簡易檢索 / 詳目顯示

研究生: 曹予馨
Tsao, Yu-Hsin
論文名稱: 紫蘇葉萃取物的治療潛力之研究
Study on Therapeutic Potential of Perilla Leaf Extract
指導教授: 李家惠
Lee, Chia-Huei
王翊青
Wang, I-Ching
口試委員: 洪錦堂
Horng, Jim-Tong
邱慶豐
Chiu, Ching-Feng
學位類別: 碩士
Master
系所名稱: 生命科學暨醫學院 - 生物科技研究所
Biotechnology
論文出版年: 2024
畢業學年度: 112
語文別: 中文
論文頁數: 35
中文關鍵詞: 紫蘇葉口腔鱗狀細胞癌咖啡酸迷迭香酸介白素-1β表皮生長因子接受器艾瑞莎順鉑
外文關鍵詞: perilla, oral squamous cell carcinoma (OSCC), caffeic acid, rosmarinic acid, interleukin (IL)-1β, epidermal growth factor receptor (EGFR), gefitinib, cisplatin
相關次數: 點閱:33下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 紫蘇葉萃取物已被證實具有多種藥理特性。研究結果顯示紫蘇葉萃取物能抑制口腔鱗狀細胞癌(以下簡稱為口腔癌)的細胞生長,且其生長抑制力正比於口腔癌細胞之惡性。經由高效液相層析分析法得知咖啡酸和迷迭香酸是紫蘇葉萃取物中主要的酚類成分,對口腔癌細胞TW2.6的72小時之半抑制濃度分別是362.3 μM和173.7 μM;對於OC3之半抑制濃度分別為477 μM和280.8 μM。咖啡酸和迷迭香酸混合能夠劑量依賴性地抑制口腔細胞株OC3與TW2.6的生長。當兩者總濃度介於300 μM‒500 μM區間時,咖啡酸和迷迭香酸以1:2比例混合有最佳抑制力,然而兩者並無協同作用。酵素連結免疫吸附分析法結果顯示咖啡酸、迷迭香酸或兩者混合均能有效抑制OC3細胞原發及腫瘤壞死因子-α激發後介白素-1β的生成。西方墨點法分析結果顯示OC3細胞生長於含有咖啡酸、迷迭香酸或兩者並存的培養液中,會增強OC3細胞本身及受腫瘤壞死因子-α激發後表皮生長因子接受器的活化,同時提高OC3細胞對低劑量 (5 μM) 表皮生長因子接受器酪氨酸激酶抑製劑艾瑞莎的敏感度。此外,集落形成實驗結果顯示口腔癌細胞株長期培養於含有迷迭香酸、咖啡酸與迷迭香酸混合物的培養液中可有效提高低劑量(1 μM)順鉑對其生長的抑制力。此外,流式細胞儀分析結果顯示口腔癌細胞OC3以咖啡酸(100 μM)、迷迭香酸(200 μM)或兩者混合處理72小時可誘導細胞凋亡。
    綜合以上實驗結果,我們認為紫蘇葉中的咖啡酸和迷迭香酸可以作為口腔癌艾瑞莎標靶治療及順鉑化療的輔劑,有效提高口腔癌細胞對艾瑞莎和順鉑的敏感度。此研究成果不僅是紫蘇葉酚類成分藥效的新發現,也具有臨床應用潛力。


    The herbal medicine perilla leaf extract (PLE) has various pharmacological properties. Our study showed that PLE inhibits the proliferation of oral squamous cell carcinoma (OSCC) cells and the proliferation inhibitory effect correlates with the malignancy of OSCC. High performance liquid chromatography analysis revealed that caffeic acid (CA) and rosmarinic acid (RA) constitutes the two main components of perilla phenols. After treatment of 72 h, CA and RA inhibits the viability of OSCC cells in a dose-dependent manner. The IC50 value for CA and RA in inhibiting the viability of TW2.6 cells were 362.3 μM and 173.7 μM, respectively. The IC50 value for CA and RA in inhibiting the viability of OC3 cells were 477 μM and 280.8 μM, respectively. The viability of TW2.6 and OC3 cells also suppressed by the mixture of CA and RA. The optimal CA/RA combination ratio was 1:2 at concentrations of 300–500 μM but had no synergistic effect on inhibiting the viability of OSCC cells. Results from enzyme-linked immunosorbent assay showed that CA, RA, or CA/RA mixture significantly suppressed OC3 cells to production of interleukin-1β (IL-1β) in the presence or absence of Tumor Necrosis Factor-α (TNF-α) stimulation. Western blot analysis indicated that treatment of CA or the combination of CA and RA enhanced the activation of EGFR in OC3 cells in the presence or absence of TNF-α stimulation. Moreover, treatment with CA or the combination of CA and RA also increased the sensitivity of OC3 cells to low-dose (5 μM) EGFR tyrosine kinase inhibitor gefitinib. Colony forming experiments showed that RA or the combination of CA and RA enhanced the growth-inhibitory effect of low-dose (1 μM) cisplatin. Furthermore, flow cytometry analysis indicated that CA (100 μM), RA (200 μM), or the combination of CA and RA induced apoptosis of OC3 cells after 72 h‒incubation.
    In summary, these results suggest that the perilla phenols, CA and RA, can serve as adjuvants in gefitinib targeted therapy and cisplatin chemotherapy of OSCC, to enhance the sensitivity of OSCC cells to gefitinib and cisplatin. Findings from this study not only revealed a novel pharmaceutical effect of perilla phenols but also uncovered the potential of perilla phenols for clinical application.

    中文摘要-ii Abstract-iii Contents-iv Chapter 1. Introduction-1 Chapter 2. Materials and Methods-4 1.Cell culture-4 2.Preparation of PLC-4 3.Cell viability assay, thiazolyl blue tetrazolium bromide (MTT)-5 4.High performance liquid chromatography (HPLC) analysis of PLE-5 5.Western blot analysis-6 6.Determination of IL-1β levels in culture media-7 7.Colony forming assay-7 8.Apoptosis assay-8 9.Quantitative real-time PCR (qPCR)-8 Chapter 3. Results-10 1.PLE selectively suppressed viability of OSCC cells-10 2.Identification of phenols in PLE-11 3.Antiproliferation effects of CA and RA on OSCC cells-11 4.CA and RA alone or in combination suppressed the release of IL-1β from TNF-α-induced or non-induced OC3 cells-12 5.CA and RA alone or in combination induced EGFR activation and enhanced antitumor activity of low-dose gefitinib-13 6.Chronic CA and RA treatment enhanced the cytotoxicity of low-dose cisplatin in TW2.6 cells-15 7.CA and RA alone or in combination induced apoptosis in OC3 cells in a time-dependent manner-16 Chapter 4. Discussion-17 Chapter 5. Conclusion-23 Reference-24 Figures-27

    1. Hou, T., et al., Perilla frutescens: A Rich Source of Pharmacological Active Compounds. Molecules, 2022. 27(11).
    2. Adam, G., et al., Applications of Perilla frutescens Extracts in Clinical Practice. Antioxidants (Basel), 2023. 12(3).
    3. Kim, D.S., et al., Inhibitory effects of rosmarinic acid on adriamycin-induced apoptosis in H9c2 cardiac muscle cells by inhibiting reactive oxygen species and the activations of c-Jun N-terminal kinase and extracellular signal-regulated kinase. Biochem Pharmacol, 2005. 70(7): p. 1066-78.
    4. Ragazinskiene, O., et al., [Common perilla (Perilla frutescens (L.) Britton.) as a perspective immunomodulator]. Medicina (Kaunas), 2004. 40(3): p. 220-4.
    5. Lee, C.H., et al., IL-1beta promotes malignant transformation and tumor aggressiveness in oral cancer. J Cell Physiol, 2015. 230(4): p. 875-84.
    6. Lee, C.H., et al., LDOC1 Suppresses Microbe-Induced Production of IL-1beta in Human Normal and Cancerous Oral Cells through the PI3K/Akt/GSK-3beta Axis. Cancers (Basel), 2020. 12(11).
    7. Shi, J., et al., Impact of inflammation and immunotherapy in renal cell carcinoma. Oncol Lett, 2020. 20(5): p. 272.
    8. Schmitt, M. and F.R. Greten, The inflammatory pathogenesis of colorectal cancer. Nat Rev Immunol, 2021. 21(10): p. 653-667.
    9. Tang, W.F., et al., Perilla (Perilla frutescens) leaf extract inhibits SARS-CoV-2 via direct virus inactivation. Biomed J, 2021. 44(3): p. 293-303.
    10. Wang, Y., et al., Extract of Perilla frutescens inhibits tumor proliferation of HCC via PI3K/AKT signal pathway. Afr J Tradit Complement Altern Med, 2013. 10(2): p. 251-7.
    11. Abd El-Hafeez, A.A., et al., Synergistic tumor suppression by a Perilla frutescens-derived methoxyflavanone and anti-cancer tyrosine kinase inhibitors in A549 human lung adenocarcinoma. Cytotechnology, 2018. 70(3): p. 913-919.
    12. Kim, C.L., et al., Extracts of Perilla frutescens var. Acuta (Odash.) Kudo Leaves Have Antitumor Effects on Breast Cancer Cells by Suppressing YAP Activity. Evid Based Complement Alternat Med, 2021. 2021: p. 5619761.
    13. Rao, C.V., et al., Prevention of familial adenomatous polyp development in APC min mice and azoxymethane-induced colon carcinogenesis in F344 Rats by omega-3 fatty acid rich perilla oil. Nutr Cancer, 2013. 65 Suppl 1: p. 54-60.
    14. Tantipaiboonwong, P., et al., Molecular Mechanism of Antioxidant and Anti-Inflammatory Effects of Omega-3 Fatty Acids in Perilla Seed Oil and Rosmarinic Acid Rich Fraction Extracted from Perilla Seed Meal on TNF-alpha Induced A549 Lung Adenocarcinoma Cells. Molecules, 2021. 26(22).
    15. Pintha, K., et al., Suppressive Effects of Rosmarinic Acid Rich Fraction from Perilla on Oxidative Stress, Inflammation and Metastasis Ability in A549 Cells Exposed to PM via C-Jun, P-65-Nf-Kappab and Akt Signaling Pathways. Biomolecules, 2021. 11(8).
    16. Voronov, E., et al., IL-1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci U S A, 2003. 100(5): p. 2645-50.
    17. Apte, R.N., et al., Effects of micro-environment- and malignant cell-derived interleukin-1 in carcinogenesis, tumour invasiveness and tumour-host interactions. Eur J Cancer, 2006. 42(6): p. 751-9.
    18. Lee, C.H., et al., Interleukin-1 beta transactivates epidermal growth factor receptor via the CXCL1-CXCR2 axis in oral cancer. Oncotarget, 2015. 6(36): p. 38866-80.
    19. Petrella, B.L. and M.P. Vincenti, Interleukin-1beta mediates metalloproteinase-dependent renal cell carcinoma tumor cell invasion through the activation of CCAAT enhancer binding protein beta. Cancer Med, 2012. 1(1): p. 17-27.
    20. Lee, C.H., et al., MCP-1/MCPIP-1 Signaling Modulates the Effects of IL-1beta in Renal Cell Carcinoma through ER Stress-Mediated Apoptosis. Int J Mol Sci, 2019. 20(23).
    21. Lin, S.C., et al., Establishment of OC3 oral carcinoma cell line and identification of NF-kappa B activation responses to areca nut extract. J Oral Pathol Med, 2004. 33(2): p. 79-86.
    22. Rheinwald, J.G. and M.A. Beckett, Tumorigenic keratinocyte lines requiring anchorage and fibroblast support cultured from human squamous cell carcinomas. Cancer Res, 1981. 41(5): p. 1657-63.
    23. Guan, Z., et al., Identification and quantitation of phenolic compounds from the seed and pomace of Perilla frutescens using HPLC/PDA and HPLC-ESI/QTOF/MS/MS. Phytochem Anal, 2014. 25(6): p. 508-13.
    24. Ma, S., et al., Actin-like protein 8 promotes cell proliferation, colony-formation, proangiogenesis, migration and invasion in lung adenocarcinoma cells. Thorac Cancer, 2020. 11(3): p. 526-536.
    25. Chen, C.Y., et al., Anti-inflammatory effects of Perilla frutescens in activated human neutrophils through two independent pathways: Src family kinases and Calcium. Sci Rep, 2015. 5: p. 18204.
    26. Abotaleb, M., et al., Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules, 2020. 10(2).
    27. Mahmoud, M.A., et al., Rosmarinic acid suppresses inflammation, angiogenesis, and improves paclitaxel induced apoptosis in a breast cancer model via NF3 kappaB-p53-caspase-3 pathways modulation. J Appl Biomed, 2021. 19(4): p. 202-209.
    28. Wan, F., et al., Caffeic Acid Supplement Alleviates Colonic Inflammation and Oxidative Stress Potentially Through Improved Gut Microbiota Community in Mice. Front Microbiol, 2021. 12: p. 784211.
    29. Zhang, K., et al., ACTB and GAPDH appear at multiple SDS-PAGE positions, thus not suitable as reference genes for determining protein loading in techniques like Western blotting. Open Life Sci, 2021. 16(1): p. 1278-1292.
    30. Atanasov, A.G., et al., Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov, 2021. 20(3): p. 200-216.
    31. Huang, S., et al., The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules, 2023. 28(15).
    32. Zhang, M., et al., Caffeic acid reduces cutaneous tumor necrosis factor alpha (TNF-alpha), IL-6 and IL-1beta levels and ameliorates skin edema in acute and chronic model of cutaneous inflammation in mice. Biol Pharm Bull, 2014. 37(3): p. 347-54.
    33. Rocha, J., et al., Anti-inflammatory effect of rosmarinic acid and an extract of Rosmarinus officinalis in rat models of local and systemic inflammation. Basic Clin Pharmacol Toxicol, 2015. 116(5): p. 398-413.
    34. Schroder, K. and J. Tschopp, The inflammasomes. Cell, 2010. 140(6): p. 821-32.
    35. Lee, H.E., et al., Targeting ASC in NLRP3 inflammasome by caffeic acid phenethyl ester: a novel strategy to treat acute gout. Sci Rep, 2016. 6: p. 38622.
    36. Ketabat, F., et al., Controlled Drug Delivery Systems for Oral Cancer Treatment-Current Status and Future Perspectives. Pharmaceutics, 2019. 11(7).
    37. Lee, C.H., et al., Therapeutic Effects of Perilla Phenols in Oral Squamous Cell Carcinoma. Int J Mol Sci, 2023. 24(19).

    QR CODE