徐子崴 | zh_TW |
dc.date.accessioned | 2021-06-17T00:18:50Z | - |
dc.date.available | 2013-09-18 | |
dc.date.copyright | 2012-09-18 | |
dc.date.issued | 2012 | |
dc.date.submitted | 2012-06-28 | |
dc.identifier.citation | 1. Kaplan, M. M. 1996. Primary biliary cirrhosis. N Engl J Med 335: 1570-1580.
2. Kaplan, M. M., and M. E. Gershwin. 2005. Primary biliary cirrhosis. N Engl J Med 353: 1261-1273. 3. Rieger, R., and M. E. Gershwin. 2007. The X and why of xenobiotics in primary biliary cirrhosis. J Autoimmun 28: 76-84. 4. Portmann, B., H. Popper, J. Neuberger, and R. Williams. 1985. Sequential and diagnostic features in primary biliary cirrhosis based on serial histologic study in 209 patients. Gastroenterology 88: 1777-1790. 5. Ishibashi, H., S. Shimoda, and M. E. Gershwin. 2005. The immune response to mitochondrial autoantigens. Semin Liver Dis 25: 337-346. 6. Kita, H., Z. X. Lian, J. Van de Water, X. S. He, S. Matsumura, M. Kaplan, V. Luketic, R. L. Coppel, A. A. Ansari, and M. E. Gershwin. 2002. Identification of HLA-A2-restricted CD8(+) cytotoxic T cell responses in primary biliary cirrhosis: T cell activation is augmented by immune complexes cross-presented by dendritic cells. J Exp Med 195: 113-123. 7. Harada, K., J. Van de Water, P. S. Leung, R. L. Coppel, A. Ansari, Y. Nakanuma, and M. E. Gershwin. 1997. In situ nucleic acid hybridization of cytokines in primary biliary cirrhosis: predominance of the Th1 subset. Hepatology 25: 791-796. 8. Leuschner, M., C. F. Dietrich, T. You, C. Seidl, J. Raedle, G. Herrmann, H. Ackermann, and U. Leuschner. 2000. Characterisation of patients with primary biliary cirrhosis responding to long term ursodeoxycholic acid treatment. Gut 46: 121-126. 9. Chuang, Y. H., W. M. Ridgway, Y. Ueno, and M. E. Gershwin. 2008. Animal models of primary biliary cirrhosis. Clin Liver Dis 12: 333-347; ix. 10. Irie, J., Y. Wu, L. S. Wicker, D. Rainbow, M. A. Nalesnik, R. Hirsch, L. B. Peterson, P. S. Leung, C. Cheng, I. R. Mackay, M. E. Gershwin, and W. M. Ridgway. 2006. NOD.c3c4 congenic mice develop autoimmune biliary disease that serologically and pathogenetically models human primary biliary cirrhosis. J Exp Med 203: 1209-1219. 11. Oertelt, S., Z. X. Lian, C. M. Cheng, Y. H. Chuang, K. A. Padgett, X. S. He, W. M. Ridgway, A. A. Ansari, R. L. Coppel, M. O. Li, R. A. Flavell, M. Kronenberg, I. R. Mackay, and M. E. Gershwin. 2006. Anti-mitochondrial antibodies and primary biliary cirrhosis in TGF-beta receptor II dominant-negative mice. J Immunol 177: 1655-1660. 12. Kaplan, M. M. 2004. Novosphingobium aromaticivorans: a potential initiator of primary biliary cirrhosis. Am J Gastroenterol 99: 2147-2149. 13. Mattner, J., P. B. Savage, P. Leung, S. S. Oertelt, V. Wang, O. Trivedi, S. T. Scanlon, K. Pendem, L. Teyton, J. Hart, W. M. Ridgway, L. S. Wicker, M. E. Gershwin, and A. Bendelac. 2008. Liver autoimmunity triggered by microbial activation of natural killer T cells. Cell Host Microbe 3: 304-315. 14. Wakabayashi, K., Z. X. Lian, P. S. Leung, Y. Moritoki, K. Tsuneyama, M. J. Kurth, K. S. Lam, K. Yoshida, G. X. Yang, T. Hibi, A. A. Ansari, W. M. Ridgway, R. L. Coppel, I. R. Mackay, and M. E. Gershwin. 2008. Loss of tolerance in C57BL/6 mice to the autoantigen E2 subunit of pyruvate dehydrogenase by a xenobiotic with ensuing biliary ductular disease. Hepatology 48: 531-540. 15. Ide, T., M. Sata, H. Suzuki, Y. Uchimura, S. Murashima, M. Shirachi, and K. Tanikawa. 1996. An experimental animal model of primary biliary cirrhosis induced by lipopolysaccharide and pyruvate dehydrogenase. Kurume med J 43: 185-188. 16. Jones, D. E., J. M. Palmer, S. J. Yeaman, J. A. Kirby, and M. F. Bassendine. 1999. Breakdown of tolerance to pyruvate dehydrogenase complex in experimental autoimmune cholangitis: a mouse model of primary biliary cirrhosis. Hepatology 30: 65-70. 17. Sasaki, M., J. Allina, J. A. Odin, S. N. Thung, R. Coppel, Y. Nakanuma, and M. E. Gershwin. 2002. Autoimmune cholangitis in the SJL/J mouse is antigen non-specific. Developmental immunology 9: 103-111. 18. Bowen, D. G., M. Zen, L. Holz, T. Davis, G. W. McCaughan, and P. Bertolino. 2004. The site of primary T cell activation is a determinant of the balance between intrahepatic tolerance and immunity. J Clin Invest 114: 701-712. 19. Crispe, I. N. 2003. Hepatic T cells and liver tolerance. Nat Rev Immunol 3: 51-62. 20. Crispe, I. N. 2009. The liver as a lymphoid organ. Annu Rev Immunol 27: 147-163. 21. Thomson, A. W., and P. A. Knolle. 2010. Antigen-presenting cell function in the tolerogenic liver environment. Nat Rev Immunol 10: 753-766. 22. Crispe, I. N. 2011. Liver antigen-presenting cells. J Hepatol 54: 357-365. 23. Bamboat, Z. M., J. A. Stableford, G. Plitas, B. M. Burt, H. M. Nguyen, A. P. Welles, M. Gonen, J. W. Young, and R. P. DeMatteo. 2009. Human liver dendritic cells promote T cell hyporesponsiveness. J Immunol 182: 1901-1911. 24. Xia, S., Z. Guo, X. Xu, H. Yi, Q. Wang, and X. Cao. 2008. Hepatic microenvironment programs hematopoietic progenitor differentiation into regulatory dendritic cells, maintaining liver tolerance. Blood 112: 3175-3185. 25. Goubier, A., B. Dubois, H. Gheit, G. Joubert, F. Villard-Truc, C. Asselin-Paturel, G. Trinchieri, and D. Kaiserlian. 2008. Plasmacytoid dendritic cells mediate oral tolerance. Immunity 29: 464-475. 26. Dubois, B., G. Joubert, M. Gomez de Aguero, M. Gouanvic, A. Goubier, and D. Kaiserlian. 2009. Sequential role of plasmacytoid dendritic cells and regulatory T cells in oral tolerance. Gastroenterology 137: 1019-1028. 27. Bamboat, Z. M., L. M. Ocuin, V. P. Balachandran, H. Obaid, G. Plitas, and R. P. DeMatteo. 2010. Conventional DCs reduce liver ischemia/reperfusion injury in mice via IL-10 secretion. J Clin Invest 120: 559-569. 28. Limmer, A., J. Ohl, C. Kurts, H. G. Ljunggren, Y. Reiss, M. Groettrup, F. Momburg, B. Arnold, and P. A. Knolle. 2000. Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med 6: 1348-1354. 29. Tang, L., J. Yang, W. Liu, X. Tang, J. Chen, D. Zhao, M. Wang, F. Xu, Y. Lu, B. Liu, Q. Sun, L. Zhang, and F. He. 2009. Liver sinusoidal endothelial cell lectin, LSECtin, negatively regulates hepatic T-cell immune response. Gastroenterology 137: 1498-1508 e1491-1495. 30. Dong, H., G. Zhu, K. Tamada, D. B. Flies, J. M. van Deursen, and L. Chen. 2004. B7-H1 determines accumulation and deletion of intrahepatic CD8(+) T lymphocytes. Immunity 20: 327-336. 31. You, Q., L. Cheng, R. M. Kedl, and C. Ju. 2008. Mechanism of T cell tolerance induction by murine hepatic Kupffer cells. Hepatology 48: 978-990. 32. Knolle, P. A., A. Uhrig, U. Protzer, M. Trippler, R. Duchmann, K. H. Meyer zum Buschenfelde, and G. Gerken. 1998. Interleukin-10 expression is autoregulated at the transcriptional level in human and murine Kupffer cells. Hepatology 27: 93-99. 33. Ellett, J. D., C. Atkinson, Z. P. Evans, Z. Amani, E. Balish, M. G. Schmidt, N. van Rooijen, R. G. Schnellmann, and K. D. Chavin. 2010. Murine Kupffer cells are protective in total hepatic ischemia/reperfusion injury with bowel congestion through IL-10. J Immunol 184: 5849-5858. 34. Wahl, C., P. Bochtler, L. Chen, R. Schirmbeck, and J. Reimann. 2008. B7-H1 on hepatocytes facilitates priming of specific CD8 T cells but limits the specific recall of primed responses. Gastroenterology 135: 980-988. 35. Bertolino, P., M. C. Trescol-Biemont, and C. Rabourdin-Combe. 1998. Hepatocytes induce functional activation of naive CD8+ T lymphocytes but fail to promote survival. Eur J Immunol 28: 221-236. 36. Winau, F., G. Hegasy, R. Weiskirchen, S. Weber, C. Cassan, P. A. Sieling, R. L. Modlin, R. S. Liblau, A. M. Gressner, and S. H. Kaufmann. 2007. Ito cells are liver-resident antigen-presenting cells for activating T cell responses. Immunity 26: 117-129. 37. Yu, M. C., C. H. Chen, X. Liang, L. Wang, C. R. Gandhi, J. J. Fung, L. Lu, and S. Qian. 2004. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology 40: 1312-1321. 38. Yang, H. R., C. C. Hsieh, L. Wang, J. J. Fung, L. Lu, and S. Qian. 2010. A critical role of TRAIL expressed on cotransplanted hepatic stellate cells in prevention of islet allograft rejection. Microsurgery 30: 332-337. 39. Jiang, G., H. R. Yang, L. Wang, G. M. Wildey, J. Fung, S. Qian, and L. Lu. 2008. Hepatic stellate cells preferentially expand allogeneic CD4+ CD25+ FoxP3+ regulatory T cells in an IL-2-dependent manner. Transplantation 86: 1492-1502. 40. Cerullo, V., W. McCormack, M. Seiler, V. Mane, R. Cela, C. Clarke, J. R. Rodgers, and B. Lee. 2007. Antigen-specific tolerance of human alpha1-antitrypsin induced by helper-dependent adenovirus. Hum Gene Ther 18: 1215-1224. 41. Breous, E., S. Somanathan, L. H. Vandenberghe, and J. M. Wilson. 2009. Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 50: 612-621. 42. Martino, A. T., S. Nayak, B. E. Hoffman, M. Cooper, G. Liao, D. M. Markusic, B. J. Byrne, C. Terhorst, and R. W. Herzog. 2009. Tolerance induction to cytoplasmic beta-galactosidase by hepatic AAV gene transfer: implications for antigen presentation and immunotoxicity. PLoS One 4: e6376. 43. Franco, L. M., B. Sun, X. Yang, A. Bird, H. Zhang, A. Schneider, T. Brown, S. P. Young, T. M. Clay, A. Amalfitano, Y. T. Chen, and D. D. Koeberl. 2005. Evasion of immune responses to introduced human acid alpha-glucosidase by liver-restricted expression in glycogen storage disease type II. Mol Ther 12: 876-884. 44. Ziegler, R. J., S. M. Lonning, D. Armentano, C. Li, D. W. Souza, M. Cherry, C. Ford, C. M. Barbon, R. J. Desnick, G. Gao, J. M. Wilson, R. Peluso, S. Godwin, B. J. Carter, R. J. Gregory, S. C. Wadsworth, and S. H. Cheng. 2004. AAV2 vector harboring a liver-restricted promoter facilitates sustained expression of therapeutic levels of alpha-galactosidase A and the induction of immune tolerance in Fabry mice. Mol Ther 9: 231-240. 45. Xu, L., M. Mei, X. Ma, and K. P. Ponder. 2007. High expression reduces an antibody response after neonatal gene therapy with B domain-deleted human factor VIII in mice. J Thromb Haemost 5: 1805-1812. 46. Mingozzi, F., Y. L. Liu, E. Dobrzynski, A. Kaufhold, J. H. Liu, Y. Wang, V. R. Arruda, K. A. High, and R. W. Herzog. 2003. Induction of immune tolerance to coagulation factor IX antigen by in vivo hepatic gene transfer. J Clin Invest 111: 1347-1356. 47. Lee, H. C., S. J. Kim, K. S. Kim, H. C. Shin, and J. W. Yoon. 2000. Remission in models of type 1 diabetes by gene therapy using a single-chain insulin analogue. Nature 408: 483-488. 48. Park, Y. M., S. Woo, G. T. Lee, J. Y. Ko, Y. Lee, Z. S. Zhao, H. J. Kim, C. W. Ahn, B. S. Cha, K. S. Kim, C. W. Park, and H. C. Lee. 2005. Safety and efficacy of adeno-associated viral vector-mediated insulin gene transfer via portal vein to the livers of streptozotocin-induced diabetic Sprague-Dawley rats. J Gene Med 7: 621-629. 49. Han, G., R. Wang, G. Chen, J. Wang, R. Xu, J. Feng, M. Yu, X. Wu, J. Qian, B. Shen, and Y. Li. 2008. Gene delivery GAD 500 autoantigen by AAV serotype 1 prevented diabetes in NOD mice: transduction efficiency do not play important roles. Immunol Lett 115: 110-116. 50. Gao, G. P., Y. Lu, X. Sun, J. Johnston, R. Calcedo, R. Grant, and J. M. Wilson. 2006. High-level transgene expression in nonhuman primate liver with novel adeno-associated virus serotypes containing self-complementary genomes. J Virol 80: 6192-6194. 51. Chen, C. C., C. P. Sun, H. I. Ma, C. C. Fang, P. Y. Wu, X. Xiao, and M. H. Tao. 2009. Comparative study of anti-hepatitis B virus RNA interference by double-stranded adeno-associated virus serotypes 7, 8, and 9. Mol Ther 17: 352-359. 52. Jooss, K., Y. Yang, K. J. Fisher, and J. M. Wilson. 1998. Transduction of dendritic cells by DNA viral vectors directs the immune response to transgene products in muscle fibers. J Virol 72: 4212-4223. 53. Zhang, Y., N. Chirmule, G. Gao, and J. Wilson. 2000. CD40 ligand-dependent activation of cytotoxic T lymphocytes by adeno-associated virus vectors in vivo: role of immature dendritic cells. J Virol 74: 8003-8010. 54. Gershwin, M. E., I. R. Mackay, A. Sturgess, and R. L. Coppel. 1987. Identification and specificity of a cDNA encoding the 70 kd mitochondrial antigen recognized in primary biliary cirrhosis. J Immunol 138: 3525-3531. 55. LoDuca, P. A., B. E. Hoffman, and R. W. Herzog. 2009. Hepatic gene transfer as a means of tolerance induction to transgene products. Curr Gene Ther 9: 104-114. 56. Miao, C. H., K. Ohashi, G. A. Patijn, L. Meuse, X. Ye, A. R. Thompson, and M. A. Kay. 2000. Inclusion of the hepatic locus control region, an intron, and untranslated region increases and stabilizes hepatic factor IX gene expression in vivo but not in vitro. Mol Ther 1: 522-532. 57. Miao, C. H., A. R. Thompson, K. Loeb, and X. Ye. 2001. Long-term and therapeutic-level hepatic gene expression of human factor IX after naked plasmid transfer in vivo. Mol Ther 3: 947-957. 58. Manno, C. S., G. F. Pierce, V. R. Arruda, B. Glader, M. Ragni, J. J. Rasko, M. C. Ozelo, K. Hoots, P. Blatt, B. Konkle, M. Dake, R. Kaye, M. Razavi, A. Zajko, J. Zehnder, P. K. Rustagi, H. Nakai, A. Chew, D. Leonard, J. F. Wright, R. R. Lessard, J. M. Sommer, M. Tigges, D. Sabatino, A. Luk, H. Jiang, F. Mingozzi, L. Couto, H. C. Ertl, K. A. High, and M. A. Kay. 2006. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med 12: 342-347. 59. Pan, W. Y., C. H. Lo, C. C. Chen, P. Y. Wu, S. R. Roffler, S. K. Shyue, and M. H. Tao. 2012. Cancer Immunotherapy Using a Membrane-bound Interleukin-12 With B7-1 Transmembrane and Cytoplasmic Domains. Mol Ther 20: 927-937. 60. Boyle, J. S., C. Koniaras, and A. M. Lew. 1997. Influence of cellular location of expressed antigen on the efficacy of DNA vaccination: cytotoxic T lymphocyte and antibody responses are suboptimal when antigen is cytoplasmic after intramuscular DNA immunization. Int Immunol 9: 1897-1906. 61. Silver, P. B., R. K. Agarwal, S. B. Su, I. Suffia, R. S. Grajewski, D. Luger, C. C. Chan, R. M. Mahdi, J. M. Nickerson, and R. R. Caspi. 2007. Hydrodynamic vaccination with DNA encoding an immunologically privileged retinal antigen protects from autoimmunity through induction of regulatory T cells. J Immunol 179: 5146-5158. 62. Luth, S., S. Huber, C. Schramm, T. Buch, S. Zander, C. Stadelmann, W. Bruck, D. C. Wraith, J. Herkel, and A. W. Lohse. 2008. Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune neuroinflammation by inducing antigen-specific Tregs. J Clin Invest 118: 3403-3410. 63. Wu, Z., H. Yang, and P. Colosi. 2010. Effect of genome size on AAV vector packaging. Mol Ther 18: 80-86. 64. Moteki, S., P. S. Leung, R. L. Coppel, E. R. Dickson, M. M. Kaplan, S. Munoz, and M. E. Gershwin. 1996. Use of a designer triple expression hybrid clone for three different lipoyl domain for the detection of antimitochondrial autoantibodies. Hepatology 24: 97-103. 65. Mays, L. E., and J. M. Wilson. 2011. The complex and evolving story of T cell activation to AAV vector-encoded transgene products. Mol Ther 19: 16-27. 66. Mount, J. D., R. W. Herzog, D. M. Tillson, S. A. Goodman, N. Robinson, M. L. McCleland, D. Bellinger, T. C. Nichols, V. R. Arruda, C. D. Lothrop, Jr., and K. A. High. 2002. Sustained phenotypic correction of hemophilia B dogs with a factor IX null mutation by liver-directed gene therapy. Blood 99: 2670-2676. 67. Nakai, H., S. Fuess, T. A. Storm, S. Muramatsu, Y. Nara, and M. A. Kay. 2005. Unrestricted hepatocyte transduction with adeno-associated virus serotype 8 vectors in mice. J Virol 79: 214-224. 68. Virgin, H. W., E. J. Wherry, and R. Ahmed. 2009. Redefining chronic viral infection. Cell 138: 30-50. 69. Rizzuto, R., A. W. Simpson, M. Brini, and T. Pozzan. 1992. Rapid changes of mitochondrial Ca2+ revealed by specifically targeted recombinant aequorin. Nature 358: 325-327. | |
dc.identifier.uri | http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/66009 | - |
dc.description.abstract | 中文摘要
原發性膽管硬化症是慢性且進程緩慢的肝臟自體免疫疾病,患者肝臟的門脈三角出現自體反應T淋巴球浸潤以及肝內膽管遭受破壞,抗粒線體抗體的產生為原發性膽管硬化症的指標,大約95%的患者體內可測得此抗體。由於目前治療方法的治療效果相當有限,因此發展有效的治療方式為當務之急。 先前研究已知,患者的自體反應T淋巴球和抗粒線體抗體以位於粒線體基質的2-oxo-acid dehydrogenase enzyme complex (2-OADC) 家族為攻擊目標,而E2 subunit of pyruvate dehydrogenase complex (PDC-E2) 為主要的自體抗原。從已知患者與小鼠模式的研究結果,推測對PDC-E2自體抗原失去免疫耐受性是原發性膽管硬化症產生的原因。 肝臟為一個傾向引發免疫耐受性的淋巴器官,是因為其具有對外來抗原產生免疫耐受性的特殊抗原呈現細胞,因此我們假設以具有肝臟特異性的腺相關病毒載體,攜帶有肝臟侷限性表現的PDC-E2,能夠回復Xenobiotic引發原發性膽管硬化症的小鼠的免疫耐受性, 我們構築腺相關病毒載體,其攜帶具有肝臟侷限性於粒腺體或細胞膜表現的PDC-E2或truncated PDC-E2,而transgene的表現以及在細胞的表現位置已經在細胞和小鼠體內被證實。在動物實驗中,我們發現PDC-E2和膜結合PDC-E2幾乎沒有抑制Xenobiotic引發anti-PDC-E2抗體產生的效果,相反地,膜結合truncated PDC-E2明顯抑制重組PDC-E2蛋白免疫引發anti-PDC-E2抗體產生的效果,此結果為將來探討小鼠模式中病理反應奠定基礎。 基於我們目前的發現,在未來的實驗中,將專注於探討膜結合truncated PDC-E2是否能減緩Xenobiotic引發的病理現象的研究。 | zh_TW |
dc.description.abstract | Primary biliary cirrhosis (PBC) is a chronic, slow progressive liver autoimmune disease. It is characterized by the infiltration of auto-reactive T cells in portal tracts and the destruction of the intrahepatic bile ducts. Anti-mitochondrial antibodies (AMA), the hallmark of PBC, are detected in approximately 95% of patients. Since current therapy of PBC has been disappointing, it is of an urgent need to develop an more effective therapy for PBC.
Considerable data shown, the auto-reactive T cells and AMAs of PBC patients target members of the 2-oxo-acid dehydrogenase enzyme complexes (2-OADC) which are located in the inner mitochondrial matrix. Among them, the major autoantigen is the E2 subunit of pyruvate dehydrogenase complex (PDC-E2) . Indeed, data in patients and in PBC animal models have shown that break of tolerance to PDC-E2 causes pathology of PBC. Liver is a lymphoid organ favoring induction of immune tolerance, which may be correlated with the tolerogenic properties of hepatic antigen presenting cells. We hypothesized that hepatotropic adeno-associated virus delivery and hepatocyte-restricted PDC-E2 expression could restore immune tolerance in Xenobiotic induced PBC model. We constructed AAV vectors expressing hepatocyte restricted- PDC-E2 or truncated PDC-E2 in mitochondria or on cell membrane. Expression and cellular localization of the transgene was confirmed in vitro and in vivo. In animal experiments, we found native and membrane-bound PDC-E2 had little effect on 2OA-BSA-induced anti-PDC-E2 antibodies. In contrast, the membrane-bound form of truncated PDC-E2 greatly improved its surface expression and significantly suppressed anti-PDC-E2 antibodies induced by recombinant PDC-E2 immunization. The finding lay the foundation for investgating the influence of PBC pathology by hepatic expression PDC-E2. Based on our current finding, we will focus on whether PBC pathology can be alleviated by hepatic expression of membrane-bound truncated PDC-E2. | en |
dc.description.provenance | Made available in DSpace on 2021-06-17T00:18:50Z (GMT). No. of bitstreams: 1 ntu-101-R99424003-1.pdf: 4943622 bytes, checksum: 355a8a92359ba9e66b71b1943b57cedf (MD5) Previous issue date: 2012 | en |
dc.description.tableofcontents | 中文摘要 i
Abstract ii 圖表目錄 vi 第一章 緒論 1 第一節、原發性膽管硬化症 (primary biliary cirrhosis ; PBC) 1 第二節、PBC的動物模式 3 第三節、肝臟微環境 (liver microenvironment) 5 第四節、腺相關病毒載體 7 第五節、研究動機與實驗目的 8 第六節、構築具肝臟侷限性表現PDC-E2抗原的重組腺相關病毒 (AAV) 載體 9 第二章 材料與方法 10 第一節、質體的構築 10 第二節、細胞株培養與細胞表現檢驗 11 第三節、包裝腺相關病毒 12 第四節、動物實驗 13 第三章 實驗結果 16 第一部分、構築腺相關病毒載體於肝臟表現PDC-E2蛋白預防或治療 2OA-BSA induced PBC小鼠 16 第一節、構築表達螢火蟲的luciferase和PDC-E2的腺相關病毒載體 16 第二節、證實在hAAT promoter控制下AAV載體具有肝臟特異性 16 第三節、在細胞和小鼠證實PDC-E2蛋白表現 17 第四節、重組AAV對於重組PDC-E2蛋白免疫生成anti-PDC-E2抗體的影響 19 第五節、探討重組AAV預防2OA-BSA引發小鼠產生anti-PDC-E2抗體的效果 20 第六節、探討重組AAV治療抑制2OA-BSA引發產生anti-PDC-E2抗體的效果 21 第二部分、構築腺相關病毒載體於肝臟表現truncated PDC-E2蛋白預防或治療2OA-BSA induced PBC小鼠 21 第一節、構築表達具肝臟特異性表現的truncated PDC-E2腺相關病毒載體 22 第二節、於小鼠可測得luciferase的活性 22 第三節、在細胞和小鼠證實truncated PDC-E2蛋白表現 22 第四節、重組AAV對於重組PDC-E2蛋白免疫生成anti-PDC-E2抗體的影響 25 第四章、討論 28 參考文獻 32 | |
dc.language.iso | zh-TW | |
dc.title | 以重組腺相關病毒載體在肝臟表現自體抗原PDC-E2來治療原發性膽管硬化症 | zh_TW |
dc.title | Treatment of Primary Biliary Cirrhosis by Recombinant AAV-mediated Hepatic PDC-E2 | en |
dc.type | Thesis | |
dc.date.schoolyear | 100-2 | |
dc.description.degree | 碩士 | |
dc.contributor.oralexamcommittee | 莊雅惠(Ya-Hui Chuang),郭敏玲(Ming-Ling Kuo) | |
dc.subject.keyword | 原發性膽管硬化症, | zh_TW |
dc.subject.keyword | PDC-E2,PBC, | en |
dc.relation.page | 62 | |
dc.rights.note | 有償授權 | |
dc.date.accepted | 2012-06-28 | |
dc.contributor.author-college | 醫學院 | zh_TW |
dc.contributor.author-dept | 醫學檢驗暨生物技術學研究所 | zh_TW |
顯示於系所單位: | 醫學檢驗暨生物技術學系 |
文件中的檔案:
檔案 | 大小 | 格式 | |
---|---|---|---|
ntu-101-1.pdf 目前未授權公開取用 | 4.83 MB | Adobe PDF |
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。