博碩士論文 103324032 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:33 、訪客IP:3.140.188.16
姓名 黃詩淳(Shih-Chun Huang)  查詢紙本館藏   畢業系所 化學工程與材料工程學系
論文名稱 以二元體形式之Indolicidin 應用於去氧寡核苷酸之輸送
(The Use of Dimeric Indolicidin for Oligodeoxynucleotide Delivery Application)
相關論文
★ 利用穿膜胜肽改善帶正電高分子之轉染效率★ 利用導電高分子聚吡咯為基材以電刺激促進幹細胞分化
★ 以電刺激增進骨髓基質細胞骨分化之最佳化探討★ 利用電場控制導電性高分子以進行基因於聚電解質多層膜的組裝
★ 以短鏈胜肽接枝聚乙烯亞胺來進行基因輸送應用之研究★ 電紡絲製備褐藻酸鈉/聚己內酯之奈米複合纖維進行原位轉染
★ 電場對於複合奈米絲進行原位基因傳送之影響★ 利用電場調控聚電解質多層膜的釋放 以應用於基因輸送
★ 發展載藥電紡聚乳酸/多壁奈米碳管/聚乙二醇纖維★ 利用寡聚精胺酸促進去氧寡核苷酸輸送
★ 利用聚己內酯/褐藻酸鈉之複合電紡絲擴增癌症幹細胞★ Indolicidin之色胺酸殘基對於轉染效率的影響
★ Indolicidin之二聚體形式對輸送去氧寡核?酸的影響★ 搭建可提供電刺激與機械刺激之生物反應器
★ 硬脂基化的Indolicidin作為傳送質體去氧核 酸的非病毒載體★ 開發促進傷口癒合之複合敷料
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 ( 永不開放)
摘要(中) 穿膜胜肽Indolicidin (IL)的毒性限制了其臨床應用性,為了增進生物適合性,本研究合成二元體形式之IL並討論其應用於輸送去氧寡核苷酸(ODN)的效果。藉由分別對Indolicidin其C端與N端接上一個半胱胺酸,胜肽得以形成雙硫鍵成為二元體,我們分別將其命名為ILC與CIL。以HPLC與MASS分析證明二元體之形成。這些二元體可以藉靜電複合的方式將之與ODN自組裝成奈米粒子。由MTT assay可以發現,相比於IL,CIL與ILC可以有效降低細胞毒性。利用動態雷射散射粒徑分析儀測量複合物之粒徑大小介於500至2000nm,在巨噬細胞可胞吞範圍內。在表面電位的表現上CIL與IL的電位可以隨著N/P比而提高其電位,但是ILC顯然地不易吸附於ODN,要到N/P 30以上才有正電位。以膠體電泳方式測試其包覆率,IL與CIL皆有良好的複合能力,可包覆ODN形成穩定的粒子,但是ILC可能因為正電區被疏水結構所遮蔽,因此無法有效包覆ODN。雖然如此,螢光顯微鏡顯示以二元體形式之ILC及CIL均有助於提高細胞攝取效率,皆有明顯的螢光亮點。雷射共軛焦顯微鏡顯示,三種胜肽均可能透過胞吞的方式使細胞攝取複合物,而CIL能助於ODN自內體逃脫而進入細胞質。抑制蛋白質實驗中,藉由ELISA分析被脂多糖(Lipopolysaccharide,LPS)刺激的Raw264.7細胞所表現TNF-α量,結果顯示CIL作為基因載體有較佳的抑制結果,且可維持長時間的藥效。因此可作為具有潛力的去氧寡核苷酸載體。
摘要(英) Indolicidin (IL) is a potential cell-penetrating peptide (CPP), however, its high cytotoxicity restricts its clinical application. In this study, we prepared dimeric IL peptides to promote their biocompatibility and investigate their oligodeoxynucleotide (ODN) delivery efficiencies. Cysteine residues were added to the C or N terminals of IL, by which peptides can be linked as dimers through disulfide bond formation, and we denoted them as ILC and CIL, respectively. HPLC and MASS analyses suggested dimers were successfully prepared in both designs. These dimeric peptides can complex with ODN to self-assembly as nanopartiocles. The MTT results suggested that both ILC and CIL exhibited low cytotoxicity compared to the IL group. The DLS results demonstrated that the sizes of formed peptideplex ranged from 500 to 2000 nm, which were suitable for macrophage uptake. Zeta potentials of peptideplex increased with increasing N/P ratio so that these complexes can be positive charged, however, the absorbance of ILC to ODN is inefficient that the surface charges were always negative except N/P 30. The ODN encapsulation efficiencies of peptides were evaluated by gel retardation assay. Both IL and CIL have good complexation ability to load ODN as stable nanocomplex. In contrast, ILC did not stably complex ODN, which probably correlative to its structure. Because the cationic domain of dimeric ILC is in the middle regions, the hydrophobic domains in two ends probably may mask the positive charges and inhibit the electrostatic interaction between peptide and ODN. However, the fluorescent labeling experiment suggested that both ILC and CIL can promote ODN internalization to Raw264.7 cells. We further investigate the internalization pathway by confocal microscopy, and the results suggested ODN delivered in all groups can be internalized through endocytosis. The long-time observation results indicated that the ODN delivered by CIL can escape from endosome to enter cytosol. Finally, the inhibition experiments were performed against the expression of TNF-α. Anti-TNF-α ODN was delivered to Raw264.7 cell by peptide carriers, and the enzyme-linked immunosorbent assay (ELISA) was performed to measure the expression of TNF-α from Raw264.7 stimulated by lipopolysaccharide (LPS). The results demonstrated CIL demonstrated the best silence against TNF-α, and the inhibition duration can be elongated, suggesting CIL is a potential vector for ODN delivery.
關鍵字(中) ★ 胜肽
★ 去氧寡核苷酸
關鍵字(英) ★ peptide
★ oligodeoxynucleotide
論文目次 目錄
摘要 I
Abstract III
致謝 V
目錄 VII
圖目錄 XI
表目錄 XIII
第一章 緒論 1
1.1 研究背景 1
1.2 研究動機 3
第二章 文獻回顧 4
2.1 免疫反應 4
2.1.1 免疫系統 4
2.1.2 Cytokine在免疫反應的角色 5
2.1.3 現有免疫疾病治療的方式 6
2.2 基因治療 8
2.2.1 基因沉默 8
2.2.2 核糖核酸干擾 9
2.2.3 反義寡核苷酸 12
2.3 用於基因沉默的載體選擇 15
2.3.1 陽離子型高分子 15
2.3.2 脂質 16
2.3.3 細胞穿膜胜肽 17
2.4 以胜肽轉染應用於基因沉默 19
2.4.1 細胞攝取胜肽之機制 19
2.4.2 利用胜肽進行藥物輸送 20
2.5 Indolicidin 22
2.5.1 Indolicidin與CPP之關係 22
2.5.2 Indolicidin穿膜機制與應用 23
2.6 以二元體形式應用於藥物輸送 24
第三章 實驗藥品、儀器設備與方法 26
3.1 實驗材料 26
3.1.1 合成材料 26
3.1.2 細胞培養 27
3.1.3 定性定量分析 28
3.2 儀器設備 30
3.3 實驗方法 32
3.3.1 配製溶液 32
3.3.2 胜肽以HPLC與MASS分析 36
3.3.3 複合物製備 37
3.3.4 粒徑大小與表面電位(DLS) 38
3.3.5 包覆率 38
3.3.6 細胞存活率(MTT) 39
3.3.7 螢光顯微鏡(Fluorescent microscope) 40
3.3.8 雷射共軛焦顯微鏡(Confocol) 41
3.3.9 蛋白質抑制實驗 43
第四章 結果與討論 46
4.1 二元體 46
4.1.1 HPLC分析 46
4.1.2 以MASS分析二元體的分子量 50
4.2 奈米粒子的物性鑑定 54
4.2.1 表面電位 54
4.2.2 粒徑大小 56
4.2.3 包覆率 57
4.3 載體應用於ODN輸送的生物適合性測試 59
4.4 穿膜胜肽對細胞攝取的影響 61
4.4.1 螢光顯微鏡分析細胞攝取量 61
4.4.2 雷射共軛焦顯微鏡探討細胞攝取 65
4.4.3 蛋白質抑制實驗 70
第五章 結論 75
第六章 參考文獻 77
參考文獻 P. Marrack, J. Kappler, and B.L. Kotzin, "Autoimmune disease: why and where it occurs," Nature Medicine, vol. 7, pp. 899-905, 2001.
[2] M. Palladino, F. Bahjat, E. Theodorakis, and L. Moldawer, "Anti-TNF-alpha therapies: the next generation," Nature Reviews Drug Discovery, vol. 2, pp. 736-46, Sep 2003.
[3] Z. Huang, Z. Zhang, Y. Zha, J. Liu, Y. Jiang, Y. Yang, J. Shao, X. Sun, X. Cai, Y. Yin, J. Chen, L. Dong, and J. Zhang, "The effect of targeted delivery of anti-TNF-α oligonucleotide into CD169+ macrophages on disease progression in lupus-prone MRL/lpr mice," Biomaterials, vol. 33, pp. 7605-7612, 2012.
[4] B. Layek and J. Singh, "Cell penetrating peptide conjugated polymeric micelles as a high performance versatile nonviral gene carrier," Biomacromolecules, vol. 14, pp. 4071-81, 2013.
[5] M. Elsabahy, A. Nazarali, and M. Foldvari, "Non-Viral Nucleic Acid Delivery: Key Challenges and Future Directions," Current Drug Delivery, vol. 8, pp.235-244, 2011.
[6] M. Elsabahy, A. Nazarali, and M. Foldvari, "Non-Viral Nucleic Acid Delivery: Key Challenges and Future Directions," vol. 8, 2011.
[7] H. Amand, B. Norden, and K. Fant, "Functionalization with C-terminal cysteine enhances transfection efficiency of cell-penetrating peptides through dimer formation," Biochem Biophys Res Commun, vol. 418, pp. 469-74, 2012.
[8] M. Moser and O. Leo, "Key concepts in immunology," Vaccine, vol. 28 Suppl 3, pp. C2-13, 2010.
[9] K. Murphy and C. Weaver, Janeway′s immunobiology: Garland Science, 2016.
[10] J. Parkin and B. Cohen, "An overview of the immune system," The Lancet, vol. 357, pp. 1777-1789, 2001.
[11] A. Hajeer and I. Hutchinson, "TNF-α Gene Polymorphism: Clinical and Biological Implications," Microscopy Research and Technique, pp. 216–228, 2000.
[12] W. Fiers, "Tumor necrosis factor Characterization at the molecular, cellular and in vivo level ." Federation of European Biochemical Societies vol. 285, pp. 199-212 1992.
[13] H. Gordon, G. Kucera, R. Salvo, and J. Boss, "Tumor necrosis factor induces genes involved in inflammation, cellular and tissue repair, and metabolism in murine fibroblasts," The Journal of Immunology, vol. 148, pp. 4021-4027, 1992.
[14] D.J. Gould, C. Bright, and Y. Chernajovsky, "Inhibition of established collagen-induced arthritis with a tumour necrosis factor-alpha inhibitor expressed from a self-contained doxycycline regulated plasmid," Arthritis Research & Therapy, vol. 6, pp. R103-13, 2004.
[15] M. Stephenson and P. Zamecnik, "Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide," Proceedings of the National Academy of Sciences of the United States of America, vol. 75, pp. 285-288, 1978.
[16] P.C. Zamecnik and M.L. Stephenson, "Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide," Proceedings of the National Academy of Sciences of the United States of America, vol. 75, pp. 280-284, 1978.
[17] J. Kurreck, "Antisense technologies," European Journal of Biochemistry, vol. 270, pp. 1628-1644, 2003.
[18] A.d. Fougerolles, H. Vornlocher, J. Maraganore, and J. Lieberman, "Interfering with disease: a progress report on siRNA-based therapeutics," Nature Reviews Drug Discovery, vol. 6, pp. 443-453, 2007.
[19] D. Grimm and M. Kay, "Therapeutic application of RNAi: is mRNA targeting finally ready for prime time?," The Journal of Clinical Investigation, vol. 117, pp. 3633-3641, 2007.
[20] T. Doerks, R. Copley, J. Schultz, C. Ponting, and P. Bork, "Systematic identification of novel protein domain families associated with nuclear functions," Genome Research, vol. 12, pp. 47-56, 2002.
[21] E. Bernstein, A. Caudy, S. Hammond, and G. Hannon, "Role for a bidentate ribonuclease in the initiation step of RNA interference," Nature, vol. 409, pp. 363-366, 2001.
[22] D. Yang, H. Lu, and J. Erickson, "Evidence that processed small dsRNAs may mediate sequence-specific mRNA degradation during RNAi in Drosophila embryos," Current Biology, vol. 10, pp. 1191-1200, 2000.
[23] J. Martinez, A. Patkaniowska, H. Urlaub, R. Lührmann, and T. Tuschl, "Single-Stranded Antisense siRNAs Guide Target RNA Cleavage in RNAi," Cell, vol. 110, pp. 563-574, 2002.
[24] T. Dowler, D. Bergeron, A. Tedeschi, L. Paquet, N. Ferrari, and M. Damha, "Improvements in siRNA properties mediated by 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid (FANA)," Nucleic Acids Research, vol. 34, pp. 1669-1675, 2006.
[25] J. Wang, Z. Lu, M. Wientjes, and J. Au, "Delivery of siRNA Therapeutics: Barriers and Carriers," The American Association of Pharmaceutical Scientists Journal, vol. 12, pp. 492-503, 2010.
[26] Y. Lee, K. Jeon, J. Lee, S. Kim, and V. Kim, "MicroRNA maturation: stepwise processing and subcellular localization," The European Molecular Biology OrganizationJournal, vol. 21, pp. 4663-4670, 2002.
[27] E. Lund, S. Güttinger, A. Calado, J. Dahlberg, and U. Kutay, "Nuclear Export of MicroRNA Precursors," Science, vol. 303, pp. 95-98, 2004.
[28] R. Yi, Y. Qin, I. Macara, and B. Cullen, "Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs," Genes & Development, vol. 17, pp. 3011-3016, 2003.
[29] Y. Zeng and B. Cullen, "Structural requirements for pre-microRNA binding and nuclear export by Exportin 5," Nucleic Acids Research, vol. 32, pp. 4776-4785, 2004.
[30] Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim, J. Yim, J. Lee, P. Provost, O. Radmark, S. Kim, and V. Kim, "The nuclear RNase III Drosha initiates microRNA processing," Nature, vol. 425, pp. 415-419, 2003.
[31] S. Sassen, E. Miska, and C. Caldas, "MicroRNA—implications for cancer," Virchows Archiv, vol. 452, pp. 1-10, 2008.
[32] P. Paddison, A. Caudy, E. Bernstein, G. Hannon, and D. Conklin, "Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells," Genes & Development, vol. 16, pp. 948-958, 2002.
[33] Y. Chen, G. Cheng, and R. Mahato, "RNAi for Treating Hepatitis B Viral Infection," Pharmaceutical Research, vol. 25, pp. 72-86, 2008.
[34] K. Karikó, P. Bhuyan, J. Capodici, and D. Weissman, "Small Interfering RNAs Mediate Sequence-Independent Gene Suppression and Induce Immune Activation by Signaling through Toll-Like Receptor 3," The Journal of Immunology, vol. 172, pp. 6545-6549, 2004.
[35] G. Deleavey and M. Damha, "Designing chemically modified oligonucleotides for targeted gene silencing," Chemistry and Biology, vol. 19, pp. 937-54, 2012.
[36] M. Mansoor and A. Melendez, "Advances in Antisense Oligonucleotide Development for Target Identification, Validation, and as Novel Therapeutics," Gene Regulation and Systems Biology, vol. 2, pp. 275-295, 2008.
[37] J. Chan, S. Lim, and W. Wong, "Antisense Oligonucleotides:From Design to Therapeutic Application," Clinical and Experimental Pharmacology and Physiology, vol. 33, pp. 533-540, 2006.
[38] P. Sazani and R. Kole, "Therapeutic potential of antisense oligonucleotides as modulators of alternative splicing," Journal of Clinical Investigation, vol. 112, pp. 481-486, 2003.
[39] E. Dinç, "Antisense Oligodeoxynucleotide Technology: A Novel Tool for Gene Silencing in Higher Plants," Institute of Plant Biology Biological Research Centre Hungarian Academy of Sciences Doctoral School of Biology University of Szeged 2012.
[40] L. Dong, S. Xia, Y. Luo, H. Diao, J. Zhang, J. Chen, and J. Zhang, "Targeting delivery oligonucleotide into macrophages by cationic polysaccharide from Bletilla striata successfully inhibited the expression of TNF-α," Journal of Controlled Release, vol. 134, pp. 214-220, 2009.
[41] F. Eckstein, "A dinucleoside phosphorothioate," Tetrahedron Letters, vol. 8, pp. 1157-1160, 1967.
[42] F. Eckstein, "Nucleoside phosphorothioates," Journal of the American Chemical Society, vol. 92, pp. 4718-4723, 1970.
[43] Y.S. Sanghvi, "A Status Update of Modified Oligonucleotides for Chemotherapeutics Applications," in Current Protocols in Nucleic Acid Chemistry, ed: John Wiley & Sons, Inc., 2001.
[44] T. Aboul-Fadl, "Antisense Oligonucleotides: The State of the Art," Current Medicinal Chemistry, vol. 12, pp. 2193-2214, 2005.
[45] B. Yu, X. Zhao, L. Lee, and R. Lee, "Targeted Delivery Systems for Oligonucleotide Therapeutics," The American Association of Pharmaceutical Scientists Journal, vol. 11, pp. 195-203, 2009.
[46] A. Koirala, S. Conley, and M. Naash, "A review of therapeutic prospects of non-viral gene therapy in the retinal pigment epithelium," Biomaterials, vol. 34, pp. 7158-7167, 2013.
[47] Y. He, G. Cheng, L. Xie, Y. Nie, B. He, and Z. Gu, "Polyethyleneimine/DNA polyplexes with reduction-sensitive hyaluronic acid derivatives shielding for targeted gene delivery," Biomaterials, vol. 34, pp. 1235-1245, 2013.
[48] M. Xun, Y. Xiao, J. Zhang, Y. Liu, Q. Peng, Q Guo, W. Wu, Y. Xu, and X. Yu, "Low molecular weight PEI-based polycationic gene vectors via Michael addition polymerization with improved serum-tolerance," Polymer, vol. 65, pp. 45-54, 2015.
[49] B. Urban-Klein, S. Werth, S. Abuharbeid, F. Czubayko, and A. Aigner, "RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo," Gene therapy, vol. 12, pp. 461-466, 2004.
[50] A. Khan, M. Benboubetra, P. Sayyed, K.W. Ng, S. Fox, G. Beck, I. Benter, and S. Akhtar, "Sustained Polymeric Delivery of Gene Silencing Antisense ODNs, siRNA, DNAzymes and Ribozymes: In Vitro and In Vivo Studies," Journal of Drug Targeting, vol. 12, pp. 393-404, 2004.
[51] P. Cullis, M. Hope, and C. Tilcock, " Lipid Polymorphism and the Roles of Lipid in Membranes," Chemistry and Physics of Lipids pp. 127-144, 1986.
[52] H. Farhood, N. Serbina, and L. Huang, "The role of dioleoyl phosphatidylethanolamine in cationic liposome mediated gene transfer," Biochimica et Biophysica Acta (BBA) - Biomembranes, vol. 1235, pp. 289-295, 1995.
[53] T. Allen and A. Chonn, "Large unilamellar liposomes with low uptake into the reticuloendothelial system," Federation of European Biochemical Societies, vol. 223, pp. 42-46, 1987.
[54] P. Hug and R. Sleight, "Liposomes for the transformation of eukaryotic cells," Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol. 1097, pp. 1-17, 1991.
[55] W. Zauner, M Ogris, and E. Wagner, "Polylysine-Based Transfection Systems Utilizing Receptor-Mediated Delivery.," Advanced Drug Delivery Reviews vol. 30, p. 97, 1998.
[56] J. Rothbard, E. Kreider, C. VanDeusen, L. Wright, B. Wylie, and P. Wender, "Ariginine-Rich Molecular Transporters for Drug Delivery:Role of Backbone Spacing in Cellular Uptake.," Journal of Medicinal Chemistry vol. 45, p. 3612, 2002.
[57] C. Pichon, C. Gonçalves, and P. Midoux, "Histidine-Rich Peptides and Polymers for Nuclei Acids Delivery.," Advanced Drug Delivery Reviews, vol. 53, p. 75, 2001.
[58] P. Midoux, C. Pichon, J. Yaouanc, and P. Jaffrès, "Chemical Vectors for Gene Delivery:A Current Review on Polymers,Peptides and Lipids Containing Histidine or Imidazole As Nuclei Acids Carriers.," British Journal of Pharmacology, vol. 157, p. 166, 2009.
[59] W. Seow and Y. Yang, "A Class of Cationic Triblock Amphiphilic Oligopeptides as Efficient Gene-Delivery Vectors.," Advanced Materials vol. 21, p. 86, 2009.
[60] D. Derossi, S. Calvet, A. Trembleau, A. Brunissen, G. Chassaing, and A. Prochiantz, "Cell Internalization of the Third Helix of the Antennapedia Homeodomain Is Receptor-independent," Journal of Biological Chemistry, vol. 271, pp. 18188-18193, 1996.
[61] K. Matsuzaki, S. Yoneyama, O. Murase, and K. Miyajima, "Transbilayer Transport of Ions and Lipids Coupled with Mastoparan X Translocation," Biochemistry, vol. 35, pp. 8450-8456, 1996.
[62] Y. Pouny, D. Rapaport, A. Mor, P. Nicolas, and Y. Shai, "Interaction of antimicrobial dermaseptin and its fluorescently labeled analogs with phospholipid membranes," Biochemistry, vol. 31, pp. 12416-12423, 1992.
[63] M. Lee, W. Hung, F. Chen, and H. Huang, "Many-Body Effect of Antimicrobial Peptides: On the Correlation Between Lipid′s Spontaneous Curvature and Pore Formation," Biophysical Journal, vol. 89, pp. 4006-4016.
[64] S. Futaki, "Arginine-rich peptides: potential for intracellular delivery of macromolecules and the mystery of the translocation mechanisms," International Journal of Pharmaceutics, vol. 245, pp. 1-7, 2002.
[65] A. Astriab-Fisher, D. Sergueev, M. Fisher, B.R. Shaw, and R. Juliano, "Antisense inhibition of P-glycoprotein expression using peptide–oligonucleotide conjugates," Biochemical Pharmacology, vol. 60, pp.83-90, 2000.
[66] S. Schwarze, A. Ho, A. Vocero-Akbani, and S. Dowdy, "In Vivo Protein Transduction: Delivery of a Biologically Active Protein into the Mouse," Science, vol. 285, pp. 1569-1572, 1999.
[67] A. Jones, "Macropinocytosis: searching for an endocytic identity and role in the uptake of cell penetrating peptides," Journal of Cellular and Molecular Medicine, vol. 11, pp. 670-684, 2007.
[68] S. Mayor and R. Pagano, "Pathways of clathrin-independent endocytosis," Nature Reviews Molecular Cell Biology, vol. 8, pp. 603-612, 2007.
[69] J. Hoyer and I. Neundorf, "Peptide Vectors for the Nonviral Delivery of Nucleic Acids," Accounts of Chemical Research, vol. 45, pp. 1048-1056,2012.
[70] U. Koppelhus, T. Shiraishi, V. Zachar, S. Pankratova, and P. Nielsen, "Improved Cellular Activity of Antisense Peptide Nucleic Acids by Conjugation to a Cationic Peptide-Lipid (CatLip) Domain," Bioconjugate Chemistry, vol. 19, pp. 1526-1534, 2008.
[71] A. Muratovska and M. Eccles, "Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells," Federation of European Biochemical Societies, vol. 558, pp. 63-68, 2004.
[72] T. Davidson, S. Harel, V. Arboleda, G. Prunell, M. Shelanski, L. Greene, and C. Troy, "Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation," The Journal of Neuroscenice, vol. 24, pp. 10040-6, 2004.
[73] N. Unnamalai, B. Kang, and W. Lee, "Cationic oligopeptide-mediated delivery of dsRNA for post-transcriptional gene silencing in plant cells," Federation of European Biochemical Societies, vol. 566, pp. 307-310, 2004.
[74] F. Simeoni, M. Morris, F. Heitz, and G.D. . "Insight into the mechanism of the peptide-based gene delivery system MPG: implications for delivery of siRNA into mammalian cells," Nucleic Acids Research, vol. 31, pp. 2717-2724, 2003.
[75] F. Madani, S. Lindberg, Ü. Langel, S. Futaki, and A. Gräslund, "Mechanisms of Cellular Uptake of Cell-Penetrating Peptides," Journal of Biophysics, vol. 2011, p. 414729, 2011.
[76] M. Yeaman and N. Yount, "Mechanisms of Antimicrobial Peptide Action and Resistance," Pharmacological Reviews, vol. 55, pp. 27-55, 2003.
[77] J. Hsu and C. Yip, "Molecular Dynamics Simulations of Indolicidin Association with Model Lipid Bilayers," Biophysical Journal, vol. 92, pp. L100-L102, 2007.
[78] J. Shaw, J. Alattia, J. Verity, G. Privé, and C. Yip., "Mechanisms of antimicrobial peptide action: Studies of indolicidin assembly at model membrane interfaces by in situ atomic force microscopy," Journal of Structural Biology, vol. 154, pp. 42-58, 2006.
[79] L. Zhang, A Rozek, and R. Hancock, "Interaction of Cationic Antimicrobial Peptides with Model Membranes," Journal of Biological Chemistry, vol. 276, pp. 35714-35722, 2001.
[80] T. Rokitskaya, N. Kolodkin, E. Kotova, and Y. Antonenko, "Indolicidin action on membrane permeability: Carrier mechanism versus pore formation," Biochimica et Biophysica Acta (BBA) - Biomembranes, vol. 1808, pp. 91-97, 2011.
[81] 林則緯, "利用穿膜胜肽改善帶正電高分子之轉染效率," 化學工程與材料工程學系, 國立中央大學, 2012.
[82] D. McKenzie, K. Kwok, and K. Rice, "A Potent New Class of Reductively Activated Peptide Gene Delivery Agents," Journal of Biological Chemistry, vol. 275, pp. 9970–9977, 2000.
[83] D. Manickam and D. Oupický, "Multiblock Reducible Copolypeptides Containing Histidine-Rich and Nuclear Localization Sequences for Gene Delivery," Bioconjugate Chemistry, vol. 17, pp. 1395-1403, 2006.
[84] M. Balakirev, G. Schoehn, and J. Chroboczek, "Lipoic acid-derived amphiphiles for redox-controlled DNA delivery," Chemistry & Biology, vol. 7, pp. 813-819.
[85] M. Read, K. Bremner, D. Oupický, N. Green, P. Searle, and L. Seymour, "Vectors based on reducible polycations facilitate intracellular release of nucleic acids," The Journal of Gene Medicine, vol. 5, pp. 232-245, 2003.
[86] X. Loh, T. Lee, Q. Dou, and G. Deen, "Utilising inorganic nanocarriers for gene delivery," Biomaterials Science, vol. 4, pp. 70-86, 2016.
[87] T. Green, J. Fisher, M. Stone, B. Wroblewski, and E. Ingham, "Polyethylene particles of a ‘critical size’ are necessary for the induction of cytokines by macrophages in vitro," Biomaterials, vol. 19, pp. 2297-2302, 1998.
[88] O. Calonius and V. Saikko, "Analysis of Polyethylene Particles Produced in Different Wear Conditions In Vitro," Clinical Orthopaedics and Related Research, vol. 399, pp. 219-230, 2002.
[89] Y. Liu, D. Peterson, H. Kimura, and D. Schubert, "Mechanism of Cellular 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) Reduction," Journal of Neurochemistry, vol. 69, pp. 581-593, 1997.
[90] R. Pan, W. Xu, F. Yuan, D. Chu, Y. Ding, B. Chen, M. Jafari, Y. Yuan, and P. Chen, "A novel peptide for efficient siRNA delivery in vitro and therapeutics in vivo," Acta Biomaterialia, vol. 21, pp. 74-84, 2015.
指導教授 胡威文(Wei-Wen Hu) 審核日期 2016-8-30
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明