博碩士論文 100233003 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:40 、訪客IP:18.216.32.116
姓名 彭奕憲(Yi-Shian Peng)  查詢紙本館藏   畢業系所 系統生物與生物資訊研究所
論文名稱 基因體功能統合分析在阿茲海默症和大腦老化-近年阿茲海默症研發藥物失敗的理論問題探討
(Commentary on recent Alzheimer’s trial failures: comparative functional genomic meta-analysis of Alzheimer’s affected and naturally aging brains)
相關論文
★ 人類陰道滴蟲之Myb2蛋白質動態性質研究★ 分析原核生物基因體複製起點與終點的反向對偶對稱現象
★ 分析基因體拷貝數變異所使用的兩種方法比較:隱藏馬可夫模型與成對高斯合併法★ 使用兩種方法偵測基因體拷貝數變異:成對高斯合併法與隱藏馬可夫模型
★ 以整體晶片數據為母體應用於分析基因差異表達的z檢定方法★ GSLHC - 運用基因組及層次類聚以生物功能群將有生物活性的複合物定性的方法
★ 一個檢定測量微晶片基因表達數據靈敏度的全統計計算法★ 運用嶄新抗體固著策略發展及驗證新式抗體微晶片平台
★ Drug-resistant colon cancer cells produce high carcinoembryonic antigen and might not be cancer-initiating cells★ 創傷性關節炎軟骨之退化進程- 大鼠模型基因體圖譜研究
★ 運用時間序列微陣列資料來預測調控基因★ 以大鼠嗜鉻性瘤細胞株建立神經訊號傳遞之細胞分子生物學模型
★ 一種找尋再利用藥物複合物來系統性治療複雜疾病的架構:大腸直腸腺瘤的應用★ 以上皮細胞間質化與增生相關功能來描述癌症幹細胞之基因型
★ 從共表達差異基因對導出正常腦老化及因阿茲海默症特定腦區導致在功能性基因途徑與樞紐基因子網絡之變化★ 以疾病進展趨勢挑選基因法識別正常腦老化與阿爾茨海默氏症在特定腦區引發的關鍵功能路徑與調節路徑之變化
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) 阿茲海默症是一種普遍的、隨著時間惡化的人類神經退化性疾病,其主要病徵是因腦中不正常的β澱粉樣蛋白(Aβ)聚集堆積所導致。人們對Aβ致病性的肇因尚不清楚。在過去的幾年中,基於阿茲海默症的Aβ假說而設計的,幾種被認為非常有潛力的抗失智症藥物,包括四種用抗體結合Aβ以防止其聚集的藥物,皆在藥物試驗後期階段失敗。在藥物抗體策略中的研究假設是減少Aβ在細胞間隙中繼續累積。我們使用全基因體的基因表達數據 (取自正常老化腦組織和阿茲海默症病人死後大腦樣本),與過去許多發現完全吻合,提及兩點作為討論: (1) 阿茲海默症致病基因與癌症之間呈現反向表現關係; (2) 微生物感染可能與阿茲海默症有關。我們最重要的推測是蛋白體酶和氧化磷酸化可能是阿茲海默症病患中受損最嚴重的兩個功能;這個狀況在病人大腦的海馬迴和後扣帶回區域最明顯。我們的研究結果顯示在阿茲海默症五個腦區的差異以及與老化的關連性,並推論出可能的標靶基因。這些結果可能有助於應用於阿茲海默症的診斷和治療。
摘要(英) Alzheimer′s disease (AD) is a prevalent progressive neurodegenerative human disease. Several initially highly hopeful anti-AD drugs based on the amyloid-β (Aβ) hypothesis of AD have failed recent late-phase tests, and the cause of AD remains unclear. Natural aging (AG) is a high risk factor for AD. Here, five sets of gene expression microarray data from different regions of AD affected brain, and one of AG, were analyzed for identifying putatively disrupted biological pathways or functions and their abnormal molecular contents. Brain-region specificity among AD cases and AG-AD differences in KEGG-termed function disruption were identified. AG was significantly more at risk to cancer than was AD; hippocampus, the region with the strongest AD signatures, showed no risk to cancer. Our results also showed that microbial related KEGG terms were enriched in five AD affected brain regions, especially in E. coli infection Pathway. Oxidative phosphorylation (OXPHOS; Fisher’s exact test p=1.1E-20) and proteasome (p=2.0E-18) were found to be the most putatively disrupted functions and 24 new target genes were identified. Our results highlighted the heterogeneity of AD in the five brain regions, and indicated a possible AG-AD connection. These may be useful in devising strategies for the early detection of AD. Based on our results we comment on a number of recent late-phase failures of anti-AD drug trials.
關鍵字(中) ★ 阿茲海默症
★ 老化
★ 蛋白質體酶
★ 氧化磷酸化
★ 類澱粉蛋白
★ 細菌感染
關鍵字(英) ★ Alzheimer′s disease
★ aging
★ proteasome
★ oxidative phosphorylation
★ amyloid-β
★ E. coli infection
論文目次 中文摘要 i
Abstract ii
Contents iv
Tables of Contents v
Figures of Contents v
Notation Illustration vi
Chapter 1. Background 1
Chapter 2. Materials and Methods 7
2.1 Gene expression microarray data source 7
2.2 Database on protein-protein interaction 8
2.3 KEGG database on biological functions and pathways 8
2.4 Databases on two type of known AD target genes 8
2.5 Computational software 9
2.6 Quality screening of data and differentially expressed genes (DEGs) 9
2.7 Selection of differentially co-expressed gene (DCE) pairs 9
2.8 Construction of networks from interacting differentially co-expressed gene (IDCE) pairs 10
2.9 Functional Profiling of the DEGs and the IGNs 11
Chapter 3. Results 12
3.1 Properties of curated gene lists 12
3.2 Thirteen genes were conspicuous in the curated gene sets 14
3.3 Five major AD culprit genes were not prominent in the curated gene sets 17
3.4 KEGG terms were enriched heterogeneously in curated gene sets 17
3.5 Oxidative phosphorylation (OXPHOS) pathway (KEGG hsa00190, 130 genes) 21
3.6 Proteasome (KEGG has03050, 47 genes) 21
3.7 Ubiquitin mediated proteolysis pathway (KEGG hsa04120, 137 genes) 21
3.8 Alzheimer’s disease pathway (KEGG has05010, 163 genes) 22
3.9 Pathogenic Escherichia coli infection pathway (KEGG hsa05130, 57 genes) 22
3.10 Pathways in cancer (KEGG hsa05200, 328 genes) 22
3.11 Thirteen genes were “AD-only” hits and ten genes were “double” hits 23
Chapter 4. Discussion 24
Chapter 5. Summary 31
Reference 32
Supplementary Figures 43
Supplementary Tables 46
參考文獻 [1] López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013) The hallmarks of aging. Cell 153, 1194-1217.
[2] Wang J, Zhang S, Wang Y, Chen L, Zhang X-S (2009) Disease-aging network reveals significant roles of aging genes in connecting genetic diseases. PLoS Comput Biol 5, e1000521-e1000521.
[3] La Rosa LR, Matrone C, Ferraina C, Panico MB, Piccirilli S, Di Certo MG, Strimpakos G, Mercuri NB, Calissano P, D′Amelio M (2013) Age-related changes of hippocampal synaptic plasticity in AβPP-null mice are restored by NGF through p75NTR. J. Alzheimers Dis 33, 265-272.
[4] Bukar Maina M, Al-Hilaly YK, Serpell LC (2016) Nuclear Tau and Its Potential Role in Alzheimer’s Disease. Biomolecules 6, 9.
[5] World Alzheimer Report 2015, https://ncdalliance.org/news-events/news/world-alzheimer-report-2015-launched,
[6] 台灣失智症協會 (2004) 機構照顧需求之調查 -長期照護機構 失智症患者之盛行率調查」研究報告.
[7] Organization WH, Top 10 global causes of deaths 2016, http://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death,
[8] Patterson C, Feightner JW, Garcia A, Hsiung G-YR, MacKnight C, Sadovnick AD (2008) Diagnosis and treatment of dementia: 1. Risk assessment and primary prevention of Alzheimer disease. Canadian Medical Association Journal 178, 548-556.
[9] Holth JK, Fritschi SK, Wang C, Pedersen NP, Cirrito JR, Mahan TE, Finn MB, Manis M, Geerling JC, Fuller PM (2019) The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science, eaav2546.
[10] Lester-Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR, de la Monte SM (2006) Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadic Alzheimer′s disease. Journal of Alzheimer′s disease: JAD 9, 13-33.
[11] Wang B-R, Shi J-Q, Ge N-N, Ou Z, Tian Y-Y, Jiang T, Zhou J-S, Xu J, Zhang Y-D (2018) PM2. 5 exposure aggravates oligomeric amyloid beta-induced neuronal injury and promotes NLRP3 inflammasome activation in an in vitro model of Alzheimer’s disease. Journal of neuroinflammation 15, 132.
[12] Hardy JA, Higgins GA (1992) Alzheimer′s disease: the amyloid cascade hypothesis. Science 256, 184.
[13] Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer′s disease: progress and problems on the road to therapeutics. science 297, 353-356.
[14] Kelleher RJ, Shen J (2017) Presenilin-1 mutations and Alzheimer’s disease. Proceedings of the National Academy of Sciences, 201619574.
[15] Cai Y, An SSA, Kim S (2015) Mutations in presenilin 2 and its implications in Alzheimer’s disease and other dementia-associated disorders. Clinical interventions in aging 10, 1163.
[16] Selkoe DJ, Hardy J (2016) The amyloid hypothesis of Alzheimer′s disease at 25 years. EMBO molecular medicine 8, 595-608.
[17] De Marco M, Vallelunga A, Meneghello F, Varma S, Frangi A, Venneri A, Alzheimers DNI (2017) ApoE ε4 allele relateted alterations in hippocampal connectivity in early Alzheimer′s disease support memory performance. Current Alzheimer research.
[18] Hyman B (2016) Tau and Alzheimer pathobiology. Neurobiology of Aging 39, S16.
[19] Bancher C, Brunner C, Lassmann H, Budka H, Jellinger K, Wiche G, Seitelberger F, Grundke-Iqbal I, Iqbal K, Wisniewski H (1989) Accumulation of abnormally phosphorylated τ precedes the formation of neurofibrillary tangles in Alzheimer′s disease. Brain research 477, 90-99.
[20] Engstrom EJ (2007) Researching dementia in Imperial Germany: Alois Alzheimer and the economies of psychiatric practice. Culture, medicine and psychiatry 31, 405-413.
[21] Glenner GG, Wong CW (1984) Alzheimer′s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochemical and biophysical research communications 120, 885-890.
[22] Brion J-P, Flament-Durand J, Dustin P (1986) Alzheimer′s disease and tau proteins. The Lancet 328, 1098.
[23] Kang J, Lemaire H-G, Unterbeck A, Salbaum JM, Masters CL, Grzeschik K-H, Multhaup G, Beyreuther K, Müller-Hill B (1987) The precursor of Alzheimer′s disease amyloid A4 protein resembles a cell-surface receptor. Nature 325, 733.
[24] St George-Hyslop P, Haines J, Rogaev E, Mortilla M, Vaula G, Pericak-Vance M, Foncin J, Montesi M, Bruni A, Sorbi S (1992) Genetic evidence for a novel familial Alzheimer′s disease locus on chromosome 14. Nature genetics 2, 330.
[25] Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small G, Roses AD, Haines J, Pericak-Vance MA (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer′s disease in late onset families. Science 261, 921-923.
[26] Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K (1995) Candidate gene for the chromosome 1 familial Alzheimer′s disease locus. Science 269, 973-977.
[27] Hroudová J, Singh N, Fišar Z, Ghosh KK (2016) Progress in drug development for Alzheimer′s disease: an overview in relation to mitochondrial energy metabolism. European journal of medicinal chemistry 121, 774-784.
[28] Markesbery WR (1997) Oxidative stress hypothesis in Alzheimer′s disease. Free Radical Biology and Medicine 23, 134-147.
[29] Zotova E, Nicoll JA, Kalaria R, Holmes C, Boche D (2010) Inflammation in Alzheimer′s disease: relevance to pathogenesis and therapy. Alzheimers Res Ther 2, 1.
[30] Jack C (2010) The vascular hypothesis of Alzheimer’s disease: bench to bedside and beyond. Neurodegenerative Diseases 7, 116-121.
[31] Wood WG, Li L, Müller WE, Eckert GP (2014) Cholesterol as a causative factor in Alzheimer′s disease: a debatable hypothesis. Journal of neurochemistry 129, 559-572.
[32] Bush AI, Tanzi RE (2008) Therapeutics for Alzheimer′s disease based on the metal hypothesis. Neurotherapeutics 5, 421-432.
[33] Neve RL, McPhie DL (2006) The cell cycle as a therapeutic target for Alzheimer′s disease. Pharmacology & therapeutics 111, 99-113.
[34] Cummings J, Lee G, Ritter A, Zhong K (2018) Alzheimer′s disease drug development pipeline: 2018. Alzheimer′s & Dementia: Translational Research & Clinical Interventions.
[35] Extance A (2010) Alzheimer′s failure raises questions about disease-modifying strategies. Nature Reviews Drug Discovery 9, 749.
[36] Philippidis A (2019) Unlucky 13: Top Clinical Trial Failures of 2018: Biopharmas pursue costly studies despite low success rates. Genetic Engineering & Biotechnology News 39, 14, 16.
[37] Mullard A (2017) New plaque psoriasis approval carries suicide warning. Nature Reviews Drug Discovery 16, 155-155.
[38] Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, Raskind M, Sabbagh M, Honig LS, Porsteinsson AP, Ferris S (2014) Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer′s disease. New England Journal of Medicine 370, 322-333.
[39] Mullard A (2017) Alzheimer amyloid hypothesis lives on. Nature Reviews Drug Discovery 16, 3-5.
[40] Panza F, Lozupone M, Seripa D, Imbimbo BP (2019) Amyloid‐β Immunotherapy for Alzheimer′s Disease–Is It Now A Long Shot…? Annals of neurology.
[41] Cao W (2019) Beyond Aducanumab and the Amyloid Hypothesis. J Neuroinflamm Neurodegener Dis 3, 100008.
[42] Hardy JA, Higgins GA (1992) Alzheimer′s disease: the amyloid cascade hypothesis. Science 256, 184-186.
[43] Semagacestat | ALZFORUM, https://www.alzforum.org/therapeutics/semagacestat,
[44] Verubecestat | ALZFORUM, https://www.alzforum.org/therapeutics/verubecestat,
[45] Atabecestat | ALZFORUM, https://www.alzforum.org/therapeutics/atabecesta,
[46] Bapineuzumab | ALZFORUM, https://www.alzforum.org/therapeutics/bapineuzumab,
[47] Solanezumab | ALZFORUM, https://www.alzforum.org/therapeutics/solanezumab,
[48] Crenezumab | ALZFORUM, https://www.alzforum.org/therapeutics/crenezumab,
[49] Aducanumab | ALZFORUM, https://www.alzforum.org/therapeutics/aducanumab,
[50] Kikuchi M, Ogishima S, Mizuno S, Miyashita A, Kuwano R, Nakaya J, Tanaka H (2016) Network-Based Analysis for Uncovering Mechanisms Underlying Alzheimer’s Disease. Systems Biology of Alzheimer′s Disease, 479-491.
[51] Chu G, Narasimhan B, Tibshirani R, Tusher V (2002) SAM,“Significance Analysis of Microarrays”: Users Guide and Technical Document. Stanford University.
[52] Avramopoulos D, Szymanski M, Wang R, Bassett S (2011) Gene expression reveals overlap between normal aging and Alzheimer′s disease genes. Neurobiology of aging 32, 2319. e2327-2319. e2334.
[53] Swerdlow RH (2011) Brain aging, Alzheimer′s disease, and mitochondria. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1812, 1630-1639.
[54] Sekar S, McDonald J, Cuyugan L, Aldrich J, Kurdoglu A, Adkins J, Serrano G, Beach TG, Craig DW, Valla J (2015) Alzheimer′s disease is associated with altered expression of genes involved in immune response and mitochondrial processes in astrocytes. Neurobiology of aging 36, 583-591.
[55] Hong L, Huang H-C, Jiang Z-F (2014) Relationship between amyloid-beta and the ubiquitin-proteasome system in Alzheimer′s disease. Neurological research 36, 276-282.
[56] Amar D, Safer H, Shamir R (2013) Dissection of regulatory networks that are altered in disease via differential co-expression. PLoS Comput Biol 9, e1002955.
[57] Berchtold NC, Coleman PD, Cribbs DH, Rogers J, Gillen DL, Cotman CW (2013) Synaptic genes are extensively downregulated across multiple brain regions in normal human aging and Alzheimer′s disease. Neurobiology of aging 34, 1653-1661.
[58] Wang X, Michaelis ML, Michaelis EK (2010) Functional genomics of brain aging and Alzheimer’s disease: focus on selective neuronal vulnerability. Current genomics 11, 618.
[59] Saetre P, Jazin E, Emilsson L (2011) Age‐related changes in gene expression are accelerated in Alzheimer′s disease. Synapse 65, 971-974.
[60] Barrett T, Troup DB, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM (2011) NCBI GEO: archive for functional genomics data sets—10 years on. Nucleic acids research 39, D1005-D1010.
[61] Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Walker DG, Caselli RJ, Kukull WA, McKeel D, Morris JC (2007) Gene expression profiles in anatomically and functionally distinct regions of the normal aged human brain. Physiological genomics 28, 311-322.
[62] Liang WS, Reiman EM, Valla J, Dunckley T, Beach TG, Grover A, Niedzielko TL, Schneider LE, Mastroeni D, Caselli R (2008) Alzheimer′s disease is associated with reduced expression of energy metabolism genes in posterior cingulate neurons. Proceedings of the National Academy of Sciences 105, 4441-4446.
[63] Lu T, Aron L, Zullo J, Pan Y, Kim H, Chen Y, Yang T-H, Kim H-M, Drake D, Liu XS (2014) REST and stress resistance in ageing and Alzheimer/′s disease. Nature 507, 448-454.
[64] Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic acids research, gkv007.
[65] Brodmann K (1925) Vergleichende Lokalisationslehre der Grosshirnrinde in ihren Prinzipien dargestellt auf Grund des Zellenbaues, Barth.
[66] Prasad TK, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A (2009) Human protein reference database—2009 update. Nucleic acids research 37, D767-D772.
[67] Consortium U (2014) Activities at the universal protein resource (UniProt). Nucleic acids research 42, 7486.
[68] Da Wei Huang BTS, Lempicki RA (2008) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols 4, 44-57.
[69] Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE (2007) Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nature genetics 39, 17-23.
[70] Bai Z, Han G, Xie B, Wang J, Song F, Peng X, Lei H (2016) AlzBase: an integrative database for gene dysregulation in Alzheimer’s disease. Molecular neurobiology 53, 310-319.
[71] Wang K, Narayanan M, Zhong H, Tompa M, Schadt EE, Zhu J (2009) Meta-analysis of inter-species liver co-expression networks elucidates traits associated with common human diseases. PLoS Comput Biol 5, e1000616.
[72] Hou C, Wang Y, Liu J, Wang C, Long J (2017) Neurodegenerative disease related proteins have negative effects on SNARE-mediated membrane fusion in pathological confirmation. Frontiers in molecular neuroscience 10, 66.
[73] Okamoto M, Gray JD, Larson CS, Kazim SF, Soya H, McEwen BS, Pereira AC (2018) Riluzole reduces amyloid beta pathology, improves memory, and restores gene expression changes in a transgenic mouse model of early-onset Alzheimer’s disease. Translational psychiatry 8.
[74] Sonntag K-C, Ryu W-I, Amirault KM, Healy RA, Siegel AJ, McPhie DL, Forester B, Cohen BM (2017) Late-onset Alzheimer’s disease is associated with inherent changes in bioenergetics profiles. Scientific reports 7, 14038.
[75] Xu J, Patassini S, Rustogi N, Riba-Garcia I, Hale BD, Phillips AM, Waldvogel H, Haines R, Bradbury P, Stevens A (2019) Regional protein expression in human Alzheimer’s brain correlates with disease severity. Communications biology 2, 43.
[76] LABUDOVA O, KITZMUELLER E, Hermann R, CAIRNS N, LUBEC G (1999) Increased phosphoglycerate kinase in the brains of patients with Down′s syndrome but not with Alzheimer′s disease. Clinical Science 96, 279-285.
[77] El Kadmiri N, Slassi I, El Moutawakil B, Nadifi S, Tadevosyan A, Hachem A, Soukri A (2014) Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Alzheimer′s disease. Pathologie Biologie 62, 333-336.
[78] Li X, Zhou J, Chen H, Wang F, Mei Q, Sun H (2017).
[79] Nordestgaard LT, Tybjærg-Hansen A, Nordestgaard BG, Frikke-Schmidt R (2015) Loss-of-function mutation in ABCA1 and risk of Alzheimer′s disease and cerebrovascular disease. Alzheimer′s & Dementia 11, 1430-1438.
[80] Liu S-L, Wang C, Jiang T, Tan L, Xing A, Yu J-T (2016) The role of Cdk5 in Alzheimer’s disease. Molecular neurobiology 53, 4328-4342.
[81] Llorens-Marítin M, Jurado J, Hernández F, Ávila J (2014) GSK-3β, a pivotal kinase in Alzheimer disease. Frontiers in molecular neuroscience 7, 46.
[82] Fowler KD, Funt JM, Artyomov MN, Zeskind B, Kolitz SE, Towfic F (2015) Leveraging existing data sets to generate new insights into Alzheimer’s disease biology in specific patient subsets. Scientific reports 5.
[83] Logotheti M, Papadodima O, Venizelos N, Chatziioannou A, Kolisis F (2013) A comparative genomic study in schizophrenic and in bipolar disorder patients, based on microarray expression profiling meta-analysis. The Scientific World Journal 2013.
[84] Greber S, Lubec G, Cairns N, Fountoulakis M (1999) Decreased levels of synaptosomal associated protein 25 in the brain of patients with Down syndrome and Alzheimer′s disease. Electrophoresis 20, 928-934.
[85] Arendt T (2009) Synaptic degeneration in Alzheimer’s disease. Acta neuropathologica 118, 167-179.
[86] Brinkmalm A, Brinkmalm G, Honer WG, Frölich L, Hausner L, Minthon L, Hansson O, Wallin A, Zetterberg H, Blennow K (2014) SNAP-25 is a promising novel cerebrospinal fluid biomarker for synapse degeneration in Alzheimer’s disease. Molecular neurodegeneration 9, 53.
[87] Erika B, Francis PT, Howlett DR, Pereira JB, Höglund K, Bogstedt A, Cedazo-Minguez A, Baek J-H, Tibor H, Attems J (2016) Synaptic proteins predict cognitive decline in Alzheimer′s disease and Lewy body dementia.
[88] Upadhya SC, Hegde AN (2007) Role of the ubiquitin proteasome system in Alzheimer′s disease. BMC biochemistry 8, S12.
[89] Ragland M, Hutter C, Zabetian C, Edwards K (2009) Association between the ubiquitin carboxyl-terminal esterase L1 gene (UCHL1) S18Y variant and Parkinson′s Disease: a HuGE review and meta-analysis. American journal of epidemiology 170, 1344-1357.
[90] McNaught KSP, Olanow CW, Halliwell B, Isacson O, Jenner P (2001) Failure of the ubiquitin–proteasome system in Parkinson′s disease. Nature Reviews Neuroscience 2, 589-594.
[91] Tramutola A, Di Domenico F, Barone E, Perluigi M, Butterfield DA (2016) It is all about (U) biquitin: role of altered ubiquitin-proteasome system and UCHL1 in Alzheimer Disease. Oxidative medicine and cellular longevity 2016.
[92] Zhang M, Cai F, Zhang S, Zhang S, Song W (2014) Overexpression of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) delays Alzheimer′s progression in vivo. Scientific reports 4, 7298.
[93] Itala M, Helenius H, Nikoskelainen J, Remes K (1992) Infections and serum IgG levels in patients with chronic lymphocytic leukemia. European journal of haematology 48, 266-270.
[94] Lue LF, Walker DG (2002) Modeling Alzheimer′s disease immune therapy mechanisms: Interactions of human postmortem microglia with antibody‐opsonized amyloid beta peptide. Journal of neuroscience research 70, 599-610.
[95] Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y (2016) The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 537, 50-56.
[96] Kadavath H, Hofele RV, Biernat J, Kumar S, Tepper K, Urlaub H, Mandelkow E, Zweckstetter M (2015) Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proceedings of the National Academy of Sciences 112, 7501-7506.
[97] Baumgartner R, Umlauf E, Veitinger M, Guterres S, Rappold E, Babeluk R, Mitulović G, Oehler R, Zellner M (2013) Identification and validation of platelet low biological variation proteins, superior to GAPDH, actin and tubulin, as tools in clinical proteomics. Journal of proteomics 94, 540-551.
[98] Kananura C, Haug K, Sander T, Runge U, Gu W, Hallmann K, Rebstock J, Heils A, Steinlein OK (2002) A splice-site mutation in GABRG2 associated with childhood absence epilepsy and febrile convulsions. Archives of neurology 59, 1137-1141.
[99] Li Y, Sun H, Chen Z, Xu H, Bu G, Zheng H (2016) Implications of GABAergic neurotransmission in Alzheimer’s disease. Frontiers in aging neuroscience 8, 31.
[100] Limon A, Reyes-Ruiz JM, Miledi R (2012) Loss of functional GABAA receptors in the Alzheimer diseased brain. Proceedings of the National Academy of Sciences 109, 10071-10076.
[101] Muslin AJ, Tanner JW, Allen PM, Shaw AS (1996) Interaction of 14-3-3 with signaling proteins is mediated by the recognition of phosphoserine. Cell 84, 889-897.
[102] Mateo I, Llorca J, Infante J, Rodríguez‐Rodríguez E, Berciano J, Combarros O (2008) Gene–gene interaction between 14‐3‐3 zeta and butyrylcholinesterase modulates Alzheimer′ s disease risk. European journal of neurology 15, 219-222.
[103] Mateo I, Sánchez-Juan P, Rodríguez-Rodríguez E, Infante J, Fernández-Viadero C, Peña N, Berciano J, Combarros O (2008) 14-3-3 zeta and tau genes interactively decrease Alzheimer’s disease risk. Dementia and geriatric cognitive disorders 25, 317-320.
[104] Qureshi HY, Li T, MacDonald R, Cho CM, Leclerc N, Paudel HK (2013) Interaction of 14-3-3ζ with Microtubule-Associated Protein Tau within Alzheimer’s Disease Neurofibrillary Tangles. Biochemistry 52, 6445-6455.
[105] Lin Z, Zhao D, Wang Y, Zhao W, Yin X, Zhou X, Zhang Z, Yang L (2015) Downregulation of β-Soluble N-Ethylmaleimide-Sensitive Factor Attachment Protein: Proteomics-Based Identification in Early-Stage Prion Disease. Neurodegenerative Diseases 15, 193-201.
[106] Yoo BC, Cairns N, Fountoulakis M, Lubec G (2001) Synaptosomal proteins, beta-soluble N-ethylmaleimide-sensitive factor attachment protein (beta-SNAP), gamma-SNAP and synaptotagmin I in brain of patients with Down syndrome and Alzheimer’s disease. Dementia and geriatric cognitive disorders 12, 219-225.
[107] Hirokawa N, Niwa S, Tanaka Y (2010) Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron 68, 610-638.
[108] Liu Z-D, Zhang S, Hao J-J, Xie T-R, Kang J-S (2016) Cellular model of neuronal atrophy induced by DYNC1I1 deficiency reveals protective roles of RAS-RAF-MEK signaling. Protein & Cell 7, 638-650.
[109] Wu F, Yao PJ (2009) Clathrin-mediated endocytosis and Alzheimer′s disease: an update. Ageing research reviews 8, 147-149.
[110] Liu QY, Lei JX, Sikorska M, Liu R (2008) A novel brain-enriched E3 ubiquitin ligase RNF182 is up regulated in the brains of Alzheimer′s patients and targets ATP6V0C for degradation. Molecular neurodegeneration 3, 4.
[111] Makin S (2018) The amyloid hypothesis on trial. Nature 559, S4.
[112] Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, Raman R, Sun X, Aisen PS (2014) Phase 3 trials of solanezumab for mild-to-moderate Alzheimer′s disease. New England Journal of Medicine 370, 311-321.
[113] Tayeb HO, Murray ED, Price BH, Tarazi FI (2013) Bapineuzumab and solanezumab for Alzheimer′s disease: is the ‘amyloid cascade hypothesis′ still alive? Expert opinion on biological therapy 13, 1075-1084.
[114] Hawkes N (2017) Merck ends trial of potential Alzheimer′s drug verubecestat. BMJ: British Medical Journal (Online) 356.
[115] Kennedy ME, Stamford AW, Chen X, Cox K, Cumming JN, Dockendorf MF, Egan M, Ereshefsky L, Hodgson RA, Hyde LA (2016) The BACE1 inhibitor verubecestat (MK-8931) reduces CNS β-amyloid in animal models and in Alzheimer’s disease patients. Science translational medicine 8, 363ra150-363ra150.
[116] Phillips NR, Simpkins JW, Roby RK (2014) Mitochondrial DNA deletions in Alzheimer′s brains: A review. Alzheimer′s & Dementia 10, 393-400.
[117] Lecker SH, Goldberg AL, Mitch WE (2006) Protein degradation by the ubiquitin–proteasome pathway in normal and disease states. Journal of the American Society of Nephrology 17, 1807-1819.
[118] Speese SD, Trotta N, Rodesch CK, Aravamudan B, Broadie K (2003) The ubiquitin proteasome system acutely regulates presynaptic protein turnover and synaptic efficacy. Current Biology 13, 899-910.
[119] Hong L, Huang H-C, Jiang Z-F (2014) Taylor & Francis.
[120] Chondrogianni N, Gonos ES (2007) Overexpression of hUMP1/POMP proteasome accessory protein enhances proteasome-mediated antioxidant defence. Experimental gerontology 42, 899-903.
[121] Chen Y, Neve RL, Liu H (2012) Neddylation dysfunction in Alzheimer′s disease. Journal of cellular and molecular medicine 16, 2583-2591.
[122] Mori F, Nishie M, Piao YS, Kito K, Kamitani T, Takahashi H, Wakabayashi K (2005) Accumulation of NEDD8 in neuronal and glial inclusions of neurodegenerative disorders. Neuropathology and applied neurobiology 31, 53-61.
[123] Krishnan V, Stoppel DC, Nong Y, Johnson MA, Nadler MJ, Ozkaynak E, Teng BL, Nagakura I, Mohammad F, Silva MA (2017) Autism gene Ube3a and seizures impair sociability by repressing VTA Cbln1. Nature 543, 507-512.
[124] Hill JM, Clement C, Pogue AI, Bhattacharjee S, Zhao Y, Lukiw WJ (2014) Pathogenic microbes, the microbiome, and Alzheimer’s disease (AD). Frontiers in aging neuroscience 6, 127.
[125] Kumar DKV, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, Lefkowitz A, McColl G, Goldstein LE, Tanzi RE (2016) Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Science translational medicine 8, 340ra372-340ra372.
[126] Boyken J, Grønborg M, Riedel D, Urlaub H, Jahn R, Chua JJE (2013) Molecular profiling of synaptic vesicle docking sites reveals novel proteins but few differences between glutamatergic and GABAergic synapses. Neuron 78, 285-297.
[127] Musicco M, Adorni F, Di Santo S, Prinelli F, Pettenati C, Caltagirone C, Palmer K, Russo A (2013) Inverse occurrence of cancer and Alzheimer disease A population-based incidence study. Neurology 81, 322-328.
[128] Ibáñez K, Boullosa C, Tabarés-Seisdedos R, Baudot A, Valencia A (2014) Molecular evidence for the inverse comorbidity between central nervous system disorders and cancers detected by transcriptomic meta-analyses. PLoS genetics 10, e1004173.
[129] Shafi O (2016) Inverse relationship between Alzheimer’s disease and cancer, and other factors contributing to Alzheimer’s disease: a systematic review. BMC neurology 16, 236.
[130] Benit P, Slama A, Cartault F, Giurgea I, Chretien D, Lebon S, Marsac C, Munnich A, Rötig A, Rustin P (2004) Mutant NDUFS3 subunit of mitochondrial complex I causes Leigh syndrome. Journal of medical genetics 41, 14-17.
[131] Nishioka K, Vilariño-Güell C, Cobb SA, Kachergus JM, Ross OA, Hentati E, Hentati F, Farrer MJ (2010) Genetic variation of the mitochondrial complex I subunit NDUFV2 and Parkinson’s disease. Parkinsonism & related disorders 16, 686-687.
[132] Föcking M, Dicker P, Lopez LM, Hryniewiecka M, Wynne K, English JA, Cagney G, Cotter DR (2016) Proteomic analysis of the postsynaptic density implicates synaptic function and energy pathways in bipolar disorder. Translational psychiatry 6, e959.
[133] Ai J, Sun L-H, Che H, Zhang R, Zhang T-Z, Wu W-C, Su X-L, Chen X, Yang G, Li K (2013) MicroRNA-195 protects against dementia induced by chronic brain hypoperfusion via its anti-amyloidogenic effect in rats. Journal of Neuroscience 33, 3989-4001.
[134] Papadopoulos GL, Reczko M, Simossis VA, Sethupathy P, Hatzigeorgiou AG (2008) The database of experimentally supported targets: a functional update of TarBase. Nucleic acids research 37, D155-D158.
[135] Chatr-Aryamontri A, Breitkreutz B-J, Heinicke S, Boucher L, Winter A, Stark C, Nixon J, Ramage L, Kolas N, O’Donnell L (2012) The BioGRID interaction database: 2013 update. Nucleic acids research 41, D816-D823.
[136] Chang Y, Paramasivam M, Girgenti MJ, Walikonis RS, Bianchi E, LoTurco JJ (2010) RanBPM regulates the progression of neuronal precursors through M‐phase at the surface of the neocortical ventricular zone. Developmental neurobiology 70, 1-15.
[137] Tufail Y, Cook D, Fourgeaud L, Powers CJ, Merten K, Clark CL, Hoffman E, Ngo A, Sekiguchi KJ, O’Shea CC (2017) Phosphatidylserine exposure controls viral innate immune responses by microglia. Neuron 93, 574-586. e578.
[138] Rojiani MV, Alidina J, Esposito N, Rojiani AM (2010) Expression of MMP-2 correlates with increased angiogenesis in CNS metastasis of lung carcinoma. International journal of clinical and experimental pathology 3, 775.
[139] Friedrich RP, Tepper K, Rönicke R, Soom M, Westermann M, Reymann K, Kaether C, Fändrich M (2010) Mechanism of amyloid plaque formation suggests an intracellular basis of Aβ pathogenicity. Proceedings of the National Academy of Sciences 107, 1942-1947.
[140] Salon ML, Pasquini L, Moreno MB, Pasquini J, Soto E (2003) Relationship between β-amyloid degradation and the 26S proteasome in neural cells. Experimental neurology 180, 131-143.
[141] Almeida CG, Takahashi RH, Gouras GK (2006) β-Amyloid accumulation impairs multivesicular body sorting by inhibiting the ubiquitin-proteasome system. Journal of Neuroscience 26, 4277-4288.
[142] Saido T, Leissring MA (2012) Proteolytic degradation of amyloid β-protein. Cold Spring Harbor perspectives in medicine 2, a006379.
指導教授 李弘謙(Hoong-Chien Lee) 審核日期 2019-8-22
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