帳號:guest(18.222.46.156)          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
作者(中文):邱鈺凱
作者(外文):Chiu, Yu Kai
論文名稱(中文):藉由建立肝臟疾病和肝癌的全基因組基因和表觀遺傳網路去探討肝癌化機制
論文名稱(外文):Constructing genome-wide genetic-and-epigenetic networks of liver diseases and liver cancer to investigate hepatocarcinogenic mechanisms via system modeling, microarray data and NGS data
指導教授(中文):陳博現
指導教授(外文):Chen, Bor Sen
口試委員(中文):楊嘉鈴
蘇士哲
王慧菁
學位類別:碩士
校院名稱:國立清華大學
系所名稱:電機工程學系
學號:102061552
出版年(民國):105
畢業學年度:104
語文別:英文
論文頁數:76
中文關鍵詞:肝癌肝臟疾病基因和表觀遺傳網路肝癌化機制
外文關鍵詞:HCCliver diseasesmiRNADNA methylationbig database mininggenetic and epigenetic network (GEN)genesishepatocarcinogenesismolecular mechanismmultiple drug targetsmultiple molecules drug
相關次數:
  • 推薦推薦:0
  • 點閱點閱:456
  • 評分評分:*****
  • 下載下載:0
  • 收藏收藏:0
在全世界,肝細胞癌是常見並且致命的惡性腫瘤。在過去,大多數的肝細胞癌患者都是由病毒感染演變,像是B型肝炎跟C型肝炎。然而近期的抗病毒療法有效的降低B型肝炎跟C型肝炎的患者數目,並且肝細胞癌中由病毒感染發展的患病率明顯地下降。有趣的是因為其他肝臟疾病發展成肝細胞癌的患病相關性也增加,所以在全世界肝癌的總患病率仍然維持在一個穩定的水平。因此,在肝細胞癌的研究中,探討正常的肝臟細胞演變到肝癌經由其他肝臟疾病的肝癌化過程是當務之急。
基於我們提出的系統生物方法,我們建立各個肝臟疾病的全基因組的基因和表觀遺傳網路 (GEN) 並且進一步的從不同肝臟狀況的GENs中萃取出各自的核心基因和表觀遺傳網路。此外,藉由比較正常肝臟細胞以及肝臟疾病的核心基因和表觀遺傳網路,我們能進一步選出特異核心網路的標誌物去研究正常肝臟細胞到肝細胞癌的演變機制。藉由更深入的探討特異核心網路,我們能得到正常的肝臟細胞演變到肝細胞癌經由其他肝臟疾病癌化過程的分子機制觀點。所以,我們識別出正常肝臟細胞到肝細胞癌經由非酒精性脂肪肝疾病(NAFLD)&非酒精性脂肪肝炎(NASH)的肝癌化過程主要可以被Wnt信號通路和MAPK信號通路通過HIST2H2BE、HSPB1、RPL30和ALDOB的DNA甲基化以及miR-21和miR-122的調控所誘導,這些可以做為肝癌化過程中的潛在複合藥物標靶。並且我們還識別出正常肝臟細胞到肝細胞癌經由原發性膽汁性肝硬化(PBC)&原發性硬化性膽管炎(PSC)的肝癌化過程主要可以被Wnt信號通路和MAPK信號通路通過RPL23A、HIST2H2BE、TIMP1、IGF2、RPL30和ALDOB的DNA甲基化以及miR-29a、miR-21和miR-122的調控所誘導,這些可以做為肝癌化過程中的潛在複合藥物標靶。最後,一個包含lactacystin、OGX-427、DIHYDROXYACETONE PHOSPHATE、curcumin 和quercetin的複合分子藥物被用來抑制NAFLD&NASH這類型肝臟疾病的肝癌化過程通過標靶上述的複合藥物標靶。另一個包含lactacystin、L-(-)-3-Phenyllactic acid、DIHYDROXYACETONE PHOSPHATE、batimastat、mannose 6-phosphate、curcumin 、quercetin和ellagic acid的複合分子藥物被用來抑制PBC&PSC這類型肝臟疾病的肝癌化過程通過標靶上述的複合藥物標靶。

Hepatocellular carcinoma (HCC) is common and deadly malignancy worldwide. In the past, most HCC patients were developed by viral infection such as hepatitis B virus (HBV) or hepatitis C virus (HCV). However, antiviral therapy has much influenced on the reduction of patients with HBV and HCV recently, and the prevalence of viral infection development of HCC is obviously decreased. Intriguingly, the total prevalence of HCC is still maintained in a stable level worldwide due to other liver diseases being developed to HCC to increase prevalence correspondingly. Hence, investigating hepatocarcinogenesis from normal liver cell to liver cancer through other liver diseases is a priority in HCC research.
Based on the proposed systems biology approach, we constructed genome-wide genetic-and-epigenetic networks (GENs) and further extracted core genetic-and-epigenetic networks from their GENs at different liver conditions. Moreover, we further selected specific core network markers by comparing core genetic-and-epigenetic networks of normal liver cell and liver diseases to investigate progression mechanisms from normal liver cells to HCC. By further investigating specific core networks, we could get insight into the molecular mechanisms of hepatocarcinogenesis from normal liver cell to HCC through different liver diseases. Furthermore, we identified that the hepatocarcinogenesis from normal liver cell to HCC through nonalcoholic fatty liver disease (NAFLD)& nonalcoholic steatohepatitis (NASH) can be induced mainly by WNT signaling pathway and MAPK signaling pathway through DNA methylation of HIST2H2BE, HSPB1, RPL30 and ALDOB, and the regulations of miR-21 and miR-122, to be potential multiple drug targets of the hepatocarcinogenesis. And we also identified that the hepatocarcinogenesis from normal liver cell to HCC through primary biliary cirrhosis (PBC)& primary sclerosing cholangitis (PSC) can be induced mainly by WNT signaling pathway and MAPK signaling pathway through DNA methylation of RPL23A, HIST2H2BE, TIMP1, IGF2, RPL30 and ALDOB, and the regulations of miR-29a, miR-21 and miR-122, to be potential multiple drug targets too. Finally, a multiple molecules drug comprising lactacystin, OGX-427, DIHYDROXYACETONE PHOSPHATE, curcumin and quercetin was proposed to target the above multiple drug targets for inhibiting the hepatocarcinogenesis through NAFLD&NASH liver diseases. Another multiple molecules drug comprising lactacystin, L-(-)-3-Phenyllactic acid, DIHYDROXYACETONE PHOSPHATE, batimastat, mannose 6-phosphate, curcumin, quercetin and ellagic acid was also proposed to target the above multiple drug targets for inhibiting the hepatocarcinogenesis through PBC&PSC liver diseases.

Keywords: HCC、liver diseases、 miRNA、DNA methylation、big database mining、genetic and epigenetic network (GEN)、genesis、hepatocarcinogenesis、molecular mechanism、multiple drug targets、multiple molecules drug
致謝 i
摘要 ii
Abstract iv
Contents vi
List of Figures vii
List of Tables viii
List of Supplemental materials viii
Introduction 1
Materials and Methods 7
2.1 Overview of the construction for genome-wide GENs, core GENs and specific core networks of genesis and hepatocarcinogenesis of HCC 7
2.2 Big data mining and preprocessing of both expression data and methylation data for liver diseases and HCC 8
2.3 Constructing genome-wide candidate genetic-and-epigenetic network 10
2.4 Constructing protein-protein interaction network (PPIN) and gene regulatory network (GRN) by system modeling method via microarray data and NGS data 10
2.5 Parameter estimation of candidate PPIN and GRN by the constrained least square estimation method and system order detection scheme 13
2.6 Determining significant GEN structures (core GENs) of the different liver diseases via Principal network projection (PNP) 18
2.7 Drug mining and design for NAFLD&NASH-developed HCC and PBC&PSC-developed HCC through the investigations of key network biomarkers of progression mechanisms in Figure 9 20
Results 22
3.1 Analysis of specific core networks to investigate underlying cellular mechanisms for genesis and hepatocarcinogenesis in the upper progression path of Figure 1 24
3.2 Analysis of specific core networks to investigate underlying cellular mechanisms for genesis and hepatocarcinogenesis in the lower progression path of Figure 1 27
3.3 The progression molecular mechanism based on specific core networks in the upper progression path and the lower progression path of Figure 1 31
Discussion 35
4.1 Dysregulations of DNA methylation and miRNA regulation contribute to genesis and hepatocarcinogenesis from normal liver cell to HCC through NAFLD&NASH or PBC&PSC 36
4.1.1 Dysregulations of DNA methylation and miR-21 contribute to genesis and hepatocarcinogenesis from the upper progression path to HCC in Figure 1 36
4.1.2 Dysregulations of DNA methylation, miR-21, miR-122 and miR-29a, contribute to genesis and hepatocarcinogenesis from the lower progression path to HCC in Figure 1 38
4.1.3 Dysregulation of miR-21, miR-122 and miR-29a contribute to human HCC tumor progression 39
4.2 Multiple molecules drug design for NAFLD&NASH-developed HCC and PBC&PSC-developed HCC by integrating the detailed investigations of progression mechanisms and drug databases 41
Conclusion 43





[1] S. Bandiera, S. Pfeffer, T. F. Baumert, and M. B. Zeisel, "miR-122--a key factor and therapeutic target in liver disease," J Hepatol, vol. 62, pp. 448-57, Feb 2015.
[2] H. Nishikawa and Y. Osaki, "Non-B, non-C hepatocellular carcinoma (Review)," Int J Oncol, vol. 43, pp. 1333-42, Nov 2013.
[3] X. Y. Duan, L. Zhang, J. G. Fan, and L. Qiao, "NAFLD leads to liver cancer: do we have sufficient evidence?," Cancer Lett, vol. 345, pp. 230-4, Apr 10 2014.
[4] L. Kikuchi, C. P. Oliveira, and F. J. Carrilho, "Nonalcoholic fatty liver disease and hepatocellular carcinoma," Biomed Res Int, vol. 2014, p. 106247, 2014.
[5] Z. F. Wu, Z. Xu, W. S. Li, H. B. Zhang, N. Yang, X. Q. Yao, et al., "Impact of occult hepatitis B virus infection on outcome after resection for non-B non-C hepatocellular carcinoma," J Surg Res, vol. 193, pp. 153-60, Jan 2015.
[6] S. Mittal, D. L. White, F. Kanwal, N. Sussman, and H. B. El-Serag, "Nonalcoholic Fatty Liver Disease (NAFLD) and Hepatocellular Carcinoma: How Common?," Current Hepatology Reports, pp. 1-12, 2015.
[7] A. Scalera and G. Tarantino, "Could metabolic syndrome lead to hepatocarcinoma via non-alcoholic fatty liver disease?," World J Gastroenterol, vol. 20, pp. 9217-28, Jul 28 2014.
[8] D. A. Sass, P. Chang, and K. B. Chopra, "Nonalcoholic fatty liver disease: a clinical review," Digestive diseases and sciences, vol. 50, pp. 171-180, 2005.
[9] K. Harada and Y. Nakanuma, "Prevalence and risk factors of hepatocellular carcinoma in Japanese patients with primary biliary cirrhosis," Hepatol Res, vol. 44, pp. 133-40, Feb 2014.
[10] Y. Sun, W. Zhang, B. Li, Z. Zou, C. Selmi, and M. E. Gershwin, "The coexistence of Sjogren's syndrome and primary biliary cirrhosis: a comprehensive review," Clin Rev Allergy Immunol, vol. 48, pp. 301-15, Jun 2015.
[11] X. X. Zhang, L. F. Wang, L. Jin, Y. Y. Li, S. L. Hao, Y. C. Shi, et al., "Primary biliary cirrhosis-associated hepatocellular carcinoma in Chinese patients: incidence and risk factors," World J Gastroenterol, vol. 21, pp. 3554-63, Mar 28 2015.
[12] T. H. Karlsen, M. Vesterhus, and K. M. Boberg, "Review article: controversies in the management of primary biliary cirrhosis and primary sclerosing cholangitis," Aliment Pharmacol Ther, vol. 39, pp. 282-301, Feb 2014.
[13] N. Razumilava, G. J. Gores, and K. D. Lindor, "Cancer surveillance in patients with primary sclerosing cholangitis," Hepatology, vol. 54, pp. 1842-52, Nov 2011.
[14] R. Zenouzi, T. J. Weismuller, P. Hubener, K. Schulze, M. Bubenheim, N. Pannicke, et al., "Low risk of hepatocellular carcinoma in patients with primary sclerosing cholangitis with cirrhosis," Clin Gastroenterol Hepatol, vol. 12, pp. 1733-8, Oct 2014.
[15] S. M. El-Haggar and T. M. Mostafa, "Comparative clinical study between the effect of fenofibrate alone and its combination with pentoxifylline on biochemical parameters and liver stiffness in patients with non-alcoholic fatty liver disease," Hepatology international, pp. 1-9, 2015.
[16] S. Francque, A. Verrijken, S. Caron, J. Prawitt, R. Paumelle, B. Derudas, et al., "PPARα gene expression correlates with severity and histological treatment response in patients with non-alcoholic steatohepatitis," Journal of hepatology, 2015.
[17] M. J. Pollheimer and P. Fickert, "Animal models in primary biliary cirrhosis and primary sclerosing cholangitis," Clin Rev Allergy Immunol, vol. 48, pp. 207-17, Jun 2015.
[18] K. D. Williamson and R. W. Chapman, "Primary sclerosing cholangitis: a clinical update," Br Med Bull, vol. 114, pp. 53-64, Jun 2015.
[19] G. A. Michelotti, M. V. Machado, and A. M. Diehl, "NAFLD, NASH and liver cancer," Nat Rev Gastroenterol Hepatol, vol. 10, pp. 656-65, Nov 2013.
[20] S. Wasserman and K. Faust, Social network analysis: Methods and applications vol. 8: Cambridge university press, 1994.
[21] A. H. Y. Tong, G. Lesage, G. D. Bader, H. Ding, H. Xu, X. Xin, et al., "Global mapping of the yeast genetic interaction network," science, vol. 303, pp. 808-813, 2004.
[22] Y.-C. Wang and B.-S. Chen, "A network-based biomarker approach for molecular investigation and diagnosis of lung cancer," BMC medical genomics, vol. 4, p. 2, 2011.
[23] S. Horvath, W. Erhart, M. Brosch, O. Ammerpohl, W. von Schonfels, M. Ahrens, et al., "Obesity accelerates epigenetic aging of human liver," Proc Natl Acad Sci U S A, vol. 111, pp. 15538-43, Oct 28 2014.
[24] A. Chatr-Aryamontri, B. J. Breitkreutz, R. Oughtred, L. Boucher, S. Heinicke, D. Chen, et al., "The BioGRID interaction database: 2015 update," Nucleic Acids Res, vol. 43, pp. D470-8, Jan 2015.
[25] G. Zheng, K. Tu, Q. Yang, Y. Xiong, C. Wei, L. Xie, et al., "ITFP: an integrated platform of mammalian transcription factors," Bioinformatics, vol. 24, pp. 2416-2417, 2008.
[26] L. A. Bovolenta, M. L. Acencio, and N. Lemke, "HTRIdb: an open-access database for experimentally verified human transcriptional regulation interactions," BMC genomics, vol. 13, p. 405, 2012.
[27] E. Wingender, "The TRANSFAC project as an example of framework technology that supports the analysis of genomic regulation," Briefings in bioinformatics, vol. 9, pp. 326-332, 2008.
[28] B. P. Lewis, C. B. Burge, and D. P. Bartel, "Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets," cell, vol. 120, pp. 15-20, 2005.
[29] A. E. Ferreira, A. M. Ponces Freire, and E. O. Voit, "A quantitative model of the generation of N(epsilon)-(carboxymethyl)lysine in the Maillard reaction between collagen and glucose," Biochem J, vol. 376, pp. 109-21, Nov 15 2003.
[30] E. O. Voit, Computational analysis of biochemical systems: a practical guide for biochemists and molecular biologists: Cambridge University Press, 2000.
[31] Y. A. Medvedeva, A. M. Khamis, I. V. Kulakovskiy, W. Ba-Alawi, M. S. I. Bhuyan, H. Kawaji, et al., "Effects of cytosine methylation on transcription factor binding sites," BMC genomics, vol. 15, p. 119, 2014.
[32] M. Weber, I. Hellmann, M. B. Stadler, L. Ramos, S. Pääbo, M. Rebhan, et al., "Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome," Nature genetics, vol. 39, pp. 457-466, 2007.
[33] Y.-C. Wang and B.-S. Chen, "Integrated cellular network of transcription regulations and protein-protein interactions," BMC Systems Biology, vol. 4, p. 20, 2010.
[34] P. Lu, C. Vogel, R. Wang, X. Yao, and E. M. Marcotte, "Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation," Nature biotechnology, vol. 25, pp. 117-124, 2007.
[35] Y.-H. Wong, R.-H. Chen, and B.-S. Chen, "Core and specific network markers of carcinogenesis from multiple cancer samples," Journal of theoretical biology, vol. 362, pp. 17-34, 2014.
[36] R. Johansson, "System Modeling & Identification," Prentice -Hall, Engle- wood Cliffs. NJ, 1993.
[37] B.-S. Chen and C.-C. Wu, "Systems biology as an integrated platform for bioinformatics, systems synthetic biology, and systems metabolic engineering," Cells, vol. 2, pp. 635-688, 2013.
[38] D. S. Wishart, C. Knox, A. C. Guo, S. Shrivastava, M. Hassanali, P. Stothard, et al., "DrugBank: a comprehensive resource for in silico drug discovery and exploration," Nucleic acids research, vol. 34, pp. D668-D672, 2006.
[39] M. Griffith, O. L. Griffith, A. C. Coffman, J. V. Weible, J. F. McMichael, N. C. Spies, et al., "DGIdb: mining the druggable genome," Nature methods, vol. 10, pp. 1209-1210, 2013.
[40] J. Lamb, E. D. Crawford, D. Peck, J. W. Modell, I. C. Blat, M. J. Wrobel, et al., "The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease," Science, vol. 313, pp. 1929-1935, 2006.
[41] D. Komander, "The emerging complexity of protein ubiquitination," Biochem Soc Trans, vol. 37, pp. 937-53, Oct 2009.
[42] H. Zheng and E. H. Koo, "The amyloid precursor protein: beyond amyloid," Molecular neurodegeneration, vol. 1, p. 5, 2006.
[43] B. De Strooper and W. Annaert, "Proteolytic processing and cell biological functions of the amyloid precursor protein," Journal of cell science, vol. 113, pp. 1857-1870, 2000.
[44] Y. Zheng, C. Zhang, D. R. Croucher, M. A. Soliman, N. St-Denis, A. Pasculescu, et al., "Temporal regulation of EGF signalling networks by the scaffold protein Shc1," Nature, vol. 499, pp. 166-171, 2013.
[45] H.-J. Shih, H.-H. Chen, Y.-A. Chen, M.-H. Wu, G.-G. Liou, W.-W. Chang, et al., "Targeting MCT-1 oncogene inhibits Shc pathway and xenograft tumorigenicity," Oncotarget, Impact Journals LLC, vol. 3, 2012.
[46] K. Oda, Y. Matsuoka, A. Funahashi, and H. Kitano, "A comprehensive pathway map of epidermal growth factor receptor signaling," Molecular systems biology, vol. 1, 2005.
[47] H. Lanaya, A. Natarajan, K. Komposch, L. Li, N. Amberg, L. Chen, et al., "EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation," Nature cell biology, vol. 16, pp. 972-981, 2014.
[48] D. Xu, T. J. Wilson, D. Chan, E. De Luca, J. Zhou, P. J. Hertzog, et al., "Ets1 is required for p53 transcriptional activity in UV‐induced apoptosis in embryonic stem cells," The EMBO journal, vol. 21, pp. 4081-4093, 2002.
[49] M. J. Bround, R. Wambolt, D. S. Luciani, J. E. Kulpa, B. Rodrigues, R. W. Brownsey, et al., "Cardiomyocyte ATP production, metabolic flexibility, and survival require calcium flux through cardiac ryanodine receptors in vivo," Journal of Biological Chemistry, vol. 288, pp. 18975-18986, 2013.
[50] C. Sun, J.-G. Fan, and L. Qiao, "Potential Epigenetic Mechanism in Non-Alcoholic Fatty Liver Disease," International journal of molecular sciences, vol. 16, pp. 5161-5179, 2015.
[51] Q. Lu, "The critical importance of epigenetics in autoimmunity," Journal of autoimmunity, vol. 41, pp. 1-5, 2013.
[52] M. A. Dawson and T. Kouzarides, "Cancer epigenetics: from mechanism to therapy," Cell, vol. 150, pp. 12-27, Jul 6 2012.
[53] J. H. Lee, S. Friso, and S.-W. Choi, "Epigenetic mechanisms underlying the link between non-alcoholic fatty liver diseases and nutrition," Nutrients, vol. 6, pp. 3303-3325, 2014.
[54] Y.-Y. Li, "Genetic and epigenetic variants influencing the development of nonalcoholic fatty liver disease," World journal of gastroenterology: WJG, vol. 18, p. 6546, 2012.
[55] Y. Murakami, T. Yasuda, K. Saigo, T. Urashima, H. Toyoda, T. Okanoue, et al., "Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues," Oncogene, vol. 25, pp. 2537-2545, 2006.
[56] J. K. Dowman, J. Tomlinson, and P. Newsome, "Pathogenesis of non-alcoholic fatty liver disease," Qjm, vol. 103, pp. 71-83, 2010.
[57] P. Pettinelli, A. Obregón, and L. Videla, "Molecular mechanisms of steatosis in nonalcoholic fatty liver disease," Nutr Hosp, vol. 26, pp. 441-50, 2011.
[58] F. Meng, R. Henson, H. Wehbe–Janek, K. Ghoshal, S. T. Jacob, and T. Patel, "MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer," Gastroenterology, vol. 133, pp. 647-658, 2007.
[59] Q. Zhu, Z. Wang, Y. Hu, J. Li, X. Li, L. Zhou, et al., "miR-21 promotes migration and invasion by the miR-21-PDCD4-AP-1 feedback loop in human hepatocellular carcinoma," Oncology reports, vol. 27, pp. 1660-1668, 2012.
[60] L. Bao, Y. Yan, C. Xu, W. Ji, S. Shen, G. Xu, et al., "MicroRNA-21 suppresses PTEN and hSulf-1 expression and promotes hepatocellular carcinoma progression through AKT/ERK pathways," Cancer letters, vol. 337, pp. 226-236, 2013.
[61] J. Xu, C. Wu, X. Che, L. Wang, D. Yu, T. Zhang, et al., "Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis," Mol Carcinog, vol. 50, pp. 136-42, Feb 2011.
[62] C. Sun, F. Huang, X. Liu, X. Xiao, M. Yang, G. Hu, et al., "miR-21 regulates triglyceride and cholesterol metabolism in non-alcoholic fatty liver disease by targeting HMGCR," International journal of molecular medicine, vol. 35, pp. 847-853, 2015.
[63] Z. Yi, Y. Li, W. Ma, D. Li, C. Zhu, J. Luo, et al., "A novel KRAB zinc-finger protein, ZNF480, expresses in human heart and activates transcriptional activities of AP-1 and SRE," Biochemical and biophysical research communications, vol. 320, pp. 409-415, 2004.
[64] E. K. Kim and E.-J. Choi, "Pathological roles of MAPK signaling pathways in human diseases," Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, vol. 1802, pp. 396-405, 2010.
[65] S.-Y. Peng, P.-L. Lai, H.-W. Pan, L.-P. Hsiao, and H.-C. Hsu, "Aberrant expression of the glycolytic enzymes aldolase B and type II hexokinase in hepatocellular carcinoma are predictive markers for advanced stage, early recurrence and poor prognosis," Oncology reports, vol. 19, pp. 1045-1053, 2008.
[66] M. Shiota, J. L. Bishop, K. M. Nip, A. Zardan, A. Takeuchi, T. Cordonnier, et al., "Hsp27 regulates epithelial mesenchymal transition, metastasis, and circulating tumor cells in prostate cancer," Cancer research, vol. 73, pp. 3109-3119, 2013.
[67] M. De Bortoli, R. C. Castellino, X.-Y. Lu, J. Deyo, L. M. Sturla, A. M. Adesina, et al., "Medulloblastoma outcome is adversely associated with overexpression of EEF1D, RPL30, and RPS20 on the long arm of chromosome 8," BMC cancer, vol. 6, p. 223, 2006.
[68] S. Hohenester, R. P. Oude-Elferink, and U. Beuers, "Primary biliary cirrhosis," in Seminars in immunopathology, 2009, pp. 283-307.
[69] J. Baier and J. Mattner, "Mechanisms of Autoimmune Liver Disease," Discovery medicine, vol. 18, pp. 255-263, 2014.
[70] X. W. Wang, N. H. Heegaard, and H. Orum, "MicroRNAs in liver disease," Gastroenterology, vol. 142, pp. 1431-43, Jun 2012.
[71] M. Gyugos, G. Lendvai, I. Kenessey, K. Schlachter, J. Halász, P. Nagy, et al., "MicroRNA expression might predict prognosis of epithelial hepatoblastoma," Virchows Archiv, vol. 464, pp. 419-427, 2014.
[72] S. H. Hsu, B. Wang, J. Kota, J. Yu, S. Costinean, H. Kutay, et al., "Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver," J Clin Invest, vol. 122, pp. 2871-83, Aug 2012.
[73] Y. Xiong, J. H. Fang, J. P. Yun, J. Yang, Y. Zhang, W. H. Jia, et al., "Effects of MicroRNA‐29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma," Hepatology, vol. 51, pp. 836-845, 2010.
[74] M. Ciechomska, S. O’Reilly, M. Suwara, K. Bogunia-Kubik, and J. M. van Laar, "MiR-29a Reduces TIMP-1 Production by Dermal Fibroblasts via Targeting TGF-β Activated Kinase 1 Binding Protein 1, Implications for Systemic Sclerosis," PloS one, vol. 9, p. e115596, 2014.
[75] Y. Sun, D. Gao, Y. Liu, J. Huang, S. Lessnick, and S. Tanaka, "IGF2 is critical for tumorigenesis by synovial sarcoma oncoprotein SYT-SSX1," Oncogene, vol. 25, pp. 1042-1052, 2006.
[76] Y. Gong, E. Scott, R. Lu, Y. Xu, W. K. Oh, and Q. Yu, "TIMP-1 promotes accumulation of cancer associated fibroblasts and cancer progression," PloS one, vol. 8, p. e77366, 2013.
[77] S. Kallappagoudar, R. K. Yadav, B. R. Lowe, and J. F. Partridge, "Histone H3 mutations—a special role for H3. 3 in tumorigenesis?," Chromosoma, vol. 124, pp. 177-189, 2015.
[78] A. A. Nagle, F.-F. Gan, G. Jones, C.-L. So, G. Wells, and E.-H. Chew, "Induction of tumor cell death through targeting tubulin and evoking dysregulation of cell cycle regulatory proteins by multifunctional cinnamaldehydes," 2012.
[79] N. V. Chaika, F. Yu, V. Purohit, K. Mehla, A. J. Lazenby, D. DiMaio, et al., "Differential expression of metabolic genes in tumor and stromal components of primary and metastatic loci in pancreatic adenocarcinoma," PloS one, vol. 7, p. e32996, 2012.
[80] Y.-W. Chen, V. Boyartchuk, and B. C. Lewis, "Differential roles of insulin-like growth factor receptor-and insulin receptor-mediated signaling in the phenotypes of hepatocellular carcinoma cells," Neoplasia, vol. 11, pp. 835-IN1, 2009.
[81] F. Ma, S. Xu, X. Liu, Q. Zhang, X. Xu, M. Liu, et al., "The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-[gamma]," Nature immunology, vol. 12, pp. 861-869, 2011.
[82] Y. Bronevetsky and K. M. Ansel, "Regulation of miRNA biogenesis and turnover in the immune system," Immunological reviews, vol. 253, pp. 304-316, 2013.
[83] R. B. Shirley, I. Kaddour-Djebbar, D. M. Patel, V. Lakshmikanthan, R. W. Lewis, and M. V. Kumar, "Combination of proteasomal inhibitors lactacystin and MG132 induced synergistic apoptosis in prostate cancer cells," Neoplasia, vol. 7, pp. 1104-1111, 2005.
[84] V. Baylot, C. Andrieu, M. Katsogiannou, D. Taieb, S. Garcia, S. Giusiano, et al., "OGX-427 inhibits tumor progression and enhances gemcitabine chemotherapy in pancreatic cancer," Cell death & disease, vol. 2, p. e221, 2011.
[85] G. W. Sledge, M. Qulali, R. Goulet, E. A. Bone, and R. Fife, "Effect of matrix metalloproteinase inhibitor batimastat on breast cancer regrowth and metastasis in athymic mice," Journal of the National Cancer Institute, vol. 87, pp. 1546-1551, 1995.
[86] J. S. Lee, J. Weiss, J. L. Martin, and C. D. Scott, "Increased expression of the mannose 6‐phosphate/insulin‐like growth factor‐II receptor in breast cancer cells alters tumorigenic properties in vitro and in vivo," International journal of cancer, vol. 107, pp. 564-570, 2003.
[87] G. Mudduluru, J. George-William, S. Muppala, I. Asangani, R. Kumarswamy, L. Nelson, et al., "Curcumin regulates miR-21 expression and inhibits invasion and metastasis in colorectal cancer," Bioscience reports, vol. 31, pp. 185-197, 2011.
[88] L. Gibellini, M. Pinti, M. Nasi, J. P. Montagna, S. De Biasi, E. Roat, et al., "Quercetin and cancer chemoprevention," Evidence-Based Complementary and Alternative Medicine, vol. 2011, 2011.
[89] A. Malik, S. Afaq, M. Shahid, K. Akhtar, and A. Assiri, "Influence of ellagic acid on prostate cancer cell proliferation: A caspase–dependent pathway," Asian Pacific journal of tropical medicine, vol. 4, pp. 550-555, 2011.
[90] E. G. Mimnaugh, H. Y. Chen, J. R. Davie, J. E. Celis, and L. Neckers, "Rapid deubiquitination of nucleosomal histones in human tumor cells caused by proteasome inhibitors and stress response inducers: effects on replication, transcription, translation, and the cellular stress response," Biochemistry, vol. 36, pp. 14418-14429, 1997.
[91] F. Lamoureux, C. Thomas, M.-J. Yin, L. Fazli, A. Zoubeidi, and M. E. Gleave, "Suppression of heat shock protein 27 using OGX-427 induces endoplasmic reticulum stress and potentiates heat shock protein 90 inhibitors to delay castrate-resistant prostate cancer," European urology, vol. 66, pp. 145-155, 2014.
[92] C. Boesch-Saadatmandi, A. E. Wagner, S. Wolffram, and G. Rimbach, "Effect of quercetin on inflammatory gene expression in mice liver in vivo–role of redox factor 1, miRNA-122 and miRNA-125b," Pharmacological Research, vol. 65, pp. 523-530, 2012.
[93] M. Corbel, S. Caulet‐Maugendre, N. Germain, S. Molet, V. Lagente, and E. Boichot, "Inhibition of bleomycin‐induced pulmonary fibrosis in mice by the matrix metalloproteinase inhibitor batimastat," The Journal of pathology, vol. 193, pp. 538-545, 2001.
[94] R. Ai, S. Wu, X. Wen, W. Xu, L. Lv, J. Rao, et al., "1, 3, 4-tri-O-galloyl-6-O-caffeoyl-β-D-glucopyranose, a new anti-proliferative ellagitannin, regulates the expression of microRNAs in HepG (2) cancer cells," Nan fang yi ke da xue xue bao= Journal of Southern Medical University, vol. 31, pp. 1641-1648, 2011.
(此全文限內部瀏覽)
電子全文
摘要
 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *