帳號:guest(3.149.214.245)          離開系統
字體大小: 字級放大   字級縮小   預設字形  

詳目顯示

以作者查詢圖書館館藏以作者查詢臺灣博碩士論文系統以作者查詢全國書目勘誤回報
作者:張瑞芳
作者(英文):Christine Chang
論文名稱:Assessing the Feasibility of Adipose-derived Stem Cells to Alleviate Neurodegeneration in MSA Animal Models
論文名稱(英文):Assessing the Feasibility of Adipose-derived Stem Cells to Alleviate Neurodegeneration in MSA Animal Models
指導教授:邱紫文
指導教授(英文):Tzyy-Wen Chiou
口試委員:李佳洪
邱紫文
韓鴻志
李茹萍
蔡昇宗
口試委員(英文):CH Li
Tzyy-Wen Chiou
Horng-Jyh Harn
Ru-Ping Lee
Sheng-Tzung Tsai
學位類別:博士
校院名稱:國立東華大學
系所名稱:生命科學系
學號:810313101
出版年(民國):109
畢業學年度:109
語文別:英文
論文頁數:90
關鍵詞(英文):MSAADSCα-synucleinGDNFautophagyapoptosis
相關次數:
  • 推薦推薦:0
  • 點閱點閱:12
  • 評分評分:系統版面圖檔系統版面圖檔系統版面圖檔系統版面圖檔系統版面圖檔
  • 下載下載:2
  • 收藏收藏:0
Patients with multiple system atrophy (MSA), a progressive neurodegenerative disorder of adult onset, were found less than 9-year life expectancy after onset. The disorders include bradykinesia and rigidity commonly seen in Parkinsonism disease and additional signs such as autonomic dysfunction, ataxia, or dementia. MSA-P, representing 80% of MSA, is characterized by striatonigral neurodegeneration (SND) where parkinsonism predominates. In clinical treatments, MSA poorly responds to levodopa, the drug used to remedy Parkinsonism disease. The exact cause of MSA is still unknown and exploring a therapeutic solution to MSA remains critical. The potential of stem cell therapy for neural degenerative diseases has been widely explored recently. With the reported abilities to differentiate into neuronal linage and to secrete neurotrophic factors for neuronal support, mesenchymal stem cells (MSC) presents a possible therapeutic option for MSA-P. Among the sources of MSC, adipose-derived stem cells (ADSC) are relatively easier to obtain and less invasive. To conduct the feasibility study in vivo, toxin-based MSA-P mouse model and transgenic MSA mouse model were established. Toxin-based model, chemically (3NP + MPTP) inducing MSA-P in C57BL/6N mice, was established to assess the effect of ADSC on improving motion behavior of MSA-P mouse. Molecular mechanism was investigated using transgenic MSA mouse model to reveal the role of ADSC in alleviating neurodegeneration of MBP1 mouse. MBP1 transgenic mice, transferred from the University of California in San Diego, USA (UCSD), have been reported in studying α-synuclein associated neurodegeneration. Our transgenic mouse model established from MBP1 mouse provides a useful tool to investigate the effect of ADSC on the biomarker for MSA, i.e., α-synuclein glial cytoplasmic inclusion (GCI) in oligodendrocytes. Human ADSC were transplanted into the striatum of MSA animals via intracerebral injection (IC). Testing intranasal (INA) delivery route was withdrawn due to brain blood barrier (BBB) issue. The grafted ADSC was traced by labeling ADSC with transduced GFP and taking photo images on live mice. The presence of ADSC was detected from immunofluorescence (IF) stains 4 weeks post grafting. The alleviation of neurodegeneration after ADSC treatment was indicated by improved performance in animal behavior testing, including rotarod test and pole test. As compared with a sham control, ADSC significantly enhanced Rotarod performance of MSA animals treated with ADSC at an effective dose (2x10^5 ADSC/mouse). Our ex vivo study supported that ADSC might alleviate striatal degeneration in MSA animal model by improving nigrostriatal pathway for dopamine, activating autophagy for α-synuclein clearance, decreasing inflammatory signal and further cell apoptosis, improving myelination and cell survival at caudate-putamen. Mitochondria was found to transfer from ADSC to injured neuron in vitro. ADSC pretreated with mitochondria-rich chemical (MR1) had beneficial trend in improving Rotarod performance although the difference against ADSC was not statistically significant.
Chapter I INTRODUCTION 1
Chapter II MATERIALS AND METHODS 7
II-A. Preparing ADSC for feasibility study 7
II-B. Establishing chemically induced mouse model and investigating the effect of ADSC 8
II-C. Establishing SHSY5Y cell model to track mitochondria of ADSC, in vitro 12
II-D. Establishing α-synuclein over-expressed transgenic mouse model and investigating the effect of ADSC 13
II-E. Studying the mechanism using the α-synuclein over-expressed transgenic mouse model 15
Chapter III RESULTS 19
III-A. Preliminary Findings from Chemically Induced Models Supported Therapeutic Potential of ADSC for Treating MSA-P Disease 19
III-B. MSA Symptoms Revealed by the Over-expression of α-synuclein Driven by MBP Promotor 20
III-C. Beneficial Effects on the MSA Transgenic Mouse Model Revealed by ADSC Transplantation 20
III-D. Nigrostriatal Pathway for Dopamine Signal Improved by ADSC Transplantation 21
III-E. Potential of ADSC Transplantation in Activating Autophagy Protein Clearance Pathway to Decrease the Levels of Un-aggregated Form of α-synuclein 22
III-F. Potential of ADSC Transplantation in Decreasing Inflammatory Signal and Further Cell Apoptosis 23
Chapter IV DISCUSSION 25
Chapter V CONCLUSIONS 31
Chapter VI FUTURE WORK 33
REFERENCES 35
TABLES AND FIGURES 49
1. Jellinger KA: Potential clinical utility of multiple system atrophy biomarkers. Expert Rev Neurother 2017, 17:1189-1208.
2. Overk C, Rockenstein E, Valera E, Stefanova N, Wenning G, Masliah E: Multiple system atrophy: experimental models and reality. Acta neuropathologica 2018, 135:33-47.
3. Stefanova N: Translational therapies for multiple system atrophy: Bottlenecks and future directions. Auton Neurosci 2018, 211:7-14.
4. Bjornsdottir A, Gudmundsson G, Blondal H, Olafsson E: Incidence and prevalence of multiple system atrophy: a nationwide study in Iceland. J Neurol Neurosurg Psychiatry 2013, 84:136-140.
5. Wedge F: The impact of resistance training on balance and functional ability of a patient with multiple system atrophy. J Geriatr Phys Ther 2008, 31:79-83.
6. Noda K, Kobayashi T, Matsuoka S, Takanashi M, Kanazawa A, Mizuno Y: [A 65-year-old man with rigid-bradykinetic parkinsonism, vertical gaze palsy, difficulty of eye-lid opening, and marked pseudo-bulbar palsy]. No to shinkei = Brain and nerve 2005, 57:73-86.
7. Krampera M, Marconi S, Pasini A, Galie M, Rigotti G, Mosna F, Tinelli M, Lovato L, Anghileri E, Andreini A, et al: Induction of neural-like differentiation in human mesenchymal stem cells derived from bone marrow, fat, spleen and thymus. Bone 2007, 40:382-390.
8. Teixeira FG, Carvalho MM, Sousa N, Salgado AJ: Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration? Cell Mol Life Sci 2013, 70:3871-3882.
9. Woodbury D, Schwarz EJ, Prockop DJ, Black IB: Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res 2000, 61:364-370.
10. Ooi YY, Dheen ST, Tay SS: Paracrine effects of mesenchymal stem cells-conditioned medium on microglial cytokines expression and nitric oxide production. Neuroimmunomodulation 2015, 22:233-242.
11. Ullah I, Subbarao RB, Rho GJ: Human mesenchymal stem cells - current trends and future prospective. Biosci Rep 2015, 35.
12. Benameur L, Charif N, Li Y, Stoltz JF, de Isla N: Toward an understanding of mechanism of aging-induced oxidative stress in human mesenchymal stem cells. Biomed Mater Eng 2015, 25:41-46.
13. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284:143-147.
14. Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, Blake J, Schwager C, Eckstein V, Ansorge W, Ho AD: Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol 2005, 33:1402-1416.
15. Zhang X, Yang M, Lin L, Chen P, Ma KT, Zhou CY, Ao YF: Runx2 overexpression enhances osteoblastic differentiation and mineralization in adipose--derived stem cells in vitro and in vivo. Calcif Tissue Int 2006, 79:169-178.
16. Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC, Chen CC: Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells 2004, 22:1330-1337.
17. Huang GT, Gronthos S, Shi S: Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res 2009, 88:792-806.
18. Kolaparthy LK, Sanivarapu S, Moogla S, Kutcham RS: Adipose Tissue - Adequate, Accessible Regenerative Material. Int J Stem Cells 2015, 8:121-127.
19. Stefanova N, Wenning GK: Animal models of multiple system atrophy. Clin Auton Res 2015, 25:9-17.
20. Fernagut PO, Diguet E, Bioulac B, Tison F: MPTP potentiates 3-nitropropionic acid-induced striatal damage in mice: reference to striatonigral degeneration. Exp Neurol 2004, 185:47-62.
21. Stefanova N, Tison F, Reindl M, Poewe W, Wenning GK: Animal models of multiple system atrophy. Trends Neurosci 2005, 28:501-506.
22. Fernagut PO, Tison F: Animal models of multiple system atrophy. Neuroscience 2012, 211:77-82.
23. Wenning GK, Granata R, Laboyrie PM, Quinn NP, Jenner P, Marsden CD: Reversal of behavioural abnormalities by fetal allografts in a novel rat model of striatonigral degeneration. Movement disorders : official journal of the Movement Disorder Society 1996, 11:522-532.
24. Scholz J, Niibori Y, P WF, J PL: Rotarod training in mice is associated with changes in brain structure observable with multimodal MRI. NeuroImage 2015, 107:182-189.
25. Heyser CJ, Vishnevetsky D, Berten S: The effect of cocaine on rotarod performance in male C57BL/6J mice. Physiology & behavior 2013, 118:208-211.
26. Stroobants S, Gantois I, Pooters T, D'Hooge R: Increased gait variability in mice with small cerebellar cortex lesions and normal rotarod performance. Behavioural brain research 2013, 241:32-37.
27. Li M, Huang Y, Ma AA, Lin E, Diamond MI: Y-27632 improves rotarod performance and reduces huntingtin levels in R6/2 mice. Neurobiology of disease 2009, 36:413-420.
28. Cendelin J, Korelusova I, Vozeh F: The effect of repeated rotarod training on motor skills and spatial learning ability in Lurcher mutant mice. Behavioural brain research 2008, 189:65-74.
29. Luszczki JJ, Andres MM, Czuczwar P, Cioczek-Czuczwar A, Wojcik-Cwikla J, Ratnaraj N, Patsalos PN, Czuczwar SJ: Levetiracetam selectively potentiates the acute neurotoxic effects of topiramate and carbamazepine in the rotarod test in mice. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology 2005, 15:609-616.
30. Rustay NR, Wahlsten D, Crabbe JC: Influence of task parameters on rotarod performance and sensitivity to ethanol in mice. Behavioural brain research 2003, 141:237-249.
31. Lutfy K, Weber E: Tolerance develops to the antinociceptive and motor impairing effects of ACEA-1416, a NMDA receptor antagonist, in the formalin and rotarod test in mice. Pharmacological research : the official journal of the Italian Pharmacological Society 1998, 37:295-302.
32. Kuhn PL, Petroulakis E, Zazanis GA, McKinnon RD: Motor function analysis of myelin mutant mice using a rotarod. International journal of developmental neuroscience : the official journal of the International Society for Developmental Neuroscience 1995, 13:715-722.
33. Stinchcomb A, Bowers BJ, Wehner JM: The effects of ethanol and Ro 15-4513 on elevated plus-maze and rotarod performance in long-sleep and short-sleep mice. Alcohol 1989, 6:369-376.
34. Stromberg C: Interactions of antidepressants and ethanol on spontaneous locomotor activity and rotarod performance in NMRI and C57BL/6 mice. Journal of psychopharmacology 1988, 2:61-66.
35. Pearl J, Stander H, McKean DB: Effects of analgesics and other drugs on mice in phenylquinone and rotarod tests. The Journal of pharmacology and experimental therapeutics 1969, 167:9-13.
36. Jones BJ, Roberts DJ: The quantiative measurement of motor inco-ordination in naive mice using an acelerating rotarod. The Journal of pharmacy and pharmacology 1968, 20:302-304.
37. Jones BJ, Roberts DJ: A rotarod suitable for quantitative measurements of motor incoordination in naive mice. Naunyn-Schmiedebergs Archiv fur experimentelle Pathologie und Pharmakologie 1968, 259:211.
38. Watzman N, Barry H, 3rd, Kinnard WJ, Jr., Buckley JP: Influence of certain parameters on the performance of mice on the rotarod. Archives internationales de pharmacodynamie et de therapie 1967, 169:362-374.
39. Plotnikoff N, Reinke D, Fitzloff J: Effects of stimulants on rotarod performance of mice. Journal of pharmaceutical sciences 1962, 51:1007-1008.
40. Diguet E, Fernagut PO, Scherfler C, Wenning G, Tison F: Effects of riluzole on combined MPTP + 3-nitropropionic acid-induced mild to moderate striatonigral degeneration in mice. Journal of neural transmission 2005, 112:613-631.
41. Bleasel JM, Halliday GM, Kim WS: Animal modeling an oligodendrogliopathy--multiple system atrophy. Acta Neuropathol Commun 2016, 4:12.
42. Shults CW, Rockenstein E, Crews L, Adame A, Mante M, Larrea G, Hashimoto M, Song D, Iwatsubo T, Tsuboi K, Masliah E: Neurological and neurodegenerative alterations in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy. J Neurosci 2005, 25:10689-10699.
43. Stefanova N, Kaufmann WA, Humpel C, Poewe W, Wenning GK: Systemic proteasome inhibition triggers neurodegeneration in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy. Acta neuropathologica 2012, 124:51-65.
44. Monzio Compagnoni G, Di Fonzo A: Understanding the pathogenesis of multiple system atrophy: state of the art and future perspectives. Acta Neuropathol Commun 2019, 7:113.
45. Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E, Goldberg MS, Shen J, Takio K, Iwatsubo T: alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nature cell biology 2002, 4:160-164.
46. Rcom-H'cheo-Gauthier AN, Osborne SL, Meedeniya AC, Pountney DL: Calcium: Alpha-Synuclein Interactions in Alpha-Synucleinopathies. Front Neurosci 2016, 10:570.
47. Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, Wood NW, Colosimo C, Durr A, Fowler CJ, et al: Second consensus statement on the diagnosis of multiple system atrophy. Neurology 2008, 71:670-676.
48. Kalivendi SV, Yedlapudi D, Hillard CJ, Kalyanaraman B: Oxidants induce alternative splicing of alpha-synuclein: Implications for Parkinson's disease. Free Radic Biol Med 2010, 48:377-383.
49. Iwata A, Maruyama M, Kanazawa I, Nukina N: alpha-Synuclein affects the MAPK pathway and accelerates cell death. J Biol Chem 2001, 276:45320-45329.
50. Refolo V, Stefanova N: Neuroinflammation and Glial Phenotypic Changes in Alpha-Synucleinopathies. Front Cell Neurosci 2019, 13:263.
51. O'Callaghan JP, Sriram K: Glial fibrillary acidic protein and related glial proteins as biomarkers of neurotoxicity. Expert opinion on drug safety 2005, 4:433-442.
52. Ito D, Tanaka K, Suzuki S, Dembo T, Fukuuchi Y: Enhanced expression of Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral ischemia in rat brain. Stroke 2001, 32:1208-1215.
53. Ubhi K, Rockenstein E, Mante M, Patrick C, Adame A, Thukral M, Shults C, Masliah E: Rifampicin reduces alpha-synuclein in a transgenic mouse model of multiple system atrophy. Neuroreport 2008, 19:1271-1276.
54. Fortin DL, Nemani VM, Nakamura K, Edwards RH: The behavior of alpha-synuclein in neurons. Movement disorders : official journal of the Movement Disorder Society 2010, 25 Suppl 1:S21-26.
55. Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, Chaudhry FA, Nicoll RA, Edwards RH: Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 2010, 65:66-79.
56. Tofaris GK, Spillantini MG: Physiological and pathological properties of alpha-synuclein. Cell Mol Life Sci 2007, 64:2194-2201.
57. Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Fullgrabe J, Jackson A, Jimenez Sanchez M, Karabiyik C, et al: Autophagy and Neurodegeneration: Pathogenic Mechanisms and Therapeutic Opportunities. Neuron 2017, 93:1015-1034.
58. Schwarz L, Goldbaum O, Bergmann M, Probst-Cousin S, Richter-Landsberg C: Involvement of macroautophagy in multiple system atrophy and protein aggregate formation in oligodendrocytes. J Mol Neurosci 2012, 47:256-266.
59. Miki Y, Tanji K, Mori F, Tatara Y, Utsumi J, Sasaki H, Kakita A, Takahashi H, Fimia GM, Wakabayashi K: AMBRA1, a novel alpha-synuclein-binding protein, is implicated in the pathogenesis of multiple system atrophy. Brain Pathol 2018, 28:28-42.
60. Kaji S, Maki T, Kinoshita H, Uemura N, Ayaki T, Kawamoto Y, Furuta T, Urushitani M, Hasegawa M, Kinoshita Y, et al: Pathological Endogenous alpha-Synuclein Accumulation in Oligodendrocyte Precursor Cells Potentially Induces Inclusions in Multiple System Atrophy. Stem Cell Reports 2018, 10:356-365.
61. Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L: Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders. Science 2000, 287:1265-1269.
62. Tanji K, Odagiri S, Maruyama A, Mori F, Kakita A, Takahashi H, Wakabayashi K: Alteration of autophagosomal proteins in the brain of multiple system atrophy. Neurobiol Dis 2013, 49:190-198.
63. Pukass K, Richter-Landsberg C: Inhibition of UCH-L1 in oligodendroglial cells results in microtubule stabilization and prevents alpha-synuclein aggregate formation by activating the autophagic pathway: implications for multiple system atrophy. Front Cell Neurosci 2015, 9:163.
64. Wakabayashi K, Tanji K: [Multiple system atrophy and autophagy]. Rinsho Shinkeigaku 2014, 54:966-968.
65. Prusiner SB, Woerman AL, Mordes DA, Watts JC, Rampersaud R, Berry DB, Patel S, Oehler A, Lowe JK, Kravitz SN, et al: Evidence for alpha-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proceedings of the National Academy of Sciences of the United States of America 2015, 112:E5308-5317.
66. Samiotaki G, Acosta C, Wang S, Konofagou EE: Enhanced delivery and bioactivity of the neurturin neurotrophic factor through focused ultrasound-mediated blood--brain barrier opening in vivo. J Cereb Blood Flow Metab 2015, 35:611-622.
67. Verma A: Prions, prion-like prionoids, and neurodegenerative disorders. Ann Indian Acad Neurol 2016, 19:169-174.
68. Fillon G, Kahle PJ: Alpha-synuclein transgenic mice: relevance to multiple system atrophy. Movement disorders : official journal of the Movement Disorder Society 2005, 20 Suppl 12:S64-66.
69. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ: Repeated mesenchymal stem cell treatment after neonatal hypoxia-ischemia has distinct effects on formation and maturation of new neurons and oligodendrocytes leading to restoration of damage, corticospinal motor tract activity, and sensorimotor function. J Neurosci 2010, 30:9603-9611.
70. Jung N, Park S, Choi Y, Park JW, Hong YB, Park HH, Yu Y, Kwak G, Kim HS, Ryu KH, et al: Tonsil-Derived Mesenchymal Stem Cells Differentiate into a Schwann Cell Phenotype and Promote Peripheral Nerve Regeneration. Int J Mol Sci 2016, 17.
71. Shahnawaz M, Tokuda T, Waragai M, Mendez N, Ishii R, Trenkwalder C, Mollenhauer B, Soto C: Development of a Biochemical Diagnosis of Parkinson Disease by Detection of alpha-Synuclein Misfolded Aggregates in Cerebrospinal Fluid. JAMA Neurol 2017, 74:163-172.
72. Yang W, Yu S: Synucleinopathies: common features and hippocampal manifestations. Cell Mol Life Sci 2017, 74:1485-1501.
73. Ashkenazi A, Bento CF, Ricketts T, Vicinanza M, Siddiqi F, Pavel M, Squitieri F, Hardenberg MC, Imarisio S, Menzies FM, Rubinsztein DC: Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature 2017, 545:108-111.
74. Stefanova N, Reindl M, Neumann M, Kahle PJ, Poewe W, Wenning GK: Microglial activation mediates neurodegeneration related to oligodendroglial alpha-synucleinopathy: implications for multiple system atrophy. Movement disorders : official journal of the Movement Disorder Society 2007, 22:2196-2203.
75. d'Anglemont de Tassigny X, Pascual A, Lopez-Barneo J: GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015, 9:10.
76. Teixeira FG, Carvalho MM, Panchalingam KM, Rodrigues AJ, Mendes-Pinheiro B, Anjo S, Manadas B, Behie LA, Sousa N, Salgado AJ: Impact of the Secretome of Human Mesenchymal Stem Cells on Brain Structure and Animal Behavior in a Rat Model of Parkinson's Disease. Stem Cells Transl Med 2017, 6:634-646.
77. Kempermann G, Gage FH, Aigner L, Song H, Curtis MA, Thuret S, Kuhn HG, Jessberger S, Frankland PW, Cameron HA, et al: Human Adult Neurogenesis: Evidence and Remaining Questions. Cell Stem Cell 2018, 23:25-30.
78. Paredes MF, Sorrells SF, Cebrian-Silla A, Sandoval K, Qi D, Kelley KW, James D, Mayer S, Chang J, Auguste KI, et al: Does Adult Neurogenesis Persist in the Human Hippocampus? Cell Stem Cell 2018, 23:780-781.
79. Moreno-Jimenez EP, Flor-Garcia M, Terreros-Roncal J, Rabano A, Cafini F, Pallas-Bazarra N, Avila J, Llorens-Martin M: Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer's disease. Nat Med 2019, 25:554-560.
80. Winner B, Kohl Z, Gage FH: Neurodegenerative disease and adult neurogenesis. Eur J Neurosci 2011, 33:1139-1151.
(此全文20251213後開放外部瀏覽)
01.pdf
 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *