博碩士論文 110821014 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:28 、訪客IP:3.146.34.191
姓名 劉晏均(Yan-Jun Liu)  查詢紙本館藏   畢業系所 生命科學系
論文名稱 探討暴露聚苯乙烯塑膠微粒對小鼠大腦學習與記憶之分子機制與神經發炎的影響
(The effects of polystyrene microplastics exposure on molecular mechanisms of learning and memory and neuroinflammation)
相關論文
★ 探討剔除Dtnbp1基因對於公和母鼠前額葉多巴胺傳遞路徑與社交行為的影響★ 探討早期壓力及成年慢性不可預測壓力對恐懼社交轉移的影響
★ 探討食入及吸入聚苯乙烯塑膠微粒對小鼠行為的影響
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]   [檢視]  [下載]
  1. 本電子論文使用權限為同意立即開放。
  2. 已達開放權限電子全文僅授權使用者為學術研究之目的,進行個人非營利性質之檢索、閱讀、列印。
  3. 請遵守中華民國著作權法之相關規定,切勿任意重製、散佈、改作、轉貼、播送,以免觸法。

摘要(中) 塑膠微粒(MPs)定義為直徑小於5 mm的塑膠顆粒,是一種新興環境污染物。因為塑膠微粒體積較小,它們能夠經由食物鏈進入生物體內,並積聚在周圍組織/器官中,隨後對器官造成損害。本實驗室先前研究發現,聚苯乙烯塑膠微粒(PS-MPs)會影響公鼠學習與記憶能力,然而,PS-MPs如何影響大腦學習及記憶的分子機制仍然未知。本篇研究結果顯示,小鼠口服2μm PS-MPs八周後,利用拉曼光譜分析,可觀察到2 μm PS-MPs在肝臟及大腦海馬迴腦區中累積,而海馬迴是學習與記憶的重要腦區,進一步針對八周PS-MPs公鼠海馬迴進行分子分析後,發現對突觸可塑性和記憶力至關重要的Activity-regulated cytoskeleton-associated protein (Arc),其mRNA和蛋白質表達在海馬迴中顯著降低,但在PS-MPs母鼠中則沒有明顯差異。由於Arc蛋白會影響α-氨基-3-羥基-5-甲基-4-異噁唑丙酸受體(α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, AMPAR)亞基之一的突觸蛋白GluA1的內吞作用,因此發現GluA1在八周PS-MPs公鼠的海馬迴中異常增加。此外,本篇研究也發現暴露PS-MPs的公鼠八周後會會增加海馬迴促炎因子TNF-α與IL-1β的mRNA表現量及小膠質細胞的活化。綜合上述結果顯示小鼠暴露PS-MPs會改變突觸相關蛋白的表達以及神經發炎,進而削弱海馬迴依賴性學習和記憶。
摘要(英) Microplastics (MPs), defined as plastics particles less than 5 mm in diameter, are an emerging environmental pollutant in worldwide. Sine MPs are smaller in size, they can enter organisms via the food chain and accumulate in tissues/organs, subsequently causing damage to the organs. Previous study has confirmed that polystyrene micropartics (PS-MPs) can affect learning and memory ability in male mice. However, the molecular mechanism on how PS-MPs affecting learning and memory is still unknown. Using Raman spectroscopy analysis, 2 μm PS-MPs were found in the liver and the hippocampus of mice with oral administration of PS-MPs for eight weeks. Since the hippocampus is an important brain areafor learning and memory, Ifurther explored the molecular mechanism in the hippocampus. I found that the mRNA and protein expressions of activity-regulated cytoskeleton-associated protein (Arc), a key regulator for synaptic plasticity and learning and memory, was significantly decreased in the hippocampus, but there was no difference in PS-MPs female mice. The Arc protein can regulate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) endocytosis. I found that GluA1, a subunit of AMPAR, was aberrantly increased in the hippocampus of PS-MPs male mice.Moreover, I also found that exposure to PS-MPs in male mice for eight weeks increased the mRNA expression of pro-inflammatory cytokines TNF-α and IL-1β in the hippocampus and induced microglia activation. Taken together, these results suggest that exposure to PS-MPs in mice alters synapse-associated protein expression and neuroinflammation, thereby impairing hippocampal-dependent learning and memory.
關鍵字(中) ★ 聚苯乙烯塑膠微粒
★ 海馬迴
★ 學習和記憶
★ 突觸可塑性分子Arc
關鍵字(英)
論文目次 中文摘要 i
英文摘要Abstract ii
聲明(Declaration) iii
誌謝 iv
目錄 v
圖目錄 viii
表目錄 ix
中英文對照表 x
一、 緒論 1
1-1 塑膠微粒 1
1-2 塑膠微粒暴露的途徑 1
1-3 人類暴露塑膠微粒相關案例 3
1-4 暴露塑膠微粒對細胞分子層次的改變 4
1-5 聚苯乙烯塑膠微粒對周邊器官的影響-以無脊椎動物、魚類為例 5
1-6 聚苯乙烯塑膠微粒對周邊器官的影響-以齧齒類動物為例 6
1-7 聚苯乙烯塑膠微粒對中樞神經系統的影響-以無脊椎動物、魚類為例 8
1-8 聚苯乙烯塑膠微粒對中樞神經系統的影響-以齧齒類動物為例 8
1-9 聚乙烯塑膠微粒對中樞神經系統的影響-以齧齒類動物為例 9
1-10 研究動機與目的 9
二、 研究方法 11
2-1 實驗動物 11
2-2 建立塑膠微粒動物模型 11
2-3 拉曼光譜之螢光PS-MPs組織萃取 11
2-4 西方墨點法 12
2-5 突觸神經體萃取 13
2-6 免疫螢光染色 14
2-7 RNA萃取 14
2-8 RNA定序(RNA -Sequencing) 14
2-9 定量聚合酶連鎖反應 15
2-10 統計分析 16
三、 實驗結果 17
3-1 PS-MPs對於公和母鼠體重、運動能力及焦慮表現之影響 17
3-2 PS-MPs會累積至小鼠肝臟與大腦中 17
3-3 餵食PS-MPs之小鼠造成即刻早期基因(Immediate early gene, IEG)改變 18
3-4 攝入PS-MPs引起公鼠海馬迴神經突觸可塑性蛋白Arc表達改變 18
3-5 攝入PS-MPs引起公鼠海馬迴突觸蛋白的變化 19
3-6 攝入PS-MPs誘導公鼠海馬迴神經發炎 19
3-7 攝入PS-MPs誘導公鼠海馬迴小膠質細胞活化 20
四、 討論 21
五、 結論 25
參考文獻 38
附錄 44
附錄一、西方墨點法使用到之抗體 44
附錄二、免疫螢光染色使用到之抗體 45
附錄三、定量聚合酶連鎖反應使用到之Primer 46
參考文獻 1. Derraik, J.G.B., The pollution of the marine environment by plastic debris: a review. Marine Pollution Bulletin, 2002. 44(9): p. 842-852.
2. Cox, K.D., et al., Human Consumption of Microplastics. Environmental Science & Technology, 2019. 53(12): p. 7068-7074.
3. 美國國家海洋暨大氣總署(National Oceanic and Atmospheric Administration, NOAA), Microplastics. Available from: https://marinedebris.noaa.gov/what-marine-debris/microplastics.
4. Rodrigues, M.O., et al., Impacts of plastic products used in daily life on the environment and human health: What is known? Environmental Toxicology and Pharmacology, 2019. 72: p. 103239.
5. Du, F., et al., Microplastics in take-out food containers. Journal of Hazardous Materials, 2020. 399: p. 122969.
6. Guzzetti, E., et al., Microplastic in marine organism: Environmental and toxicological effects. Environmental Toxicology and Pharmacology, 2018. 64: p. 164-171.
7. Revel, M., A. Châtel, and C. Mouneyrac, Micro(nano)plastics: A threat to human health? Current Opinion in Environmental Science & Health, 2018. 1: p. 17-23.
8. Mohamed Nor, N.H., et al., Lifetime Accumulation of Microplastic in Children and Adults. Environmental Science & Technology, 2021. 55(8): p. 5084-5096.
9. Prata, J.C., Airborne microplastics: Consequences to human health? Environmental Pollution, 2018. 234: p. 115-126.
10. Carr, S.A., J. Liu, and A.G. Tesoro, Transport and fate of microplastic particles in wastewater treatment plants. Water Research, 2016. 91: p. 174-182.
11. Güven, O., et al., Microplastic litter composition of the Turkish territorial waters of the Mediterranean Sea, and its occurrence in the gastrointestinal tract of fish. Environmental Pollution, 2017. 223: p. 286-294.
12. Bråte, I.L.N., et al., Plastic ingestion by Atlantic cod (Gadus morhua) from the Norwegian coast. Marine Pollution Bulletin, 2016. 112(1): p. 105-110.
13. Kosuth, M., S.A. Mason, and E.V. Wattenberg, Anthropogenic contamination of tap water, beer, and sea salt. PLOS ONE, 2018. 13(4): p. e0194970.
14. Liebezeit, G. and E. Liebezeit, Synthetic particles as contaminants in German beers. Food Additives & Contaminants: Part A, 2014. 31(9): p. 1574-1578.
15. Karami, A., et al., The presence of microplastics in commercial salts from different countries. Scientific Reports, 2017. 7(1): p. 46173.
16. Oliveri Conti, G., et al., Micro- and nano-plastics in edible fruit and vegetables. The first diet risks assessment for the general population. Environmental Research, 2020. 187: p. 109677.
17. Amato-Lourenço, L.F., et al., An emerging class of air pollutants: Potential effects of microplastics to respiratory human health? Science of The Total Environment, 2020. 749: p. 141676.
18. Dris, R., et al., Microplastic contamination in an urban area: a case study in Greater Paris. Environmental Chemistry, 2015. 12(5): p. 592-599.
19. Dris, R., et al., A first overview of textile fibers, including microplastics, in indoor and outdoor environments. Environmental Pollution, 2016. 221.
20. Schwabl, P., et al., Detection of Various Microplastics in Human Stool. Annals of Internal Medicine, 2019. 171(7): p. 453-457.
21. Schmidt, C., et al., Nano- and microscaled particles for drug targeting to inflamed intestinal mucosa—A first in vivo study in human patients. Journal of Controlled Release, 2013. 165(2): p. 139-145.
22. Ragusa, A., et al., Plasticenta: First evidence of microplastics in human placenta. Environment International, 2021. 146: p. 106274.
23. Leslie, H.A., et al., Discovery and quantification of plastic particle pollution in human blood. Environment International, 2022. 163: p. 107199.
24. Amato-Lourenço, L.F., et al., Presence of airborne microplastics in human lung tissue. Journal of Hazardous Materials, 2021. 416: p. 126124.
25. Jenner, L.C., et al., Detection of microplastics in human lung tissue using μFTIR spectroscopy. Science of The Total Environment, 2022. 831: p. 154907.
26. Wang, Y.-L., et al., The Kidney-Related Effects of Polystyrene Microplastics on Human Kidney Proximal Tubular Epithelial Cells HK-2 and Male C57BL/6 Mice. Environmental Health Perspectives. 129(5): p. 057003.
27. Wu, B., et al., Size-dependent effects of polystyrene microplastics on cytotoxicity and efflux pump inhibition in human Caco-2 cells. Chemosphere, 2019. 221: p. 333-341.
28. Lee, H.-S., et al., Adverse effect of polystyrene microplastics (PS-MPs) on tube formation and viability of human umbilical vein endothelial cells. Food and Chemical Toxicology, 2021. 154: p. 112356.
29. Espinosa, C., et al., In vitro effects of virgin microplastics on fish head-kidney leucocyte activities. Environmental Pollution, 2018. 235: p. 30-38.
30. Zwollo, P., et al., Polystyrene microplastics reduce abundance of developing B cells in rainbow trout (Oncorhynchus mykiss) primary cultures. Fish & Shellfish Immunology, 2021. 114: p. 102-111.
31. Liu, T., et al., Polystyrene microplastics induce mitochondrial damage in mouse GC-2 cells. Ecotoxicology and Environmental Safety, 2022. 237: p. 113520.
32. Liu, L., et al., Cellular internalization and release of polystyrene microplastics and nanoplastics. Science of The Total Environment, 2021. 779: p. 146523.
33. Huang, J.-N., et al., Exposure to microplastics impairs digestive performance, stimulates immune response and induces microbiota dysbiosis in the gut of juvenile guppy (Poecilia reticulata). Science of The Total Environment, 2020. 733: p. 138929.
34. Feng, S., et al., Polystyrene microplastics alter the intestinal microbiota function and the hepatic metabolism status in marine medaka (Oryzias melastigma). Science of The Total Environment, 2021. 759: p. 143558.
35. Kaloyianni, M., et al., Toxicity and Functional Tissue Responses of Two Freshwater Fish after Exposure to Polystyrene Microplastics. Toxics, 2021. 9(11).
36. Yu, Y., et al., Polystyrene microplastics (PS-MPs) toxicity induced oxidative stress and intestinal injury in nematode Caenorhabditis elegans. Science of The Total Environment, 2020. 726: p. 138679.
37. Paul-Pont, I., et al., Exposure of marine mussels Mytilus spp. to polystyrene microplastics: Toxicity and influence on fluoranthene bioaccumulation. Environmental Pollution, 2016. 216: p. 724-737.
38. Liu, Z., et al., Effects of microplastics on the innate immunity and intestinal microflora of juvenile Eriocheir sinensis. Science of The Total Environment, 2019. 685: p. 836-846.
39. Deng, Y., et al., Tissue accumulation of microplastics in mice and biomarker responses suggest widespread health risks of exposure. Scientific Reports, 2017. 7(1): p. 46687.
40. Lu, L., et al., Polystyrene microplastics induce gut microbiota dysbiosis and hepatic lipid metabolism disorder in mice. (1879-1026 (Electronic)).
41. Zheng, H., et al., Proinflammatory properties and lipid disturbance of polystyrene microplastics in the livers of mice with acute colitis. Science of The Total Environment, 2021. 750: p. 143085.
42. Jin, H., et al., Polystyrene microplastics induced male reproductive toxicity in mice. Journal of Hazardous Materials, 2021. 401: p. 123430.
43. Wei, Y., et al., Polystyrene microplastics disrupt the blood-testis barrier integrity through ROS-Mediated imbalance of mTORC1 and mTORC2. Environmental Pollution, 2021. 289: p. 117904.
44. Hou, J., et al., Polystyrene microplastics lead to pyroptosis and apoptosis of ovarian granulosa cells via NLRP3/Caspase-1 signaling pathway in rats. Ecotoxicology and Environmental Safety, 2021. 212: p. 112012.
45. An, R., et al., Polystyrene microplastics cause granulosa cells apoptosis and fibrosis in ovary through oxidative stress in rats. Toxicology, 2021. 449: p. 152665.
46. Li, X., et al., Intratracheal administration of polystyrene microplastics induces pulmonary fibrosis by activating oxidative stress and Wnt/β-catenin signaling pathway in mice. Ecotoxicology and Environmental Safety, 2022. 232: p. 113238.
47. Lim, D., et al., Inhalation toxicity of polystyrene micro(nano)plastics using modified OECD TG 412. Chemosphere, 2021. 262: p. 128330.
48. Fournier, S.B., et al., Nanopolystyrene translocation and fetal deposition after acute lung exposure during late-stage pregnancy. Particle and Fibre Toxicology, 2020. 17(1): p. 55.
49. Prüst, M., J. Meijer, and R.H.S. Westerink, The plastic brain: neurotoxicity of micro- and nanoplastics. Particle and Fibre Toxicology, 2020. 17(1): p. 24.
50. Lei, L., et al., Polystyrene (nano)microplastics cause size-dependent neurotoxicity, oxidative damage and other adverse effects in Caenorhabditis elegans. Environmental Science: Nano, 2018. 5(8): p. 2009-2020.
51. Chen, H., et al., Chronic exposure to UV-aged microplastics induces neurotoxicity by affecting dopamine, glutamate, and serotonin neurotransmission in Caenorhabditis elegans. Journal of Hazardous Materials, 2021. 419: p. 126482.
52. Zhang, S., et al., Interactive effects of polystyrene microplastics and roxithromycin on bioaccumulation and biochemical status in the freshwater fish red tilapia (Oreochromis niloticus). (1879-1026 (Electronic)).
53. Hoyo-Alvarez, E., et al., Effects of pollutants and microplastics ingestion on oxidative stress and monoaminergic activity of seabream brains. Aquatic Toxicology, 2022. 242: p. 106048.
54. Kwon, W., et al., Microglial phagocytosis of polystyrene microplastics results in immune alteration and apoptosis in vitro and in vivo. Science of The Total Environment, 2022. 807: p. 150817.
55. Liang, B., et al., Brain single-nucleus transcriptomics highlights that polystyrene nanoplastics potentially induce Parkinson’s disease-like neurodegeneration by causing energy metabolism disorders in mice. Journal of Hazardous Materials, 2022. 430: p. 128459.
56. Wang, S., et al., Polystyrene microplastics affect learning and memory in mice by inducing oxidative stress and decreasing the level of acetylcholine. Food and Chemical Toxicology, 2022. 162: p. 112904.
57. Zaheer, J., et al., Pre/post-natal exposure to microplastic as a potential risk factor for autism spectrum disorder. Environment International, 2022. 161: p. 107121.
58. da Costa Araújo, A.P. and G. Malafaia, Microplastic ingestion induces behavioral disorders in mice: A preliminary study on the trophic transfer effects via tadpoles and fish. Journal of Hazardous Materials, 2021. 401: p. 123263.
59. Lee, C.W., et al., Exposure to polystyrene microplastics impairs hippocampus-dependent learning and memory in mice. J Hazard Mater, 2022. 430: p. 128431.
60. Chowdhury, S., et al., Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron, 2006. 52(3): p. 445-59.
61. Plath, N., et al., Arc/Arg3.1 is essential for the consolidation of synaptic plasticity and memories. Neuron, 2006. 52(3): p. 437-44.
62. Rial Verde, E.M., et al., Increased expression of the immediate-early gene arc/arg3.1 reduces AMPA receptor-mediated synaptic transmission. Neuron, 2006. 52(3): p. 461-74.
63. Shepherd, J.D., et al., Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron, 2006. 52(3): p. 475-84.
64. Stampanoni Bassi, M., et al., Neurophysiology of synaptic functioning in multiple sclerosis. Clinical Neurophysiology, 2017. 128(7): p. 1148-1157.
65. Rizzo, F.R., et al., Tumor Necrosis Factor and Interleukin-1<i>β</i> Modulate Synaptic Plasticity during Neuroinflammation. Neural Plasticity, 2018. 2018: p. 8430123.
66. Bourgognon, J.-M. and J. Cavanagh, The role of cytokines in modulating learning and memory and brain plasticity. Brain and Neuroscience Advances, 2020. 4: p. 2398212820979802.
67. Miranda, A. and A. Lorke, Stability of suspended monolayer graphene membranes in alkaline environment. Materials Research Letters, 2017. 6(1): p. 49-54.
68. Wright, S.L. and F.J. Kelly, Plastic and Human Health: A Micro Issue? Environmental Science & Technology, 2017. 51(12): p. 6634-6647.
69. Louveau, A., et al., Structural and functional features of central nervous system lymphatic vessels. (1476-4687 (Electronic)).
70. Louveau, A., et al., CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nature Neuroscience, 2018. 21(10): p. 1380-1391.
71. Louveau, A., et al., Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. (1558-8238 (Electronic)).
72. Kaur, C. and E.A. Ling, The circumventricular organs. (1699-5848 (Electronic)).
73. Fry, W.M. and A.V. Ferguson, Circumventricular Organs, in Encyclopedia of Neuroscience, L.R. Squire, Editor. 2009, Academic Press: Oxford. p. 997-1002.
74. Minatohara, K., M. Akiyoshi, and H. Okuno, Role of Immediate-Early Genes in Synaptic Plasticity and Neuronal Ensembles Underlying the Memory Trace. Frontiers in Molecular Neuroscience, 2016. 8.
75. Gallo, F.T., et al., Immediate Early Genes, Memory and Psychiatric Disorders: Focus on c-Fos, Egr1 and Arc. Frontiers in Behavioral Neuroscience, 2018. 12.
76. Chowdhury, S., et al., Arc/Arg3.1 Interacts with the Endocytic Machinery to Regulate AMPA Receptor Trafficking. Neuron, 2006. 52(3): p. 445-459.
77. Rial Verde, E.M., et al., Increased Expression of the Immediate-Early Gene <em>Arc/Arg3.1</em> Reduces AMPA Receptor-Mediated Synaptic Transmission. Neuron, 2006. 52(3): p. 461-474.
78. Shepherd, J.D., et al., Arc/Arg3.1 Mediates Homeostatic Synaptic Scaling of AMPA Receptors. Neuron, 2006. 52(3): p. 475-484.
79. Khairova, R.A., et al., A potential role for pro-inflammatory cytokines in regulating synaptic plasticity in major depressive disorder. International Journal of Neuropsychopharmacology, 2009. 12(4): p. 561-578.
80. Stellwagen, D. and R.C. Malenka, Synaptic scaling mediated by glial TNF-α. Nature, 2006. 440(7087): p. 1054-1059.
81. Gegenhuber, B. and J. Tollkuhn, Signatures of sex: Sex differences in gene expression in the vertebrate brain. WIREs Developmental Biology, 2020. 9(1): p. e348.
82. Choleris, E., et al., Sex differences in the brain: Implications for behavioral and biomedical research. Neuroscience & Biobehavioral Reviews, 2018. 85: p. 126-145.
83. Jazin, E. and L. Cahill, Sex differences in molecular neuroscience: from fruit flies to humans. Nature Reviews Neuroscience, 2010. 11(1): p. 9-17.
84. Cosgrove, K.P., J.K. Mazure Cm Fau - Staley, and J.K. Staley, Evolving knowledge of sex differences in brain structure, function, and chemistry. (0006-3223 (Print)).
85. Dalla, C., et al., Female rats learn trace memories better than male rats and consequently retain a greater proportion of new neurons in their hippocampi. (1091-6490 (Electronic)).
86. Sherwin, B.B., Estrogen and/or androgen replacement therapy and cognitive functioning in surgically menopausal women. Psychoneuroendocrinology, 1988. 13(4): p. 345-357.
87. Leranth, C., T. Hajszan, and N.J. MacLusky, Androgens Increase Spine Synapse Density in the CA1 Hippocampal Subfield of Ovariectomized Female Rats. The Journal of Neuroscience, 2004. 24(2): p. 495.
88. Leranth, C., O. Petnehazy, and N.J. MacLusky, Gonadal Hormones Affect Spine Synaptic Density in the CA1 Hippocampal Subfield of Male Rats. The Journal of Neuroscience, 2003. 23(5): p. 1588.
89. Tabatadze, N., et al., Sex Differences in Molecular Signaling at Inhibitory Synapses in the Hippocampus. The Journal of Neuroscience, 2015. 35(32): p. 11252.
90. Elderman, M., et al., Sex and strain dependent differences in mucosal immunology and microbiota composition in mice. Biology of Sex Differences, 2018. 9(1): p. 26.
91. Fransen, F., et al., The Impact of Gut Microbiota on Gender-Specific Differences in Immunity. Frontiers in Immunology, 2017. 8.
92. Kim, Y.A.-O., et al., Sex Differences in Gut Microbiota. (2287-4208 (Print)).
93. Gareau, M.G., Microbiota-Gut-Brain Axis and Cognitive Function, in Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease, M. Lyte and J.F. Cryan, Editors. 2014, Springer New York: New York, NY. p. 357-371.
94. Tang, W., et al., Roles of Gut Microbiota in the Regulation of Hippocampal Plasticity, Inflammation, and Hippocampus-Dependent Behaviors. Frontiers in Cellular and Infection Microbiology, 2021. 10.
指導教授 黃佳瑜 審核日期 2022-7-26
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