博碩士論文 972404002 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:13 、訪客IP:3.15.221.67
姓名 黃暐捷(Wei-Chieh Huang)  查詢紙本館藏   畢業系所 生命科學系
論文名稱 抑制口腔癌和乳癌細胞轉移的微型核醣核酸
(MicroRNAs acting as potent metastasis suppressors in oral cancer and breast cancer)
相關論文
★ Thirst control of water-seeking behavior in Drosophila★ KLHL17在癲癇與自閉症中之角色
★ MyoD對於PGC-1α 基因表現之調控機制★ 雄性素受體對於肌肉前驅細胞決定的功用
★ Nanog和Oct4表現對肌肉分化之影響★ 大量表現幹細胞專有轉錄因子抑制肌肉細胞走向分化
★ FOXOs 轉錄調控因子家族對肌肉細胞末期分化的影響★ 大量表現 Oct4 與 Nanog 抑制肌纖維母細胞 C2C12 分化
★ 在終極肌肉分化時,肌肉性bHLH轉錄因子對PGC-1α的調控★ FoxOs 大量表現對肌肉細胞末期分化的影響
★ 觀察肌肉生成轉錄因子如何調控 M- 和N- cadherin 表現★ Oc4和Nanog共同抑制末端肌肉分化
★ FoxO6在肌原母細胞中的代謝及分化中所扮演的角色★ PGC-1α 與 Stra13 間之交互作用
★ 探討大量表現 FoxO6 對肌肉終極分化的影響以及尋找 FoxO6 蛋白質在 PGC-1 alpha 啟動子上的結合位★ 探討丙戊酸 (Valporic acid) 於肌肉細胞中活化 Oct4 promoter 的機制
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) 轉移是癌症患者治療失敗的主要臨床因數。微RNA分子在癌症進展和轉移扮演一個重要的角色。此項研究是深入探討微RNA分子與高侵入性的口腔癌及乳癌細胞之間的關係。我們分別使用了體外和體內的篩選方式建立了高度侵入性的口腔鱗狀細胞癌(OSCC)及乳癌細胞,並且藉由微小RNA微陣列技術分析這些高度轉移細胞株與其低侵入性母代細胞株的轉錄圖譜。我們以基因表現差異兩倍為門檻,並且使用gene ontology軟體分析找出與侵入相關的微RNA分子以及其下游標的基因。研究顯示所篩選出的全部高侵入口腔癌細胞株及乳癌細胞株其miR-491-5p或miR-149分別表現顯著減少。同時研究也顯示當miR-491-5p或miR-149分別大量表達在高侵入性口腔癌或乳癌細胞時會抑制其爬動和侵入能力,在老鼠模式中也發現大量表達這些微RNA分子會抑制癌細胞轉移的能力。藉由3′UTR報告基因分析證實G protein-coupled receptor kinase-interacting protiein 1(GIT1)同時是miR-491-5p及miR-149的目標基因。另一方面在口腔癌細胞中大量表達GIT1可以回復miR-491-5p及miR-149所抑制的爬動、侵入及轉移的能力。若抑制GIT1的表現則會抑制口腔癌細胞的爬動、侵入和肺轉移的能力。這個研究也證實miR-491-5p藉由抑制GIT1的表現進而減少口腔癌細胞的focal adhesion,同時導致paxillin蛋白的降解及減少paxillin、FAK及EGF/EGFR調控ERK1/2的活性,也抑制了MMP2/9的表現量與活性。在乳癌方面,miR-149同樣藉由抑制GIT1的表現進而促使paxillin及α5β1 integrin蛋白分別走向proteasome及lysosome的分解路徑。此外,我們發現miR-491-5p和FOCAD同時位於染色體9p21.3,我們的實驗證明miR-491-5p是一個intronic微RNA分子,其座落在FOCAD的intron 4。實驗證明當FOCAD的表現被抑制時也會導致口腔癌細胞癌轉移及侵入的能力減少。進一步我們也證實FOCAD與miR-491-5p會一同被表現出來,並且藉由抑制GIT1調控路徑來減少口腔癌細胞的爬動、侵入和轉移的能力。因此,我們的結論是miR-491-5p及miR-149可以同時藉由GIT1的調控路徑有效的抑制口腔癌及乳癌細胞的轉移能力,這些發現暗示著本研究探討的調控機制可能成為臨床診斷和治療的標的。
摘要(英) Metastasis is an important clinical parameter for patient prognosis and the major cause of treatment failure for cancer. MicroRNAs are molecules that could play an abstrusive role in cancer progression and metastasis. The study is to identify relevant microRNAs associated with invasive phenotype of oral and breast cancers. We have established isogenic highly invasive oral squamous cell carcinoma (OSCC) lines and breast cancer lines from their respective low invasive parental lines via in vitro and in vivo selection protocols. MicroRNA array analysis was used for transcriptome profiling between each pair of the parental and the highly invasive subline. Using threshold of 2-fold change of gene expression, we analyzed the microarray data by gene ontology enrichment of Partek and identify genes as well as microRNAs revealed to be significantly associated with invasive phenotype. We found that miR-491-5p and miR-149 level were significantly decreased in the selected OSCC invasive lines and highly invasive breast cancer lines respectively. Overexpression of miR-491-5p or miR-149 in those highly invasive cells suppressed their migration/invasion in vitro and metastatic ability in a xenograft mouse model. The G protein-coupled receptor kinase-interacting protiein 1 (GIT1) is a direct target of miR-491-5p and miR-149 as revealed by 3′UTR reporter assays. The miR-491-5p- and miR-149-mediated inhibition of migration/invasion and lung metastasis could be rescued by overexpression of GIT1. Depletion of GIT1 inhibited migration/invasion and lung metastasis of OSCC and breast cancer cells. MiR-491-5p-mediated GIT1 repression reduced focal adhesion and concurrently decreased steady state levels of paxillin, phospho-paxillin,phospho-FAK, EGF/EGFR-mediated ERK1/2 activation as well as decreased MMP2/9 levels and activities in OSCC cells. Overexpression of miR-149 or depletion of GIT1 led to enhanced protein degradation of paxillin and α5β1 integrin via proteasome and lysosome pathways respectively. In addition, we found that miR-491-5p and focadhesin (KIAA1797/FOCAD) gene are located on chromosome 9p21.3 together. Our data suggest that miR-491-5p is an intronic miRNA processed form FOCAD intron 4 rather than being transcribed as a separate RNA. Depletion of FOCAD promoted cell migration/invasion abilities in human oral cancer cells. Furthermore, FOCAD/miR-491-5p were co-expressed in OSCC cells and they suppresses OSCC cell migration/invasion and metastasis, suggesting potential application of the miR-491-5p/GIT1 pathways in OSCC prognosis and therapy. Therefore, we conclude that miR-491-5p and miR-149 suppresses migration/invasion and metastasis of OSCC and breast cancer cells, respectively, by targeting GIT1, suggesting potential application of the miR-491-5p/GIT1 and miR-149/GIT1 pathways in clinical diagnosis and therapeutics.
關鍵字(中) ★ 口腔癌
★ 乳癌
★ 轉移
★ 微型核醣核酸
★ 局部黏著斑激酶
★ 口腔鱗狀上皮細胞癌
關鍵字(英) ★ oral cancer
★ breast cancer
★ metastasis
★ microRNA
★ focal adhesion kinase
★ OSCC
論文目次 Table of contents
Declaration I
Publications arising from this thesis II
(A). Referred papers: II
(B). Abstracts presented in meetings: II
中文摘要 IV
Abstract VI
Acknowledgments VIII
Table of contents XI
Abbreviation XVII
Chapter I: Introduction 1
Epidemiology of oral cancer and breast cancer 1
Neck lymph node metastasis of oral squamous cell carcinoma 2
Breast cancer metastasis 2
microRNA biogenesis and nomenclature 3
The role of microRNAs in oral cancer and breast cancer invasion and metastasis 5
Regulation of focal adhesion complexs in cancer cell migration, invasion and metastasis 6
Figures 11
Figure I-1. The microRNA processing pathways and their regulation 11
Figure I-2. GW (glycine-tryptophan repeats) proteins are recruited to mRNA via direct interaction with the slicer activity (miRNA–AGO complex) (49). 13
Figure I-3. Schematic of a miRNA precursor 14
Figure I-4. Focal adhesion components 15
Figure I-5. GIT1 domain structure 16
Figure I-6. Summary of the GIT1 protein interaction and signaling pathways 17
Figure I-7. Focal adhesion kinase (FAK)-SRC-ERK-paxillin signaling regulating cell migration and focal adhesion formation 18
Figure I-8. The model showing that FOCAD is a novel component of the focal adhesion complex. 20
Chapter II: Materials and Methods 21
Tissue samples acquisition. 21
Cell culture, DNA and RNA transfections and stable cell line generation. 21
Vectors, antibodies and reagents. 23
3′UTR reporter assays. 24
Reverse transcriptase (RT)-PCR and qRT-PCR. 25
Cell chemotatic migration and invasion assay. 25
In vivo metastasis assays. 26
Focal adhesion assay and Immunofluorescence microscopy. 27
Western blotting 27
Paxillin degradation assays 28
Immunohistochemistry (IHC) and Fluorescence in situ hybridization (FISH). 28
Gelatin zymography 29
Transfection efficiency of miRNAs 29
Statistical analysis. 30
Figure 31
Figure II. The predicted highly conserved miR-491-5p and miR-149 target sequences located in the 3′UTR of GIT1 mRNA. 31
Chapter III: Functional role and clinical evidence of microRNA-491-5p in oral cancer metastasis 32
Rationale 32
Results 34
Establishment of isogenic pairs of high and low invasive OSCC lines. 34
Down-regulation of miR-491-5p was found in the highly invasive OSCC cells and was correlated with poor survival of OSCC patients. 35
The miR-491-5p inhibits migration, invasion and lung metastasis of OSCC cells. 36
GIT1 is a direct target of miR-491-5p. 37
Suppression of GIT1 by its siRNA inhibits OSCC cell migration, invasion and metastasis. 38
Re-expression of GIT1 significantly reverses miR-491-5p-mediated suppression of invasion and metastasis of C9-lung-IV2 cells. 39
Inhibtion of GIT1 by miR-491-5p enhances degradation of paxillin and impaires focal adhesion signaling in OSCC cells. 39
Dominant-negative form of FAK (DN-FAK) partially inhibits OSCC cells invasiveness which could be rescued by GIT1. 41
miR-491-5p targets GIT1 to regulate expression level and activity of Matrix metalloproteinases2/9 (MMP2/9) via EGFR/ERK1/2 signaling pathway . 41
Correlation of miR-491-5p and GIT1 expression with grades of OSCC and inverse correlation between miR-491-5p and GIT1 . 43
Discussion: 44
Figures 50
Figure III-1. Selection of highly invasive oral cancer cells. 50
Figure III-2. The miR-491-5p expression is downregulated in highly invasive OSCC lines and oral cancers. 52
Figure III-3. The miR-491-5p inhibits migration, invasion and metastasis of OSCC cells. 54
Figure III-4. Overexpressing miR-491-5p inhibits lung metastasis in SCID mice. 56
Figure III-5. Identification of GIT1 as the direct target of miR-491-5p. 57
Figure III-6. The GIT1 mRNA level is upregulated in highly invasive OSCC lines. 59
Figure III-7. GIT1 is overexpressed in the highly invasive oral cancer lines. 60
Figure III-8. GIT1 expression correlates with lymph node metastasis and knockdown of GIT1 inhibits OSCC cell migration, invasion and metastasis. 61
Figure III-9. GIT1 depletion enhances degradation of paxillin, reduces FAK phosphorylation and decreases focal adhesion formation. 63
Figure III-10. Knockdown of GIT1 by siRNA-3 has no effect on paxillin mRNA expression. 65
Figure III-11. DN-FAK suppresses GIT1-induced OSCC cell migration and invasion. 66
Figure III-12. Effect of GIT1 on activation and expression of MMP2/9 and on EGF-induced ERK1/2 phosphorylation. 67
Figure III-13. Effect of ERK1/2 on MMP2/9 mRNA expression in OSCC cells. 69
Figure III-14. Overexpression of miR-491-5p has no effect on proliferation of OSCC cells measured up to 72h. 71
Figure III-15. Effect of the DN-MEK or PD98059 on GIT1 induced MMP2/9 mRNA expression in OSCC cells. 73
Figure III-16. miR-491-5p and GIT1 expression levels exhibits an inverse relationship in OSCC specimens. 74
Figure III-17. Depletion of FAK and GIT1 decreases the paxillin and FAK phosphorylation. 76
Figure III-18. CGHNC9 cells transfected with Fluorescein amidite (FAM)-labeled miR-491-5p. 77
Tables 78
Table III-1: Clinicopathologic features of OSCC patients 78
Table III-2: The list of primers used in oral cancer study 79
Table III-3: Venn digram showing miR-491-5p targets derived from the TargetScan and DIANA-microT 80
Chapter IV: The role of microRNA-149 in migration, invasion and metastasis of breast cancer 81
Rationale 81
Results 83
MiRNA microarray profiling identified miRNAs differentially expressed in the in vivo-selected metastatic breast cancer cells versus their parental cells. 83
miR-149 suppresses breast cancer cell migration and invasion in vitro 84
miR-149 functions as a metastasis suppressor in vivo 84
GIT1 is a direct target of miR-149 85
Low miR-149 and high GIT1 level correlates with the invasive phenotype of breast cancer cell lines 86
Knockdown of the endogenous GIT1 expression by siRNAs impairs cell migration and invasion 87
Re-expression of GIT1 partially reverses miR-149-mediated suppression of migration and invasion 88
Re-expression of GIT1 partially rescues miR-149-suppressed lung metastasis 88
Inhibition of GIT1 expression by miR-149 or GIT1 siRNAs impairs focal adhesions and focal adhesion signaling, which is significantly rescued by re-expression of GIT1 89
Depletion of GIT1 leads to enhanced proteasome-mediated degradation of paxillin 90
Depletion of GIT1 leads to enhanced lysosome-mediated protein degradation of α5β1 integrin 91
Depletion of GIT1 impairs α5β1-mediated cell adhesion to fibronectin and collagen 93
miR-149 and GIT1 expression significantly correlates with clinical stages and lymph node metastasis of breast cancer 93
Discussion 95
Figures 99
Figure IV-1. Schematics for the establishment of highly metastatic breast cancer cells using a mouse model. 99
Figure IV-2. Metastatic IV2 sublines exhibit increased invasive ability as compared to the parental 231 cells. 100
Figure IV-3. Whole organ photographs and histological analysis of lung metastases generated by parental 231 and IV2-1 cells. 101
Figure IV-4. Hierarchical clustering of miRNA expression among the parental 231 cells and three metastatic IV2 sublines, IV2-1, IV2-2 and IV2-3, reveals a set of differentially expressed miRNAs. 102
Figure IV-5. Taqman qRT-PCR validation of selected candidate miRNAs. 103
Figure IV-6. miR-149 is down-regulated in highly metastatic IV2 sublines and suppresses breast cancer cell migration, invasion and metastasis. 104
Figure IV-7. Overexpression of pre-miR-149 has no effect on proliferation of the IV2-1 cells. 106
Figure IV-8. Overexpression of pre-miR-149 has no effect on proliferation of the Hs578T breast cancer cells. 107
Figure IV-9. miR-149 reduces lung targeting ability of metastatic IV2-1 cells. 108
Figure IV-10. GIT1 is a direct target of miR-149 in breast cancer cells. 109
Figure IV-11. miR-149 suppresses GIT1 expression at the mRNA level in IV2-1 cells. 111
Figure IV-12. miR-149 sponge suppresses miR-149 expression in parental 231 cells. 112
Figure IV-13. miR-149 and GIT1 expression are associated with the invasive phenotype of breast cancer cell lines. 113
Figure IV-14. miR-149-dependent inhibitory effect on cell migration/invasion and metastasis can be partially reversed by re-expression of GIT1 in vitro and in vivo. 114
Figure IV-15. Knockdown of GIT1 using siRNA transfection has no effect on GIT2 mRNA expression. 116
Figure IV-16. Depletion of GIT1 has no effect on proliferation of the IV2-1 cells. 117
Figure IV-17. Inhibition of GIT1 in the invasive Hs578T cells reduces their migration and invasion. 118
Figure IV-18. ArfGAP activity of GIT1 is not required for GIT1-mediated cell migration and invasion of IV2 cells. 119
Figure IV-19. Inhibition of GIT1 expression by miR-149 or GIT1 siRNAs suppresses phosphorylation of FAK and paxillin and focal adhesion formation of metastatic IV2 cells. 121
Figure IV-20. Depletion of GIT1 leads to enhanced protein degradation of paxillin and α5β1 integrin in metastatic IV2 cells. 123
Figure IV-21. GIT1 depletion greatly reduces α5β1integrin-mediated cell adhesion to fibronectin in metastatic IV2 cells. 125
Figure IV-22. Effect of depleting GIT1 on paxillin and α5β1 integrin level under different matrix-coating conditions. 126
Figure IV-23. Expression level of miR-149 and GIT1 in 90 breast tumor specimens and 50 adjacent normal tissues as determined by qRT-PCR. 127
Figure IV-24. Low level of miR-149 and high level of GIT1 correlate with lymph node metastasis. 128
Figure IV-25. Model of miR-149-GIT1 pathways in the regulation of breast cancer metastasis. 130
Figure IV-26. Evaluation of the transfection efficiency of pre-miR-149 using FAM-labeled pre-miR-149. 131
Tables 132
Table IV-1. Metastasis ability between parental 231 cells and IV2-1 cells. 132
Table IV-2. Candidate miRNAs down-regulated in IV2 lines. 133
Table IV-3. The list of primers and oligomers used in this study. 134
Table IV-3 continued. The list of primers and oligomers used in this study. 136
Table IV-4. Information of antibodies and reagents. 138
Chapter V: General discussion and future work 139
The Role of miRNA-491-5p and miR-149 in tumor progression 139
miRNAs as therapeutic targets and tools for cancer growth and metastasis 140
Role of GIT1, a target of miR-491-5p, as a therapeutic targets and tools for cancer progression 142
MiR-491-5p as an intronic microRNA is likely co-expressed with its host gene, FOCAD 144
Figures 148
Figure V-1. MiR-491-5p coding pre-sequence is located in intron 4 of the FOCAD gene 148
Figure V-2. Expression of intronic miR-491-5p and FOCAD 149
Figure V-3. miR-491-5p and FOCAD expression are down-regulated in the highly invasive oral cancer lines. 151
Figure V-4. Expression of targets of miR-491-5p are downregulated in OSCC cells overexpressing FOCAD intron 4 filament. 152
Figure V-5. Transfection of CGHNC9 cells with three set of FOCAD siRNAs effectively suppressed FOCAD expression. 154
Figure V-6. Inhibition of FOCAD expression promotes migration and invasion of human OSCC cells 155
References: 156

參考文獻 1. Saman W. Global epidemiology of oral and oropharyngeal cancer. Oral Oncology. 2009;45:309-16.
2. Chen Y-J, Chang JT-C, Liao C-T, Wang H-M, Yen T-C, Chiu C-C, et al. Head and neck cancer in the betel quid chewing area: recent advances in molecular carcinogenesis. Cancer Science. 2008;99:1507-14.
3. Ho P-S, Ko Y-C, Yang Y-HC, Shieh T-Y, Tsai C-C. The incidence of oropharyngeal cancer in Taiwan: an endemic betel quid chewing area. Journal of Oral Pathology & Medicine. 2002;31:213-9.
4. Punglia RS, Morrow M, Winer EP, Harris JR. Local Therapy and Survival in Breast Cancer. New England Journal of Medicine. 2007;356:2399-405.
5. Lin C-H, Liau J-Y, Lu Y-S, Huang C-S, Lee W-C, Kuo K-T, et al. Molecular Subtypes of Breast Cancer Emerging in Young Women in Taiwan: Evidence for More Than Just Westernization as a Reason for the Disease in Asia. Cancer Epidemiology Biomarkers & Prevention. 2009;18:1807-14.
6. Shen Y-C, Chang C-J, Hsu C, Cheng C-C, Chiu C-F, Cheng A-L. Significant Difference in the Trends of Female Breast Cancer Incidence Between Taiwanese and Caucasian Americans: Implications from Age-Period-Cohort Analysis. Cancer Epidemiology Biomarkers & Prevention. 2005;14:1986-90.
7. Lee K-D, Chen S-C, Chan CH, Lu C-H, Chen C-C, Lin J-T, et al. Increased Risk for Second Primary Malignancies in Women with Breast Cancer Diagnosed at Young Age: A Population-Based Study in Taiwan. Cancer Epidemiology Biomarkers & Prevention. 2008;17:2647-55.
8. Ferlito A, Rinaldo A, Devaney KO, MacLennan K, Myers JN, Petruzzelli GJ, et al. Prognostic significance of microscopic and macroscopic extracapsular spread from metastatic tumor in the cervical lymph nodes. Oral Oncology. 2002;38:747-51.
9. Mamelle G, Pampurik J, Luboinski B, Lancar R, Lusinchi A, Bosq J. Lymph node prognostic factors in head and neck squamous cell carcinomas. The American Journal of Surgery. 1994;168:494-8.
10. Liao C-T, Hsueh C, Lee L-Y, Lin C-Y, Fan K-H, Wang H-M, et al. Neck dissection field and lymph node density predict prognosis in patients with oral cavity cancer and pathological node metastases treated with adjuvant therapy. Oral Oncology. 2012;48:329-36.
11. Kurahara S-i, Shinohara M, Ikebe T, Nakamura S, Beppu M, Hiraki A, et al. Expression of MMPS, MT-MMP, and TIMPs in squamous cell carcinoma of the oral cavity: Correlations with tumor invasion and metastasis. Head & Neck. 1999;21:627-38.
12. American Cancer Society. Cancer statistics 1998. CA Cancer J Clin. 1998;48:1 - 63.
13. Taneja P, Maglic D, Kai F, Zhu S, Kendig RD, Fry EA, et al. Classical and Novel Prognostic Markers for Breast Cancer and their Clinical Significance. Clinical Medicine Insights: Oncology. 2010;4:15-34.
14. Boyd D. Invasion and metastasis. Cancer Metastasis Rev. 1996;15:77 - 89.
15. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol. 2009;11:228-34.
16. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509-24.
17. Bartel DP. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell. 2004;116:281-97.
18. Zeng Y, Cullen BR. Structural requirements for pre-microRNA binding and nuclear export by Exportin 5. Nucleic Acids Research. 2004;32:4776-85.
19. Valencia-Sanchez MA, Liu J, Hannon GJ, Parker R. Control of translation and mRNA degradation by miRNAs and siRNAs. Genes & Development. 2006;20:515-24.
20. Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs Exhibit Strand Bias. Cell. 2003;115:209-16.
21. Li S-C, Chan W-C, Ho M-R, Tsai K-W, Hu L-Y, Lai C-H, et al. Discovery and characterization of medaka miRNA genes by next generation sequencing platform. BMC Genomics. 2010;11:S8.
22. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Research. 2006;34:D140-D4.
23. Nicoloso MS, Spizzo R, Shimizu M, Rossi S, Calin GA. MicroRNAs [mdash] the micro steering wheel of tumour metastases. Nat Rev Cancer. 2009;9:293-302.
24. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-8.
25. Jiang L, Liu X, Kolokythas A, Yu J, Wang A, Heidbreder CE, et al. Downregulation of the Rho GTPase signaling pathway is involved in the microRNA-138-mediated inhibition of cell migration and invasion in tongue squamous cell carcinoma. International Journal of Cancer. 2010;127:505-12.
26. Pérez-Sayáns M, Pilar G-D, Barros-Angueira F, Suárez-Peñaranda JM, Fernández AC, Gándara-Rey JM, et al. Current trends in miRNAs and their relationship with oral squamous cell carcinoma. Journal of Oral Pathology & Medicine. 2012;41:433-43.
27. Negrini M, Calin G. Breast cancer metastasis: a microRNA story. Breast Cancer Research. 2008;10:303.
28. Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451:147-52.
29. Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR‐200 family promotes EMT and invasion in cancer cells. EMBO reports. 2008;9:582-9.
30. Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo Y-Y. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 0000;18:350-9.
31. Turnover of Focal Adhesions and Cancer Cell Migration. International Journal of Cell Biology. 2012;2012.
32. Hoefen RJ, Berk BC. The multifunctional GIT family of proteins. Journal of Cell Science. 2006;119:1469-75.
33. Hsu R-M, Tsai M-H, Hsieh Y-J, Lyu P-C, Yu J-S. Identification of MYO18A as a Novel Interacting Partner of the PAK2/βPIX/GIT1 Complex and Its Potential Function in Modulating Epithelial Cell Migration. Molecular Biology of the Cell. 2010;21:287-301.
34. Manabe R-i, Kovalenko M, Webb DJ, Horwitz AR. GIT1 functions in a motile, multi-molecular signaling complex that regulates protrusive activity and cell migration. Journal of Cell Science. 2002;115:1497-510.
35. Hajdo-Milasinovic A, van der Kammen RA, Moneva Z, Collard JG. Rac3 inhibits adhesion and differentiation of neuronal cells by modifying GIT1 downstream signaling. Journal of Cell Science. 2009;122:2127-36.
36. Schmalzigaug R, Garron M-L, Roseman JT, Xing Y, Davidson CE, Arold ST, et al. GIT1 utilizes a focal adhesion targeting-homology domain to bind paxillin. Cellular Signalling. 2007;19:1733-44.
37. Zhao Z-s, Manser E, Loo T-H, Lim L. Coupling of PAK-Interacting Exchange Factor PIX to GIT1 Promotes Focal Complex Disassembly. Molecular and Cellular Biology. 2000;20:6354-63.
38. Zhao J, Guan J-L. Signal transduction by focal adhesion kinase in cancer. Cancer and Metastasis Reviews. 2009;28:35-49.
39. Brunton VG, Frame MC. Src and focal adhesion kinase as therapeutic targets in cancer. Current Opinion in Pharmacology. 2008;8:427-32.
40. Subauste MC, Pertz O, Adamson ED, Turner CE, Junger S, Hahn KM. Vinculin modulation of paxillin–FAK interactions regulates ERK to control survival and motility. The Journal of Cell Biology. 2004;165:371-81.
41. Parsons JT. Focal adhesion kinase: the first ten years. Journal of Cell Science. 2003;116:1409-16.
42. Schlaepfer DD, Mitra SK, Ilic D. Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2004;1692:77-102.
43. Mitra SK, Hanson DA, Schlaepfer DD. Focal adhesion kinase: in command and control of cell motility. Nat Rev Mol Cell Biol. 2005;6:56-68.
44. Nicholson RI, Gee JMW, Harper ME. EGFR and cancer prognosis. European Journal of Cancer. 2001;37, Supplement 4:9-15.
45. Ciardiello F, Tortora G. A Novel Approach in the Treatment of Cancer: Targeting the Epidermal Growth Factor Receptor. Clinical Cancer Research. 2001;7:2958-70.
46. Tsai P-C, Hsieh C-Y, Chiu C-C, Wang C-K, Chang L-S, Lin S-R. Cardiotoxin III suppresses MDA-MB-231 cell metastasis through the inhibition of EGF/EGFR-mediated signaling pathway. Toxicon.
47. Yin G, Haendeler J, Yan C, Berk BC. GIT1 Functions as a Scaffold for MEK1-Extracellular Signal-Regulated Kinase 1 and 2 Activation by Angiotensin II and Epidermal Growth Factor. Molecular and Cellular Biology. 2004;24:875-85.
48. Brockschmidt A, Trost D, Peterziel H, Zimmermann K, Ehrler M, Grassmann H, et al. KIAA1797/FOCAD encodes a novel focal adhesion protein with tumour suppressor function in gliomas. Brain. 2012;135:1027-41.
49. Chekulaeva M, Mathys H, Zipprich JT, Attig J, Colic M, Parker R, et al. miRNA repression involves GW182-mediated recruitment of CCR4–NOT through conserved W-containing motifs. Nat Struct Mol Biol. 2011;18:1218-26.
50. Peng C-H, Liao C-T, Peng S-C, Chen Y-J, Cheng A-J, Juang J-L, et al. A Novel Molecular Signature Identified by Systems Genetics Approach Predicts Prognosis in Oral Squamous Cell Carcinoma. PLoS ONE. 2011;6:e23452.
51. Lu Y-C, Chen Y-J, Wang H-M, Tsai C-Y, Chen W-H, Huang Y-C, et al. Oncogenic Function and Early Detection Potential of miRNA-10b in Oral Cancer as Identified by microRNA Profiling. Cancer Prevention Research. 2012;5:665-74.
52. Gong BD, Xie Q, Wang L, Xiang XG, Lin LY, Zhao GD, et al. [Real-time quantification of microRNAs in Huh7 cells by stem-loop reverse transcriptase polymerase chain reaction]. Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese journal of hepatology. 2009;17:603-6.
53. Rong-Mei Wu1 MW, Anthony Thrush2, Eric F. Walton1, and Erika Varkonyi-Gasic1*. Real-Time PCR Quantification of Plant miRNAs Using Universal ProbeLibrary Technology. Biochemica. 2007:12-5.
54. Yeh Y-M, Chuang C-M, Chao K-C, Wang L-H. MicroRNA-138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1α. International Journal of Cancer. 2013:n/a-n/a.
55. Silahtaroglu AN, Nolting D, Dyrskjot L, Berezikov E, Moller M, Tommerup N, et al. Detection of microRNAs in frozen tissue sections by fluorescence in situ hybridization using locked nucleic acid probes and tyramide signal amplification. Nat Protocols. 2007;2:2520-8.
56. Weigelt B, Peterse JL, van′t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5:591-602.
57. Yamamoto E, Miyakawa A, Kohama G-I. Mode of invasion and lymph node metastasis in squamous cell carcinoma of the oral cavity. Head & Neck Surgery. 1984;6:938-47.
58. Cunningham MJ, Johnson JT, Myers EN, Schramm Jr VL, Thearle PB. Cervical lymph node metastasis after local excision of early squamous cell carcinoma of the oral cavity. The American Journal of Surgery. 1986;152:361-6.
59. Sakamoto S, McCann RO, Dhir R, Kyprianou N. Talin1 Promotes Tumor Invasion and Metastasis via Focal Adhesion Signaling and Anoikis Resistance. Cancer Research. 2010;70:1885-95.
60. Ren Y, Yu L, Fan J, Rui Z, Hua Z, Zhang Z, et al. Phosphorylation of GIT1 tyrosine 321 is required for association with FAK at focal adhesions and for PDGF-activated migration of osteoblasts. Molecular and Cellular Biochemistry. 2012:1-10.
61. Zhang ZM, Simmerman JA, Guibao CD, Zheng JJ. GIT1 Paxillin-binding Domain Is a Four-helix Bundle, and It Binds to Both Paxillin LD2 and LD4 Motifs. Journal of Biological Chemistry. 2008;283:18685-93.
62. Mendes O, Kim H-T, Lungu G, Stoica G. MMP2 role in breast cancer brain metastasis development and its regulation by TIMP2 and ERK1/2. Clin Exp Metastasis. 2007;24:341-51.
63. Liu Z, Li L, Yang Z, Luo W, Li X, Yang H, et al. Increased expression of MMP9 is correlated with poor prognosis of nasopharyngeal carcinoma. BMC Cancer. 2010;10:270.
64. Luo Y, Liang F, Zhang Z-Y. PRL1 Promotes Cell Migration and Invasion by Increasing MMP2 and MMP9 Expression through Src and ERK1/2 Pathways†. Biochemistry. 2009;48:1838-46.
65. Shim Y-J, Kang B-H, Jeon H-S, Park I-S, Lee K-U, Lee I-K, et al. Clusterin induces matrix metalloproteinase-9 expression via ERK1/2 and PI3K/Akt/NF-κB pathways in monocytes/macrophages. Journal of Leukocyte Biology. 2011;90:761-9.
66. Liao C-T, Huang S-F, Chen IH, Chang J-C, Wang H-M, Ng S-H, et al. Risk Stratification of Patients with Oral Cavity Squamous Cell Carcinoma and Contralateral Neck Recurrence Following Radical Surgery. Ann Surg Oncol. 2009;16:159-70.
67. Capote A, Escorial V, Muñoz-Guerra MF, Rodríguez-Campo FJ, Gamallo C, Naval L. Elective neck dissection in early-stage oral squamous cell carcinoma—does it influence recurrence and survival? Head & Neck. 2007;29:3-11.
68. Greenberg JS, El Naggar AK, Mo V, Roberts D, Myers JN. Disparity in pathologic and clinical lymph node staging in oral tongue carcinoma. Cancer. 2003;98:508-15.
69. Capote-Moreno A, Naval L, Muñoz-Guerra MF, Sastre J, Rodríguez-Campo FJ. Prognostic Factors Influencing Contralateral Neck Lymph Node Metastases in Oral and Oropharyngeal Carcinoma. Journal of Oral and Maxillofacial Surgery. 2010;68:268-75.
70. Yan W, Zhang W, Sun L, Liu Y, You G, Wang Y, et al. Identification of MMP-9 specific microRNA expression profile as potential targets of anti-invasion therapy in glioblastoma multiforme. Brain Research. 2011;1411:108-15.
71. Rutnam ZJ, Yang BB. The non-coding 3′ UTR of CD44 induces metastasis by regulating extracellular matrix functions. Journal of Cell Science. 2012;125:2075-85.
72. Wang J, Taba Y, Pang J, Yin G, Yan C, Berk BC. GIT1 Mediates VEGF-Induced Podosome Formation in Endothelial Cells. Arteriosclerosis, Thrombosis, and Vascular Biology. 2009;29:202-8.
73. Yin G, Zheng Q, Yan C, Berk BC. GIT1 Is a Scaffold for ERK1/2 Activation in Focal Adhesions. Journal of Biological Chemistry. 2005;280:27705-12.
74. Zhang N, Cai W, Yin G, Nagel DJ, Berk BC. GIT1 is a novel MEK1-ERK1/2 scaffold that localizes to focal adhesions. Cell biology international. 2010;34:41-7.
75. Kornberg LJ. Focal adhesion kinase and its potential involvement in tumor invasion and metastasis. Head & Neck. 1998;20:745-52.
76. Turner CE. Paxillin and focal adhesion signalling. Nat Cell Biol. 2000;2:E231-E6.
77. Hagel M, George EL, Kim A, Tamimi R, Opitz SL, Turner CE, et al. The Adaptor Protein Paxillin Is Essential for Normal Development in the Mouse and Is a Critical Transducer of Fibronectin Signaling. Molecular and Cellular Biology. 2002;22:901-15.
78. Shuyi Y, Feng W, Jing T, Hongzhang H, Haiyan W, Pingping M, et al. Human beta-defensin-3 (hBD-3) upregulated by LPS via epidermal growth factor receptor (EGFR) signaling pathways to enhance lymphatic invasion of oral squamous cell carcinoma. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology. 2011;112:616-25.
79. Sheu JJ-C, Hua C-H, Wan L, Lin Y-J, Lai M-T, Tseng H-C, et al. Functional Genomic Analysis Identified Epidermal Growth Factor Receptor Activation as the Most Common Genetic Event in Oral Squamous Cell Carcinoma. Cancer Research. 2009;69:2568-76.
80. Luo M, Guan J-L. Focal adhesion kinase: A prominent determinant in breast cancer initiation, progression and metastasis. Cancer Letters. 2010;289:127-39.
81. Liu S, Calderwood DA, Ginsberg MH. Integrin cytoplasmic domain-binding proteins. Journal of Cell Science. 2000;113:3563-71.
82. Hunziker W, Geuze HJ. Intracellular trafficking of lysosomal membrane proteins. BioEssays. 1996;18:379-89.
83. Luo Z, Zhang L, Li Z, Li X, Li G, Yu H, et al. An in silico analysis of dynamic changes in microRNA expression profiles in stepwise development of nasopharyngeal carcinoma. BMC Medical Genomics. 2012;5:3.
84. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5:816-26.
85. Wang Y, Zheng X, Zhang Z, Zhou J, Zhao G, Yang J, et al. MicroRNA-149 Inhibits Proliferation and Cell Cycle Progression through the Targeting of ZBTB2 in Human Gastric Cancer. PLoS ONE. 2012;7:e41693.
86. Calalb MB, Zhang X, Polte TR, Hanks SK. Focal Adhesion Kinase Tyrosine-861 Is a Major Site of Phosphorylation by Src. Biochemical and Biophysical Research Communications. 1996;228:662-8.
87. Parsons JT, Horwitz AR, Schwartz MA. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat Rev Mol Cell Biol. 2010;11:633-43.
88. Guo R, Wang Y, Shi W-Y, Liu B, Hou S-Q, Liu L. MicroRNA miR-491-5p Targeting both TP53 and Bcl-XL Induces Cell Apoptosis in SW1990 Pancreatic Cancer Cells through Mitochondria Mediated Pathway. Molecules. 2012;17:14733-47.
89. Nakano H, Miyazawa T, Kinoshita K, Yamada Y, Yoshida T. Functional screening identifies a microRNA, miR-491 that induces apoptosis by targeting Bcl-XL in colorectal cancer cells. International Journal of Cancer. 2010;127:1072-80.
90. Zhou Y, Li Y, Ye J, Jiang R, Yan H, Yang X, et al. MicroRNA-491 is involved in metastasis of hepatocellular carcinoma by inhibitions of matrix metalloproteinase and epithelial to mesenchymal transition. Liver International. 2013;33:1271-80.
91. Hui Z, Yiling C, Wenting Y, XuQun H, ChuanYi Z, Hui L. miR-491-5p functions as a tumor suppressor by targeting JMJD2B in ERα-positive breast cancer. FEBS Letters. 2015;589:812-21.
92. Lin R-J, Lin Y-C, Yu AL. miR-149* induces apoptosis by inhibiting Akt1 and E2F1 in human cancer cells. Molecular Carcinogenesis. 2010;49:719-27.
93. Molina-Pinelo S, Gutiérrez G, Pastor MD, Hergueta M, Moreno-Bueno G, García-Carbonero R, et al. MicroRNA-Dependent Regulation of Transcription in Non-Small Cell Lung Cancer. PLoS ONE. 2014;9:e90524.
94. Tu H-F, Liu C-J, Chang C-L, Wang P-W, Kao S-Y, Yang C-C, et al. The Association between Genetic Polymorphism and the Processing Efficiency of miR-149 Affects the Prognosis of Patients with Head and Neck Squamous Cell Carcinoma. PLoS ONE. 2012;7:e51606.
95. He D-X, Gu X-T, Li Y-R, Jiang L, Jin J, Ma X. Methylation-regulated miR-149 modulates chemoresistance by targeting GlcNAc N-deacetylase/N-sulfotransferase-1 in human breast cancer. FEBS Journal. 2014;281:4718-30.
96. Xu K, Liu X, Mao X, Xue L, Wang R, Chen L, et al. MicroRNA-149 Suppresses Colorectal Cancer Cell Migration and Invasion by Directly Targeting Forkhead Box Transcription Factor FOXM1. Cellular Physiology and Biochemistry. 2015;35:499-515.
97. Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Molecular Medicine. 2012;4:143-59.
98. Hall TMT. Structure and Function of Argonaute Proteins. Structure. 2005;13:1403-8.
99. Koshkin AA, Singh SK, Nielsen P, Rajwanshi VK, Kumar R, Meldgaard M, et al. LNA (Locked Nucleic Acids): Synthesis of the adenine, cytosine, guanine, 5-methylcytosine, thymine and uracil bicyclonucleoside monomers, oligomerisation, and unprecedented nucleic acid recognition. Tetrahedron. 1998;54:3607-30.
100. Ma L, Reinhardt F, Pan E, Soutschek J, Bhat B, Marcusson EG, et al. Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model. Nat Biotech. 2010;28:341-7.
101. Elmén J, Lindow M, Silahtaroglu A, Bak M, Christensen M, Lind-Thomsen A, et al. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Research. 2008;36:1153-62.
102. Rettig WJ, Old LJ. Immunogenetics of Human Cell Surface Differentiation. Annual Review of Immunology. 1989;7:481-511.
103. Hudis CA. Trastuzumab — Mechanism of Action and Use in Clinical Practice. New England Journal of Medicine. 2007;357:39-51.
104. Zhang L-L, Mu G-G, Ding Q-S, Li Y-X, Shi Y-B, Dai J-F, et al. PTEN represses colon cancer progression through inhibiting paxillin transcription via PI3K/AKT/NF-kB pathway. Journal of Biological Chemistry. 2015.
105. Deakin NO, Turner CE. Distinct roles for paxillin and Hic-5 in regulating breast cancer cell morphology, invasion, and metastasis. Molecular Biology of the Cell. 2011;22:327-41.
106. Deakin NO, Pignatelli J, Turner CE. Diverse Roles for the Paxillin Family of Proteins in Cancer. Genes & Cancer. 2012;3:362-70.
107. Lin S-L, Miller JD, Ying S-Y. Intronic MicroRNA (miRNA). Journal of Biomedicine and Biotechnology. 2006;2006:13.
108. Geiger B, Spatz JP, Bershadsky AD. Environmental sensing through focal adhesions. Nat Rev Mol Cell Biol. 2009;10:21-33.
109. Venkatachalam R, Verwiel ETP, Kamping EJ, Hoenselaar E, Görgens H, Schackert HK, et al. Identification of candidate predisposing copy number variants in familial and early-onset colorectal cancer patients. International Journal of Cancer. 2011;129:1635-42.
指導教授 王陸海、陳盛良(Lu-Hai Wang Shen-Liang Chen) 審核日期 2015-6-11
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