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
  • 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. 醫學院
  3. 解剖學暨細胞生物學科所
請用此 Handle URI 來引用此文件: http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/90440
完整後設資料紀錄
DC 欄位值語言
dc.contributor.advisor林能裕zh_TW
dc.contributor.advisorNeng-Yu Linen
dc.contributor.author林容安zh_TW
dc.contributor.authorJung-An Linen
dc.date.accessioned2023-10-02T16:13:38Z-
dc.date.available2023-11-10-
dc.date.copyright2023-10-02-
dc.date.issued2023-
dc.date.submitted2023-08-10-
dc.identifier.citation"Assessment of NIH Research on Autoimmune Diseases". www.nationalacademies.org. Retrieved 2022-06-13.
Varma Avi. Everything to Know About Autoimmune Diseases. Healthline, 2023 May 24.
Rosenblum MD, Gratz IK, Paw JS, Abbas AK. Treating human autoimmunity: current practice and future prospects. Sci Transl Med. 2012 Mar 14;4(125):125sr1.
Li P, Zheng Y, Chen X. Drugs for Autoimmune Inflammatory Diseases: From Small Molecule Compounds to Anti-TNF Biologics. Front Pharmacol. 2017 Jul 12; 8:460.
Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discov. 2021 Mar;20(3):179-199.
Bonelli M, Göschl L, Blüml S, Karonitsch T, Hirahara K, Ferner E, Steiner CW, Steiner G, Smolen JS, Scheinecker C. Abatacept (CTLA-4Ig) treatment reduces T cell apoptosis and regulatory T cell suppression in patients with rheumatoid arthritis. Rheumatology (Oxford). 2016 Apr;55(4):710-20.
Dalakas MC. Intravenous immunoglobulin in autoimmune neuromuscular diseases. JAMA. 2004 May 19;291(19):2367-75.
Aletaha D, Smolen JS. Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA. 2018;320(13):1360–1372.
Cross M, Smith E, Hoy D, Carmona L, Wolfe F, Vos T, Williams B, Gabriel S, Lassere M, Johns N, Buchbinder R, Woolf A, March L. The global burden of rheumatoid arthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. 2014 Jul;73(7):1316-22.
Lee YH, Tsou HK, Kao SL, Gau SY, Bai YC, Lin MC, Wei JC. Patients With Rheumatoid Arthritis Increased Risk of Developing Osteoarthritis: A Nationwide Population-Based Cohort Study in Taiwan. Front Med (Lausanne). 2020 Sep 10; 7:392.
Angum F, Khan T, Kaler J, Siddiqui L, Hussain A. The Prevalence of Autoimmune Disorders in Women: A Narrative Review. Cureus. 2020 May 13;12(5): e8094.
Baker JF. Diagnosis and differential diagnosis of rheumatoid arthritis. UpToDate. 2022 May 28.
Kelly S. Rethinking the role of pathogenic drivers in rheumatoid arthritis. Bristol Myers Squibb. 2019 Dec 16.
Blüml, S., Redlich, K. & Smolen, J.S. Mechanisms of tissue damage in arthritis. Semin Immunopathol 36, 531–540 (2014).
Müller-Ladner, U., Pap, T., Gay, R. et al. Mechanisms of Disease: the molecular and cellular basis of joint destruction in rheumatoid arthritis. Nat Rev Rheumatol 1, 102–110 (2005).
Zwerina J, Redlich K, Polzer K, Joosten L, Kronke G, Distler J et al (2007) TNF-induced structural joint damage is mediated by IL-1. Proc Natl Acad Sci U S A 104(28):11742–11747.
Mahmoud DE, Kaabachi W, Sassi N, Tarhouni L, Rekik S, Jemmali S, Sehli H, Kallel-Sellami M, Cheour E, Laadhar L. The synovial fluid fibroblast-like synoviocyte: A long-neglected piece in the puzzle of rheumatoid arthritis pathogenesis. Front Immunol. 2022 Aug;13: 942417.
Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami M, Kotake S et al (2000) Tumor necrosis factor alpha stimulates osteoclast differentiation by a mechanism independent of the ODF/RANKL-RANK interaction. J Exp Med 191(2):275–286.
Korb-Pap A, Stratis A, Muhlenberg K, Niederreiter B, Hayer S, Echtermeyer F et al (2012) Early structural changes in cartilage and bone are required for the attachment and invasion of inflamed synovial tissue during destructive inflammatory arthritis. Ann Rheum Dis 71(6):1004–1011.
Goldring MB, Marcu KB (2009) Cartilage homeostasis in health and rheumatic diseases. Arthritis Res Ther 11(3):224.
Zhu J, Paul WE (2008) CD4 T cells: fates, functions, and faults. Blood 112(5):1557–1569.
Ogura H, Murakami M, Okuyama Y, Tsuruoka M, Kitabayashi C, Kanamoto M et al (2008) Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction. Immunity 29(4):628–636.
Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S et al (1999) Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature 402(6759):304–309.
Tu J, Zhang Y, Kim S, Wiebe E, Spies CM, Buttgereit F, Cooper MS, Seibel MJ, Zhou H. Transgenic Disruption of Glucocorticoid Signaling in Osteoblasts Attenuates Joint Inflammation in Collagen Antibody-Induced Arthritis. Am J Pathol. 2016 May;186(5):1293-301.
Bullock J, Rizvi SAA, Saleh AM, Ahmed SS, Do DP, Ansari RA, Ahmed J. Rheumatoid Arthritis: A Brief Overview of the Treatment. Med Princ Pract. 2018;27(6):501-507.
Ong CK, Lirk P, Tan CH, Seymour RA. An evidence-based update on nonsteroidal anti-inflammatory drugs. Clin Med Res. 2007 Mar;5((1)):19–34.
Silva JC, Mariz HA, Rocha LF, Jr, Oliveira PS, Dantas AT, Duarte AL, et al. Hydroxychloroquine decreases Th17-related cytokines in systemic lupus erythematosus and rheumatoid arthritis patients. Clinics (São Paulo) 2013 Jun;68((6)):766–71.
Gay RD, Clarke AW, Elgundi Z, Domagala T, Simpson RJ, Le NB, et al. Anti-TNFα domain antibody construct CEP-37247: full antibody functionality at half the size. MAbs. 2010 Nov-Dec;2((6)):625–38.
Lis K, Kuzawińska O, Bałkowiec-Iskra E. Tumor necrosis factor inhibitors - state of knowledge. Arch Med Sci. 2014 Dec;10((6)):1175–85.
Feldmann M, Brennan FM, Williams RO, Woody JN, Maini RN: The transfer of a laboratory based hypothesis to a clinically useful therapy: the development of anti-TNF therapy of rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2004, 18: 59-80. 10.1016/j.berh.2003.09.010.
Arend WP, Dayer JM: Inhibition of the production and effects of interleukin-1 and tumor necrosis factor alpha in rheumatoid arthritis. Arthritis Rheum. 1995, 38: 151-160.
Karonitsch T, von Dalwigk K, Steiner CW, Bluml S, Steiner G, Kiener HP et al (2012) Interferon signals and monocytic sensitization of the interferon-gamma signaling pathway in the peripheral blood of patients with rheumatoid arthritis. Arthritis Rheum 64(2):400–408.
Axmann R, Bohm C, Kronke G, Zwerina J, Smolen J, Schett G (2009) Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum 60(9):2747–2756.
Harrington R, Al Nokhatha SA, Conway R. JAK Inhibitors in Rheumatoid Arthritis: An Evidence-Based Review on the Emerging Clinical Data. J Inflamm Res. 2020 Sep 14;13: 519-531.
Rosman Z, Shoenfeld Y, Zandman-Goddard G. Biologic therapy for autoimmune diseases: an update. BMC Med. 2013 Apr;11((1)):88.
Chung KC, Pushman AG. Current concepts in the management of the rheumatoid hand. J Hand Surg Am. 2011 Apr;36((4)):736–47.
Ruderman EM, Pope RM. The evolving clinical profile of abatacept (CTLA4-Ig): a novel co-stimulatory modulator for the treatment of rheumatoid arthritis. Arthritis Res Ther. 2005;7 Suppl 2(Suppl 2): S21-5.
Knut L. Radiosynovectomy in the therapeutic management of arthritis. World J Nucl Med. 2015 Jan-Apr;14((1)):10–5.
Shiel WC., Jr Rheumatoid Arthritis. 2017 (accesses May 12, 2017) http://www.medicinenet.com/rheumatoid_arthritis/article.htm.
Smilek DE, Ehlers MR, Nepom GT. Restoring the balance: immunotherapeutic combinations for autoimmune disease. Dis Model Mech. 2014 May;7(5):503-13.
Mann DL. Innate immunity and the failing heart: the cytokine hypothesis revisited. Circ Res. 2015 Mar;116((7)):1254–68.
Perpétuo IP, Caetano-Lopes J, Rodrigues AM, Campanilho-Marques R, Ponte C, Canhão H, et al. Effect of Tumor Necrosis Factor Inhibitor Therapy on Osteoclasts Precursors in Rheumatoid Arthritis. BioMed Res Int. 2017; 2017:2690402.
Curtis JR, Singh JA. Use of biologics in rheumatoid arthritis: current and emerging paradigms of care. Clin Ther. 2011 Jun;33((6)):679–707.
Reily C, Stewart TJ, Renfrow MB, Novak J. Glycosylation in health and disease. Nat Rev Nephrol. 2019 Jun;15(6):346-366.
Marth JD, Grewal PK. Mammalian glycosylation in immunity. Nat Rev Immunol. 2008.
Pandey VK, Sharma R, Prajapati GK, Mohanta TK, Mishra AK. N-glycosylation, a leading role in viral infection and immunity development. Mol Biol Rep. 2022 Aug;49(8):8109-8120.
Esmail S, Manolson MF. Advances in understanding N-glycosylation structure, function, and regulation in health and disease. Eur J Cell Biol. 2021 Sep-Nov;100(7-8):151186.
Magalhães A, Duarte HO, Reis CA. The role of O-glycosylation in human disease. Mol Aspects Med. 2021 Jun; 79:100964.
González-Morelo KJ, Vega-Sagardía M, Garrido D. Molecular Insights Into O-Linked Glycan Utilization by Gut Microbes. Front Microbiol. 2020 Nov 5; 11: 591568.
Breloy I, Hanisch FG. Functional Roles of O-Glycosylation. Molecules. 2018 Nov 23;23(12):3063.
Radovani B, Gudelj I. N-Glycosylation and Inflammation; the Not-So-Sweet Relation. Front Immunol. 2022 Jun 27; 13:893365.
Jellusova J, Nitschke L. Regulation of B cell functions by the sialic acid-binding receptors siglec-G and CD22. Front Immunol. 2012 Jan 11; 2:96.
Müller J, Obermeier I, Wöhner M, Brandl C, Mrotzek S, Angermüller S, Maity PC, Reth M, Nitschke L. CD22 ligand-binding and signaling domains reciprocally regulate B-cell Ca2+ signaling. Proc Natl Acad Sci U S A. 2013 Jul 23;110(30):12402-7.
Clark EA, Giltiay NV. CD22: A Regulator of Innate and Adaptive B Cell Responses and Autoimmunity. Front Immunol. 2018 Sep 28; 9:2235.
O'Keefe TL, Williams GT, Batista FD, Neuberger MS. Deficiency in CD22, a B cell-specific inhibitory receptor, is sufficient to predispose to development of high affinity autoantibodies. J Exp Med. 1999 Apr 19;189(8):1307-13.
Dörner T, Shock A, Goldenberg DM, Lipsky PE. The mechanistic impact of CD22 engagement with epratuzumab on B cell function: Implications for the treatment of systemic lupus erythematosus. Autoimmun Rev. 2015 Dec;14(12):1079-86.
Clark MC, Baum LG. T cells modulate glycans on CD43 and CD45 during development and activation, signal regulation, and survival. Ann N Y Acad Sci. 2012 Apr; 1253:58-67.
Seo W, Ziltener HJ. CD43 processing and nuclear translocation of CD43 cytoplasmic tail are required for cell homeostasis. Blood. 2009 Oct 22;114(17):3567-77.
Coughlin S, Noviski M, Mueller JL, Chuwonpad A, Raschke WC, Weiss A, Zikherman J. An extracatalytic function of CD45 in B cells is mediated by CD22. Proc Natl Acad Sci U S A. 2015 Nov 24;112(47): E6515-24.
Grabowska J, Lopez-Venegas MA, Affandi AJ, den Haan JMM. CD169+ Macrophages Capture and Dendritic Cells Instruct: The Interplay of the Gatekeeper and the General of the Immune System. Front Immunol. 2018 Oct 26; 9:2472.
Friedrich SK, Lang PA, Friebus-Kardash J, Duhan V, Bezgovsek J, Lang KS. Mechanisms of lymphatic system-specific viral replication and its potential role in autoimmune disease. Clin Exp Immunol. 2019 Jan;195(1):64-73.
Nguyen JT, Evans DP, Galvan M, Pace KE, Leitenberg D, Bui TN, Baum LG. CD45 modulates galectin-1-induced T cell death: regulation by expression of core 2 O-glycans. J Immunol. 2001 Nov 15;167(10):5697-707.
Zhou X, Motta F, Selmi C, Ridgway WM, Gershwin ME, Zhang W. Antibody glycosylation in autoimmune diseases. Autoimmun Rev. 2021 May;20(5):102804.
Morel M, Pochard P, Echchih W, Dueymes M, Bagacean C, Jousse-Joulin S, Devauchelle-Pensec V, Cornec D, Jamin C, Pers JO, Bordron A. Abnormal B cell glycosylation in autoimmunity: A new potential treatment strategy. Front Immunol. 2022 Aug 25; 13:975963.
Zhang Z, Shah B, Richardson J. Impact of Fc N-glycan sialylation on IgG structure. MAbs. 2019 Nov-Dec;11(8):1381-1390.
Nandakumar KS, Holmdahl R. Antibody-induced arthritis: disease mechanisms and genes involved at the effector phase of arthritis. Arthritis Res Ther. 2006;8(6):223.
Kagari T, Doi H, Shimozato T: The importance of IL-1 beta and TNF-alpha, and the noninvolvement of IL-6, in the development of monoclonal antibody-induced arthritis. J Immunol. 2002, 169: 1459-1466.
Williams RO. Collagen-induced arthritis as a model for rheumatoid arthritis. Methods Mol Med. 2004; 98:207-16.
Nandakumar KS, Svensson L, Holmdahl R. Collagen type II-specific monoclonal antibody-induced arthritis in mice: description of the disease and the influence of age, sex, and genes. Am J Pathol. 2003 Nov;163(5): 1827-37.
Cho YG, Cho ML, Min SY, Kim HY. Type II collagen autoimmunity in a mouse model of human rheumatoid arthritis. Autoimmun Rev. 2007 Nov;7(1):65-70.
Luross JA, Williams NA. The genetic and immunopathological processes underlying collagen-induced arthritis. Immunology. 2001 Aug;103(4):407-16.
Paul Monach, Kimie Hattori, Haochu Huang, Elzbieta Hyatt, Jody Morse, Linh Nguyen, Adriana Ortiz-Lopez, Hsin-Jung Wu, Diane Mathis, Christophe Benoist. The K/BxN Mouse Model of Inflammatory Arthritis. Arthritis Research, 2007, Volume 136.
Christensen AD, Haase C, Cook AD, Hamilton JA. K/BxN Serum-Transfer Arthritis as a Model for Human Inflammatory Arthritis. Front Immunol. 2016 Jun 2; 7:213.
Kissel T, Toes REM, Huizinga TWJ, Wuhrer M. Glycobiology of rheumatic diseases. Nat Rev Rheumatol. 2023 Jan;19(1):28-43. doi: 10.1038/s41584-022-00867-4. Epub 2022 Nov 23. Erratum in: Nat Rev Rheumatol. 2023 Apr;19(4):253.
Maleitzke T, Weber J, Hildebrandt A, Dietrich T, Zhou S, Tsitsilonis S, Keller J. Standardized protocol and outcome measurements for the collagen antibody-induced arthritis mouse model. STAR Protoc. 2022 Dec 16; 3(4): 101718.
Williams B, Lees F, Tsangari H, Hutchinson MR, Perilli E, Crotti TN. Effects of Mild and Moderate Monoclonal Antibody Dose on Inflammation, Bone Loss, and Activation of the Central Nervous System in a Female Collagen Antibody-induced Arthritis Mouse Model. J Histochem Cytochem. 2021 Aug;69(8):511-522.
Monach PA, Mathis D, Benoist C. The K/BxN arthritis model. Curr Protoc Immunol. 2008 May; Chapter 15:15.22.1-15.22.12.
Montilla-García Á, Tejada MÁ, Perazzoli G, Entrena JM, Portillo-Salido E, Fernández-Segura E, Cañizares FJ, Cobos EJ. Grip strength in mice with joint inflammation: A rheumatology function test sensitive to pain and analgesia. Neuropharmacology. 2017 Oct; 125:231-242.
Hayer, S., Vervoordeldonk, M.J., Denis, M.C., Armaka, M., Hoffmann, M.H., Bäcklund, J., Nandakumar, K., Niederreiter, B., Geka, C., Fischer, A., Woodworth, N., Blüml, S., Kollias, G., Holmdahl, R., Apparailly, F., & Koenders, M.I. (2021). ‘SMASH’ recommendations for standardised microscopic arthritis scoring of histological sections from inflammatory arthritis animal models. Annals of the Rheumatic Diseases, 80, 714 - 726.
Hayman AR. Tartrate-resistant acid phosphatase (TRAP) and the osteoclast/immune cell dichotomy. Autoimmunity. 2008 Apr;41(3):218-23.
Raphael I, Joern RR, Forsthuber TG. Memory CD4+ T Cells in Immunity and Autoimmune Diseases. Cells. 2020 Feb 25;9(3):531.
Yang W, Yu T, Cong Y. CD4+ T cell metabolism, gut microbiota, and autoimmune diseases: implication in precision medicine of autoimmune diseases. Precis Clin Med. 2022 Jul 6;5(3): pbac018.
Jacobs BY, Kloefkorn HE, Allen KD. Gait analysis methods for rodent models of osteoarthritis. Current Pain and Headache Reports. 2014;18(456):1–11.
Ferland CE, Laverty S, Beaudry F, Vachon P. Gait analysis and pain response of two rodent models of osteoarthritis. Pharmacology Biochemistry and Behavior. 2011;97(2011):603–610.
Lakes EH, Allen KD. Gait analysis methods for rodent models of arthritic disorders: reviews and recommendations. Osteoarthritis Cartilage. 2016 Nov;24(11):1837-1849.
Boettger MK, Weber K, Schmidt M, Gajda M, Bräuer R, Schaible HG. Gait abnormalities differentially indicate pain or structural joint damage in monoarticular antigen-induced arthritis. Pain. 2009;145(2009):142–150.
Pereira MS, Alves I, Vicente M, Campar A, Silva MC, Padrão NA, Pinto V, Fernandes Â, Dias AM, Pinho SS. Glycans as Key Checkpoints of T Cell Activity and Function. Front Immunol. 2018 Nov 27; 9:2754.
Lowe JB. Glycosylation, immunity, and autoimmunity. Cell. 2001 Mar 23;104(6):809-12.
Alves I, Fernandes Â, Santos-Pereira B, Azevedo CM, Pinho SS. Glycans as a key factor in self and nonself discrimination: impact on the breach of immune tolerance. FEBS Lett. 2022 Jun;596(12):1485-1502.
Earl LA, Bi S, Baum LG. N- and O-glycans modulate galectin-1 binding, CD45 signaling, and T cell death. J Biol Chem. 2010 Jan 22;285(4):2232-44.
Hobbs SJ, Nolz JC. Regulation of T Cell Trafficking by Enzymatic Synthesis of O-Glycans. Front Immunol. 2017 May 24; 8:600.
van Kooyk Y, Rabinovich GA. Protein-glycan interactions in the control of innate and adaptive immune responses. Nat Immunol. 2008 Jun;9(6):593-601.
van Gaalen FA, Toes RE, Ditzel HJ, Schaller M, Breedveld FC, Verweij CL, Huizinga TW. Association of autoantibodies to glucose-6-phosphate isomerase with extraarticular complications in rheumatoid arthritis. Arthritis Rheum. 2004 Feb;50(2):395-9.
Schaller M, Stohl W, Benoit V, Tan SM, Johansen L, Ditzel HJ. Patients with inflammatory arthritic diseases harbor elevated serum and synovial fluid levels of free and immune-complexed glucose-6-phosphate isomerase (G6PI). Biochem Biophys Res Commun. 2006 Oct 20;349(2):838-45.
Xiong G, Lei T, Dong S, Xu L, Li M, Wang R. Roles of CD3, CD4 and CD8 in synovial lymphocytes of rheumatoid arthritis. Pol J Pathol. 2022;73(1):21-26.
-
dc.identifier.urihttp://tdr.lib.ntu.edu.tw/jspui/handle/123456789/90440-
dc.description.abstract類風濕性關節炎(Rheumatoid arthritis, RA)是一種慢性自體免疫疾病,由關節滑膜的慢性發炎所引起。RA的發病機制涉及T細胞的活化和促發炎因子的分泌,如GM-CSF、IL-17A、IL-17F和IL-23等,這些促發炎因子促進Th17細胞分化和增值,導致軟骨侵蝕、硬骨侵蝕以及免疫細胞浸潤,最終導致關節結構受損。近年研究發現異常的醣基化與自體免疫疾病的誘發有關,C1GALT1 (Core 1 beta1,3-galactosyltransferase, Core 1 synthase) 是使氧型醣基化中的醣類結構產生分支和複雜度的關鍵酵素。然而,C1GALT1在RA發病機制中的調控機制和影響仍不清楚。本研究旨在探討C1GALT1在RA發病機制中的作用以及對T細胞分化的影響。首先,我們使用免疫組織化學染色法分析了膠原抗體誘導性關節炎(Collagen Antibody-Induced Arthritis, CAIA)小鼠脾臟中C1GALT1的蛋白表達,結果顯示C1GALT1在發炎反應中表達增加。而為了進一步探討C1GALT1對T細胞分化的影響,我們繁殖出了C1GALT1f/f CD4-Cre (C1GALT1 KO)小鼠,其C1GALT1基因在naïve T細胞分化為CD4+ T細胞的過程中被敲除,從而影響CD4+ T 細胞表面醣類結構的修飾。流式細胞儀分析C1GALT1f/f和C1GALT1 KO小鼠的脾臟、胸腺和淋巴結,發現敲除C1GALT1基因後VVA lectin(Vicia Villosa Lectin)的表達增加,表明CD4+ T細胞表面醣類結構發生了改變。此外,在小鼠誘導關節炎的實驗中發現,CD4+ T細胞特異性敲除C1GALT1改善了小鼠的踝關節腫脹、爪力強度以及臨床評分。並在骨表面掃描結果顯示C1GALT1 KO小鼠的腳掌骨表面骨侵蝕減緩。這些研究結果表明,在CAIA關節炎模型中,CD4+ T細胞特異性敲除C1GALT1可能減輕關節發炎所引起的踝關節腫脹,說明C1GALT1或許具有抑制發炎的潛力。瞭解C1GALT1在RA發病機制和T細胞分化中的作用,可以有助於提供醣基化治療RA和其他自體免疫疾病的方法。zh_TW
dc.description.abstractRheumatoid arthritis (RA) is a chronic autoimmune disease characterized by chronic inflammation targeting synovial membrane in the joints, resulting in arthritis. The pathogenesis of RA involves T cell activation and the secretion of proinflammatory cytokines, such as GM-CSF, IL-17A, IL-17F, and IL-23. These proinflammatory cytokines promote T cell differentiation and proliferation, leading to cartilage and bone erosion, and immune cell infiltration, which ultimately result in damage to joint structures. Abnormal glycosylation has also been implicated in autoimmune diseases, with the glycosyltransferase C1GALT1 playing a critical role in O-glycosylation and glycan complexity. However, the regulatory mechanisms and impact of C1GALT1 on RA pathogenesis remain unclear. This study aimed to investigate the role of C1GALT1 in RA pathogenesis and its effect on T differentiation. Initially, we analyzed the protein expression of C1GALT1 in the spleens of Collagen Antibody-Induced Arthritis (CAIA) induced C57BL/6 mice using immunohistochemistry, revealing increased C1GALT1 expression in response to inflammation. To explore the influence of C1GALT1 on Th17 differentiation, we generated C1GALT1f/f CD4-Cre mice (C1GALT1 KO), in which the C1GALT1 gene was knocked out during the differentiation of naïve T cells into CD4+ T cells, impacting surface glycan modification. Flow cytometry analysis of spleens, thymus, and lymph nodes from C1GALT1f/f and C1GALT1f/f CD4-Cre mice showed increased lectin expression of VVA following C1GALT1 gene knockout, indicating altered O-glycans on the surface of CD4+ T cells. Furthermore, we employed the CAIA model to simulate RA in mice and compared C1GALT1f/f CD4-Cre mice to C1GALT1f/f mice. The results demonstrated that C1GALT1 knockout in T cells alleviated ankle swelling in the CAIA model, with reduced swelling observed from Day 6 to Day 10. Additionally, bone scan analysis revealed a deceleration of bone erosion on the paw bone surface in C1GALT1 KO. These findings suggest that T cell-specific knockout of C1GALT1 may mitigate ankle swelling in the CAIA model, emphasizing the crucial role of C1GALT1 in suppressing inflammation. Understanding the role of C1GALT1 in RA pathogenesis and T cell differentiation could provide insights into potential therapeutic strategies targeting glycosylation pathways for the treatment of RA and other autoimmune diseases.en
dc.description.provenanceSubmitted by admin ntu (admin@lib.ntu.edu.tw) on 2023-10-02T16:13:38Z
No. of bitstreams: 0
en
dc.description.provenanceMade available in DSpace on 2023-10-02T16:13:38Z (GMT). No. of bitstreams: 0en
dc.description.tableofcontents口試委員會審定書 ⅰ
致謝 ⅰi
中文摘要 ⅰii
Abstract iv
Abbreviation v
Chapter 1 Introductions 1
1.1 Autoimmune disease 1
1.1.1 The definition of autoimmune diseases 1
1.1.2 Rheumatoid arthritis 1
1.1.3 The prevalence and diagnostic methods of rheumatoid arthritis 2
1.1.4 The mechanisms of rheumatoid arthritis 2
1.1.5 Treatment strategies and challenges in rheumatoid arthritis 4
1.2 Glycosylation 7
1.2.1 The definition and biological functions of glycosylation 7
1.2.2 The type of glycosylation 7
1.2.3 Glycosylation and immunity 9
1.3 Mouse models that mimic the pathogenesis of human RA 11
1.3.1 Collagen Antibody-Induced Arthritis (CAIA) model 11
1.3.2 K/BxN serum-transfer arthritis (STA) model 12
1.4 Research value of C1GALT1 in autoimmune diseases 13
Chapter 2 Study objectives and Hypotheses 15
Chapter 3 Materials and Methods 18
3.1 Materials 18
3.1.1 Antibody 18
3.1.2 Primers 19
3.1.3 Chemical and Reagents 19
3.1.3.1 Buffer Solutions and Reagents 22
3.1.4 Instruments and Software 26
3.2 Methods 28
3.2.1 Mice primary cells isolation 28
3.2.2 Animal Handling 28
3.2.3 Tissue Decalcification 29
3.2.4 Immunohistochemistry staining 29
3.2.5 Tartrate resistant acid phosphatase staining (TRAP) 30
3.2.6 Toluidine Blue Staining 30
3.2.7 Flow cytometry 31
3.2.8 Bradford protein assay 31
3.2.9 Western blot 32
3.2.10 Collagen Antibody-Induced Arthritis model (CAIA) 32
3.2.11 CAIA clinical arthritis score 33
3.2.12 K/BxN Serum-Transfer Arthritis model (K/BxN STA) 33
3.2.13 K/BxN STA clinical arthritis score 33
3.2.14 Malleoli size measurements 34
3.2.15 Grip strength measurements 34
3.2.16 Toluidine blue staining histopathological scoring 35
3.2.17 H&E staining histopathological scoring 35
3.2.18 TRAP-positive osteoclasts counting 36
3.2.19 Trajectory analysis 36
3.2.20 Bone surface erosion by micro CT analysis 36
3.2.21 Mouse TNF-α Enzyme-linked immunosorbent assay 37
3.2.22 Statistical analysis 37
Chapter 4 Result 38
4.1 C1GALT1 increase during collagen antibody-induced arthritis mouse model 38
4.2 Generation of C1GALT1 conditional knockdout mice for study the mechanisms of RA 38
4.2.1 The impact of knockout C1GALT1 in mouse thymus on immune organs 38
4.2.2 The impact of knockout C1GALT1 in mouse thymus on cell surface glycan structures 39
4.2.3 The impact of knockout C1GALT1 on the population of CD4+ T cells, CD8+ T cells, and B cells in mouse immune organs 39
4.3 C1GALT1f/f CD4+Cre mice alleviate the clinical index in CAIA 41
4.3.1 C1GALT1f/f CD4+Cre mice showed improvement in weight loss during CAIA 41
4.3.2 C1GALT1f/f CD4+Cre mice showed alleviated ankle swelling during CAIA 41
4.3.3 C1GALT1f/f CD4+Cre mice alleviated the functional decline in joint inflammation induced by CAIA 42
4.3.4 C1GALT1f/f CD4+Cre mice exhibited reduced paw swelling scores during CAIA 42
4.3.5 C1GALT1f/f CD4+Cre mice did not exhibit any improvement in walking distance and velocity of the mice in CAIA 43
4.4 C1GALT1f/f CD4+Cre mice alleviate the histopathological scoring in CAIA 43
4.4.1 C1GALT1f/f CD4+Cre mice exhibited amelioration of cartilage erosion and proteoglycan loss in the CAIA model 43
4.4.2 C1GALT1f/f CD4+Cre mice exhibited amelioration of bone erosion in the CAIA model 44
4.4.3 C1GALT1f/f CD4+Cre mice showed reduced inflammation activity and synovial lining hyperplasia in the CAIA model 45
4.5 The level of TNF-α in the serum of C1GALT1f/f CD4+Cre mice is decreased 45
4.6 The population of CD3+ T cells, CD4+ T cells and CD8+ T cells are decreased in the spleen and lymph nodes of C1GALT1f/f CD4+Cre mice 46
4.7 C1GALT1 increase during K/BxN Serum-Transfer Arthritis model 47
4.8 C1GALT1f/f CD4+Cre mice alleviate the clinical index in K/BxN STA 47
4.8.1 C1GALT1f/f CD4+Cre mice improvement in weight loss during K/BxN STA 47
4.8.2 C1GALT1f/f CD4+Cre mice alleviated ankle swelling during K/BxN STA 48
4.8.3 C1GALT1f/f CD4+Cre mice alleviated the functional decline in joint inflammation induced by K/BxN STA 48
4.8.4 C1GALT1f/f CD4+Cre mice exhibited reduced paw swelling scores during K/BxN STA 49
4.9 C1GALT1f/f CD4+Cre mice alleviate the histopathological scoring in K/BxN STA 49
4.9.1 C1GALT1f/f CD4+Cre mice exhibited amelioration of cartilage erosion and proteoglycan loss in the K/BxN STA model 49
4.9.2 C1GALT1f/f CD4+Cre mice exhibited amelioration of bone erosion in the K/BxN STA model 50
4.9.3 C1GALT1f/f CD4+Cre mice reduced inflammation activity and synovial lining hyperplasia in the K/BxN STA model 51
Chapter 5 Conclusion 53
Chapter 6 Discussion 54
6.1 Knockout of C1GALT1 did not improve walking distance and velocity in arthritic mice 54
6.2 Knockout of C1GALT1 in CD4+ T cell resulted in a decreased population of CD3+ T cells, CD4+ T cells and CD8+ T cells in the spleen and lymph nodes 54
References 56
List of Tables 65
List of Figures 67
-
dc.language.isoen-
dc.title探討敲除CD4+ T細胞中C1GALT1對類風濕性關節炎小鼠模型的治療效果zh_TW
dc.titleTo investigate the therapeutic effect of deleting C1GALT1 in CD4+ T cells in a rheumatoid arthritis mouse modelen
dc.typeThesis-
dc.date.schoolyear111-2-
dc.description.degree碩士-
dc.contributor.coadvisor黃敏銓zh_TW
dc.contributor.coadvisorMin-Chuan Huangen
dc.contributor.oralexamcommittee郭靜穎;莊雅婷zh_TW
dc.contributor.oralexamcommitteeChing-Ying Kuo;Ya-Ting Chuangen
dc.subject.keywordCore 1 synthase,CD4+ T cell,膠原抗體誘導性關節炎模型,T 細胞分化,zh_TW
dc.subject.keywordCore 1 synthase,CD4+ T cell,Collagen Antibody-Induced Arthritis,en
dc.relation.page94-
dc.identifier.doi10.6342/NTU202303703-
dc.rights.note未授權-
dc.date.accepted2023-08-10-
dc.contributor.author-college醫學院-
dc.contributor.author-dept解剖學暨細胞生物學研究所-
顯示於系所單位:解剖學暨細胞生物學科所

文件中的檔案:
檔案 大小格式 
ntu-111-2.pdf
  目前未授權公開取用
7.59 MBAdobe PDF
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