研究生: |
吳柏毅 Wu, Po-Yi |
---|---|
論文名稱: |
藉由大數據探勘及NGS數據判別方法來研究乳癌發展的全基因組之基因和表觀遺傳基因網路及其分子機制 Constructing the Genome-wide Genetic and Epigenetic Networks for Investigating the Molecular Mechanisms of Breast Cancer Development by Big Database Mining and NGS Data Identification |
指導教授: |
陳博現
Chen, Bor-Sen |
口試委員: |
詹鴻霖
Chan, Hong-Lin 王慧菁 Wang, Hui Ching 蘇士哲 Sue, Shih-che 王禹超 Wang, Yu-Chao |
學位類別: |
碩士 Master |
系所名稱: |
電機資訊學院 - 電機工程學系 Department of Electrical Engineering |
論文出版年: | 2017 |
畢業學年度: | 105 |
語文別: | 英文 |
論文頁數: | 66 |
中文關鍵詞: | 乳癌 、全基因組 、表觀遺傳 、生物標誌物 、藥物 |
外文關鍵詞: | breast cancer, Genome-wide, epigenetic, biomarker, drug |
相關次數: | 點閱:2 下載:0 |
分享至: |
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報 |
乳癌是全世界婦女中最常見的癌症,也是婦女因癌症死亡最主要的原因。越來越多的研究表明乳癌是由基因、表觀遺傳基因和環境因素引起的。表觀遺傳調控在乳癌生物學中逐漸受到重視。我們藉由大數據探勘的方法來建立人類細胞的候選基因和表觀遺傳基因網路(GEN)。我們根據正常乳房細胞和乳癌細胞的次世代定序數據(NGS數據),並藉由系統識別和系統階層測定方法來刪減候選GEN中的偽陽性並獲得四期乳癌的全基因組GEN。接著,為了研究重要的分子機制,我們藉由主成份網路投影方法(PNP)從各期的GEN萃取出對應的核心GEN。為了分析乳癌的發展機制,我們比較連續兩期乳癌的核心GEN並進一步萃取出核心生物途徑來找出差異的生物途徑並挑選出乳癌發展機制中的關鍵基因和表觀遺傳基因生物標誌物。此外,我們也確定了表觀遺傳調控和微環境因素在乳癌發展機制中的作用。這些重要的發展機制以及表觀遺傳調控的改變顯示,早期乳癌細胞比起正常細胞有較高的增生能力並促進腫瘤附近的淋巴管形成。淋巴管附近的中期乳癌細胞有趨化性移動的傾向並與淋巴內皮細胞的膜蛋白結合而促進淋巴轉移。晚期乳癌細胞受到巨噬細胞分泌因子的刺激以及上皮間質轉換(EMT)的影響使其細胞黏附能力被抑制。根據這些發展機制,我們識別了數個促進乳癌發展的基因和表觀遺傳基因生物標誌物作為藥物目標。最後,我們提出三組多重藥物針對這些基因和表觀遺傳基因生物標誌物來治療其對應的各期乳癌。
Breast cancer is the most common cancer and also the major cause of death by cancer among women throughout the world. Accumulating studies indicate that breast cancer is caused by complex combination of genetic, epigenetic and environmental factors. Epigenetic regulation recently has become an increasingly important aspect of breast cancer biology. We constructed the candidate genetic and epigenetic network (GEN) of human cells by using big database mining. Based on next generation sequencing data (NGS data) of breast normal and cancer cells, we identified four genome-wide GENs of each stage of breast cancer by pruning false-positives in candidate GENs through system identification method and system order detection scheme. Then, in order to investigate the significant molecular mechanisms, we extracted the corresponding core GENs from their GENs in each stage via principal network projection (PNP) method. To analyze the progression mechanisms of breast cancer more conveniently, we further extracted the core pathways by comparing two core GENs of two consecutive stages of breast cancer to find differential pathways and choosing the most crucial genetic and epigenetic biomarkers playing the key roles on the progression mechanisms in breast carcinogenesis. In addition, we also identified the roles of identified epigenetic regulations and microenvironment factors in the progression mechanism of breast cancer in their interacting with these core pathways. According to these key progression mechanisms, early-stage breast cancer cells have a higher ability of cell proliferation than normal cells to promote lymphangiogenesis around tumor resulting from the change of related epigenetic regulations. Mid-stage breast cancer cells around lymphatic vessels tend to chemotactic migration and binding to the membrane proteins of lymphatic endothelial cells to promote lymphatic metastasis due to the change of related epigenetic regulations. Late-stage breast cancer cells are stimulated by the factors secreted from macrophages and affected by epithelial-mesenchymal transition (EMT), and their cell adhesion ability is also inhibited due to the change of related epigenetic regulations. Based on these progression mechanisms, we identified several genetic and epigenetic biomarkers crucial to promote breast carcinogenesis as drug targets. Finally, we proposed three multiple drugs to target these genetic and epigenetic biomarkers for therapeutic treatment of breast cancer at three corresponding carcinogenic stages.
Reference
1. Edge, S.B. and C.C. Compton, The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol, 2010. 17(6): p. 1471-4.
2. Hsu, C.W., H.F. Juan, and H.C. Huang, Characterization of microRNA-regulated protein-protein interaction network. Proteomics, 2008. 8(10): p. 1975-9.
3. Lin, C.C., et al., Dynamic functional modules in co-expressed protein interaction networks of dilated cardiomyopathy. BMC Syst Biol, 2010. 4: p. 138.
4. Chen, C.Y., et al., Coregulation of transcription factors and microRNAs in human transcriptional regulatory network. BMC Bioinformatics, 2011. 12 Suppl 1: p. S41.
5. Lee, R.C. and V. Ambros, An extensive class of small RNAs in Caenorhabditis elegans. Science, 2001. 294(5543): p. 862-4.
6. Stefani, G. and F.J. Slack, Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol, 2008. 9(3): p. 219-30.
7. Jones, P.A. and S.B. Baylin, The fundamental role of epigenetic events in cancer. Nat Rev Genet, 2002. 3(6): p. 415-28.
8. Jones, P.A. and S.B. Baylin, The epigenomics of cancer. Cell, 2007. 128(4): p. 683-92.
9. Lu, P., et al., Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nat Biotechnol, 2007. 25(1): p. 117-24.
10. Li, C.W. and B.S. Chen, Network Biomarkers of Bladder Cancer Based on a Genome-Wide Genetic and Epigenetic Network Derived from Next-Generation Sequencing Data. Dis Markers, 2016. 2016: p. 4149608.
11. Shannon, P., et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 2003. 13(11): p. 2498-504.
12. Forbes, S.A., et al., COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in Cancer. Curr Protoc Hum Genet, 2016. 91: p. 10 11 1-10 11 37.
13. Twigg, S.R., et al., Mutations of ephrin-B1 (EFNB1), a marker of tissue boundary formation, cause craniofrontonasal syndrome. Proc Natl Acad Sci U S A, 2004. 101(23): p. 8652-7.
14. Maxwell, C.A., et al., RHAMM is a centrosomal protein that interacts with dynein and maintains spindle pole stability. Molecular Biology of the Cell, 2003. 14(6): p. 2262-2276.
15. O'Donovan, P.J. and D.M. Livingston, BRCA1 and BRCA2: breast/ovarian cancer susceptibility gene products and participants in DNA double-strand break repair. Carcinogenesis, 2010. 31(6): p. 961-967.
16. Lu, Y.L., et al., The PTEN/MMAC1/TEP tumor suppressor gene decreases cell growth and induces apoptosis and anoikis in breast cancer cells. Oncogene, 1999. 18(50): p. 7034-7045.
17. Baeg, G.H., et al., The Tumor-Suppressor Gene-Product Apc Blocks Cell-Cycle Progression from G(0)/G(1) to S-Phase. Embo Journal, 1995. 14(22): p. 5618-5625.
18. Dikovskaya, D., et al., Loss of APC induces polyploidy as a result of a combination of defects in mitosis and apoptosis (vol 176, pg 183, 2007). Journal of Cell Biology, 2007. 176(3).
19. Singh, K.K., et al., BRCA1 is a novel target to improve endothelial dysfunction and retard atherosclerosis. Journal of Thoracic and Cardiovascular Surgery, 2013. 146(4): p. 949-+.
20. Huang, W., et al., FW-04-806 inhibits proliferation and induces apoptosis in human breast cancer cells by binding to N-terminus of Hsp90 and disrupting Hsp90-Cdc37 complex formation. Molecular Cancer, 2014. 13.
21. Mostovich, L.A., et al., The TCF4/beta-catenin pathway and chromatin structure cooperate to regulate D-glucuronyl C5-epimerase expression in breast cancer. Epigenetics, 2012. 7(8): p. 930-939.
22. Prudnikova, T.Y., et al., Antiproliferative effect of D-glucuronyl C5-epimerase in human breast cancer cells. Cancer Cell International, 2010. 10.
23. Jadhav, R.R., et al., Genome-wide DNA methylation analysis reveals estrogen-mediated epigenetic repression of metallothionein-1 gene cluster in breast cancer. Clinical Epigenetics, 2015. 7.
24. Tilghman, J., et al., HMMR Maintains the Stemness and Tumorigenicity of Glioblastoma Stem-like Cells. Cancer Research, 2014. 74(11): p. 3168-3179.
25. Joukov, V., et al., The BRCA1/BARD1 heterodimer modulates ran-dependent mitotic spindle assembly. Cell, 2006. 127(3): p. 539-552.
26. Zhang, J.M., et al., Kallistatin antagonizes Wnt/beta-catenin signaling and cancer cell motility via binding to low-density lipoprotein receptor-related protein 6. Molecular and Cellular Biochemistry, 2013. 379(1-2): p. 295-301.
27. Milovanovic, T., et al., Expression of Wnt genes and frizzled 1 and 2 receptors in normal breast epithelium and infiltrating breast carcinoma. International Journal of Oncology, 2004. 25(5): p. 1337-1342.
28. Burwinkel, B., et al., Transcription factor 7-like 2 (TCF7L2) variant is associated with familial breast cancer risk: a case-control study. Bmc Cancer, 2006. 6.
29. King, T.D., M.J. Suto, and Y.H. Li, The wnt/beta-catenin signaling pathway: A potential therapeutic target in the treatment of triple negative breast cancer. Journal of Cellular Biochemistry, 2012. 113(1): p. 13-18.
30. Naidu, R., C.H. Yip, and N.A.M. Taib, Genetic variations in transcription factor 7-like 2 (TCF7L2) gene: association of TCF7L2 rs12255372(G/T) or rs7903146(C/T) with breast cancer risk and clinico-pathological parameters. Medical Oncology, 2012. 29(2): p. 411-417.
31. Zhang, H., et al., Interference of Frizzled 1 (FZD1) reverses multidrug resistance in breast cancer cells through the Wnt/beta-catenin pathway. Cancer Letters, 2012. 323(1): p. 106-113.
32. Liu, W.G., et al., Mutations in AXIN2 cause colorectal cancer with defective mismatch repair by activating beta-catenin/TCF signalling (vol 26, pg 146, 2000). Nature Genetics, 2000. 26(4): p. 501-501.
33. Koinuma, K., H. Nagai, and H. Mano, Epigenetic silencing of AXIN2 in colorectal carcinoma with microsatellite instability. Gastroenterology, 2006. 130(4): p. A716-A716.
34. Aristizabal-Pachon, A.F., et al., AXIN2 Polymorphisms, the beta-Catenin Destruction Complex Expression Profile and Breast Cancer Susceptibility. Asian Pac J Cancer Prev, 2015. 16(16): p. 7277-84.
35. Pang, M.F., et al., TGF-beta 1-induced EMT promotes targeted migration of breast cancer cells through the lymphatic system by the activation of CCR7/CCL21-mediated chemotaxis. Oncogene, 2016. 35(6): p. 748-760.
36. Tutunea-Fatan, E., et al., The role of CCL21/CCR7 chemokine axis in breast cancer-induced lymphangiogenesis. Molecular Cancer, 2015. 14.
37. Jiang, J.H., A. Jedinak, and D. Sliva, Ganodermanontriol (GDNT) exerts its effect on growth and invasiveness of breast cancer cells through the down-regulation of CDC20 and uPA. Biochemical and Biophysical Research Communications, 2011. 415(2): p. 325-329.
38. Whitehurst, B., et al., Anti-VEGF-A therapy reduces lymphatic vessel density and expression of VEGFR-3 in an orthotopic breast tumor model. International Journal of Cancer, 2007. 121(10): p. 2181-2191.
39. Gasparini, G. and A.L. Harris, Clinical Importance of the Determination of Tumor Angiogenesis in Breast-Carcinoma - Much More Than a New Prognostic Tool. Journal of Clinical Oncology, 1995. 13(3): p. 765-782.
40. Sfiligoi, C., et al., Angiopoietin-2 expression in breast cancer correlates with lymph node invasion and short survival. International Journal of Cancer, 2003. 103(4): p. 466-474.
41. Chae, S.S., et al., Angiopoietin-2 interferes with anti-VEGFR2-induced vessel normalization and survival benefit in mice bearing gliomas. Clin Cancer Res, 2010. 16(14): p. 3618-27.
42. Zhang, W., et al., A monoclonal antibody that blocks VEGF binding to VEGFR2 (KDR/Flk-1) inhibits vascular expression of Flk-1 and tumor growth in an orthotopic human breast cancer model. Angiogenesis, 2002. 5(1-2): p. 35-44.
43. Yang, F., et al., MALAT-1 interacts with hnRNP C in cell cycle regulation. FEBS Lett, 2013. 587(19): p. 3175-81.
44. Shirure, V.S., et al., CD44 variant isoforms expressed by breast cancer cells are functional E-selectin ligands under flow conditions. Am J Physiol Cell Physiol, 2015. 308(1): p. C68-78.
45. Conacci-Sorrell, M., J. Zhurinsky, and A. Ben-Ze'ev, The cadherin-catenin adhesion system in signaling and cancer. J Clin Invest, 2002. 109(8): p. 987-91.
46. Park, J. and J.E. Schwarzbauer, Mammary epithelial cell interactions with fibronectin stimulate epithelial-mesenchymal transition. Oncogene, 2014. 33(13): p. 1649-57.
47. Bae, Y.K., et al., Fibronectin expression in carcinoma cells correlates with tumor aggressiveness and poor clinical outcome in patients with invasive breast cancer. Hum Pathol, 2013. 44(10): p. 2028-37.
48. Jinga, D.C., et al., MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: correlations with prognostic factors. J Cell Mol Med, 2006. 10(2): p. 499-510.
49. Nagase, H. and J.F. Woessner, Jr., Matrix metalloproteinases. J Biol Chem, 1999. 274(31): p. 21491-4.
50. Hagemann, T., et al., Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol, 2005. 175(2): p. 1197-205.
51. Jain, M., et al., Rudhira/BCAS3 is a cytoskeletal protein that controls Cdc42 activation and directional cell migration during angiogenesis. Experimental Cell Research, 2012. 318(6): p. 753-767.
52. Raftopoulou, M. and A. Hall, Cell migration: Rho GTPases lead the way. Dev Biol, 2004. 265(1): p. 23-32.
53. Li, H., et al., MicroRNA-217, down-regulated in clear cell renal cell carcinoma and associated with lower survival, suppresses cell proliferation and migration. Neoplasma, 2013. 60(5): p. 511-5.
54. Wang, G., et al., Induction of metastasis by S100P in a rat mammary model and its association with poor survival of breast cancer patients. Cancer Res, 2006. 66(2): p. 1199-207.
55. Bischoff, F.R., et al., Co-activation of RanGTPase and inhibition of GTP dissociation by Ran-GTP binding protein RanBP1. EMBO J, 1995. 14(4): p. 705-15.
56. Comijn, J., et al., The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell, 2001. 7(6): p. 1267-78.
57. Kulbe, H., et al., The inflammatory cytokine tumor necrosis factor-alpha regulates chemokine receptor expression on ovarian cancer cells. Cancer Research, 2005. 65(22): p. 10355-10362.
58. Oguma, K., et al., Activated macrophages promote Wnt signalling through tumour necrosis factor-alpha in gastric tumour cells. Embo Journal, 2008. 27(12): p. 1671-1681.
59. Lamb, J., et al., The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science, 2006. 313(5795): p. 1929-35.
60. Inbar-Rozensal, D., et al., A selective eradication of human nonhereditary breast cancer cells by phenanthridine-derived polyADP-ribose polymerase inhibitors. Breast Cancer Res, 2009. 11(6): p. R78.
61. Nagasawa, H. and J. Meites, Suppression by ergocornine and iproniazid of carcinogen-induced mammary tumors in rats; effects on serum and pituitary prolactin levels. Proc Soc Exp Biol Med, 1970. 135(2): p. 469-72.
62. Villar-Garea, A., et al., Procaine is a DNA-demethylating agent with growth-inhibitory effects in human cancer cells. Cancer Research, 2003. 63(16): p. 4984-4989.
63. Papendorp, J.T., et al., On the Role of 17-Alpha-Estradiol and 17-Beta-Estradiol in the Proliferation of Mcf7 and T47d-A11 Human-Breast Tumor-Cells. Journal of Cellular Physiology, 1985. 125(3): p. 591-595.
64. Youngren, J.F., et al., Nordihydroguaiaretic acid (NDGA) inhibits the IGF-1 and c-erbB2/HER2/neu receptors and suppresses growth in breast cancer cells. Breast Cancer Research and Treatment, 2005. 94(1): p. 37-46.
65. Bonadonna, G., et al., Adjuvant cyclophosphamide, methotrexate, and fluorouracil in node-positive breast cancer: the results of 20 years of follow-up. N Engl J Med, 1995. 332(14): p. 901-6.
66. Chiang, C.T., et al., Diosgenin, a naturally occurring steroid, suppresses fatty acid synthase expression in HER2-overexpressing breast cancer cells through modulating Akt, mTOR and JNK phosphorylation. Febs Letters, 2007. 581(30): p. 5735-5742.