全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Molecular Signatures of Recurrent Hepatocellular Carcinoma Secondary to Hepatitis C Virus following Liver Transplantation

DOI: 10.1155/2013/878297

Full-Text   Cite this paper   Add to My Lib

Abstract:

Chronic hepatitis C virus (HCV) induced hepatocellular carcinoma (HCC) is a primary indication for liver transplantation (LT). In western countries, the estimated rate of HCC recurrence following LT is between 15% and 20% and is a major cause of mortality. Currently, there is no standard method to treat patients who are at high risk for HCC recurrence. The aim of this study was to investigate the molecular signatures underlying HCC recurrence that may lead to future studies on gene regulation contributing to new therapeutic options. Two groups of patients were selected, one including patients with HCV who developed HCC recurrence (HCC-R) ≤3 years from LT and the second group including patients with HCV who did not have recurrent HCC (HCC-NR). Microarray analysis containing more than 29,000 known genes was performed on formalin-fixed-paraffin-embedded (FFPE) liver tissue from explanted livers. Gene expression profiling revealed 194 differentially regulated genes between the two groups. These genes belonged to cellular networks including cell cycle G1/S checkpoint regulators, RAN signaling, chronic myeloid leukemia signaling, molecular mechanisms of cancer, FXR/RXR activation and hepatic cholestasis. A subset of molecular signatures associated with HCC recurrence was found. The expression levels of these genes were validated by quantitative PCR analysis. 1. Introduction Hepatitis C virus (HCV) infection is the most common cause of hepatocellular carcinoma (HCC) in the USA, Europe, and Japan, accounting for 47%–49%, 56%, and 75% of cases, respectively [1, 2]. HCC causes >600,000 deaths annually worldwide and is the most common primary liver cancer [3]. Definitive treatment for HCC is surgical resection when possible or liver transplantation for patients with end-stage liver disease and liver tumors. In USA, transplant guidelines stipulate that eligibility for liver transplantation is determined by the patient’s liver tumor(s) meeting the Milan criteria (a single tumor ≤5?cm in diameter or up to 3 tumors with individual diameters ≤3?cm and no macrovascular invasion) [4]. Unfortunately, the recurrence of HCC is a major cause of mortality in surgically treated patients [5]. There is no standard therapy for patients who are at high risk for HCC recurrence. Hence, a better understanding of the molecular mechanisms involved in the recurrence of HCC post LT is necessary to develop an efficient surveillance protocol and seek new potential therapies. Gene expression profiling is best performed on fresh or frozen tissue to lessen the degradation of RNA. However,

References

[1]  A. M. di Bisceglie, A. C. Lyra, M. Schwartz et al., “Hepatitis C-related hepatocellular carcinoma in the United States: influence of ethnic status,” American Journal of Gastroenterology, vol. 98, no. 9, pp. 2060–2063, 2003.
[2]  K. Kiyosawa, T. Umemura, T. Ichijo et al., “Hepatocellular carcinoma: recent trends in Japan,” Gastroenterology, vol. 127, no. 5, supplement 1, pp. S17–S26, 2004.
[3]  J. M. Llovet, A. Burroughs, and J. Bruix, “Hepatocellular carcinoma,” The Lancet, vol. 362, no. 9399, pp. 1907–1917, 2003.
[4]  V. Mazzaferro, E. Regalia, R. Doci et al., “Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis,” The New England Journal of Medicine, vol. 334, no. 11, pp. 693–699, 1996.
[5]  E. Y. Chan, A. M. Larson, O. K. Fix et al., “Identifying risk for recurrent hepatocellular carcinoma after liver transplantation: implications for surveillance studies and new adjuvant therapies,” Liver Transplantation, vol. 14, no. 7, pp. 956–965, 2008.
[6]  S. von Ahlfen, A. Missel, K. Bendrat, and M. Schlumpberger, “Determinants of RNA quality from FFPE samples,” PLoS One, vol. 2, no. 12, Article ID e1261, 2007.
[7]  S. M. Farragher, A. Tanney, R. D. Kennedy, and D. Paul Harkin, “RNA expression analysis from formalin fixed paraffin embedded tissues,” Histochemistry and Cell Biology, vol. 130, no. 3, pp. 435–445, 2008.
[8]  L. Turner, J. D. Heath, and N. Kurn, “Gene expression profiling of RNA extracted from FFPE tissues: NuGEN technologies' whole-transcriptome amplification system,” Methods in Molecular Biology, vol. 724, pp. 269–280, 2011.
[9]  R. A. Coudry, S. I. Meireles, R. Stoyanova et al., “Successful application of microarray technology to microdissected formalin-fixed, paraffin-embedded tissue,” Journal of Molecular Diagnostics, vol. 9, no. 1, pp. 70–79, 2007.
[10]  C. April, B. Klotzle, T. Royce et al., “Whole-genome gene expression profiling of formalin-fixed, paraffin-embedded tissue samples,” PLoS One, vol. 4, no. 12, Article ID e8162, 2009.
[11]  N. Iizuka, M. Oka, H. Yamada-Okabe et al., “Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection,” The Lancet, vol. 361, no. 9361, pp. 923–929, 2003.
[12]  Y. Kurokawa, R. Matoba, I. Takemasa et al., “Molecular-based prediction of early recurrence in hepatocellular carcinoma,” Journal of Hepatology, vol. 41, no. 2, pp. 284–291, 2004.
[13]  V. R. Mas, R. A. Fisher, K. J. Archer et al., “Genes associated with progression and recurrence of hepatocellular carcinoma in hepatitis C patients waiting and undergoing liver transplantation: preliminary results,” Transplantation, vol. 83, no. 7, pp. 973–981, 2007.
[14]  C. L. Lai, V. Ratziu, M.-F. Yuen, and T. Poynard, “Viral hepatitis B,” The Lancet, vol. 362, no. 9401, pp. 2089–2094, 2003.
[15]  T. Poynard, M.-F. Yuen, V. Ratziu, and C. Lung Lai, “Viral hepatitis C,” The Lancet, vol. 362, no. 9401, pp. 2095–2100, 2003.
[16]  J.-B. Fan, J. M. Yeakley, M. Bibikova et al., “A versatile assay for high-throughput gene expression profiling on universisal array matrices,” Genome Research, vol. 14, no. 5, pp. 878–885, 2004.
[17]  Y. Hoshida, A. Villanueva, M. Kobayashi, et al., “Gene expression in fixed tissues and outcome in hepatocellular carcinoma,” The New England Journal of Medicine, vol. 359, no. 19, pp. 1995–2004, 2008.
[18]  M. Abramovitz, M. Ordanic-Kodani, Y. Wang et al., “Optimization of RNA extraction from FFPE tissues for expression profiling in the DASL assay,” BioTechniques, vol. 44, no. 3, pp. 417–423, 2008.
[19]  C. C. Ton, N. Vartanian, X. Chai et al., “Gene expression array testing of FFPE archival breast tumor samples: an optimized protocol for WG-DASL sample preparation,” Breast Cancer Research and Treatment, vol. 125, no. 3, pp. 879–883, 2011.
[20]  M. Ravo, M. Mutarelli, L. Ferraro et al., “Quantitative expression profiling of highly degraded RNA from formalin-fixed, paraffin-embedded breast tumor biopsies by oligonucleotide microarrays,” Laboratory Investigation, vol. 88, no. 4, pp. 430–440, 2008.
[21]  L. Mittempergher, J. J. de Ronde, M. Nieuwland et al., “Gene expression profiles from formalin fixed paraffin embedded breast cancer tissue are largely comparable to fresh frozen matched tissue,” PLoS One, vol. 6, no. 2, Article ID e17163, 2011.
[22]  Y. Z. Chen, E. Soeda, H. W. Yang et al., “Homozygous deletion in a neuroblastoma cell line defined by a high-density STS map spanning human chromosome band Ip36,” Genes Chromosomes and Cancer, vol. 31, no. 4, pp. 326–332, 2001.
[23]  X. Liu, H. Zou, C. Slaughter, and X. Wang, “DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis,” Cell, vol. 89, no. 2, pp. 175–184, 1997.
[24]  W. Yang, L. M. Rozan, E. R. McDonald III et al., “CARPs are ubiquitin ligases that promote MDM2-independent p53 and phospho-p53ser20 degradation,” Journal of Biological Chemistry, vol. 282, no. 5, pp. 3273–3281, 2007.
[25]  A. Vinayagam, U. Stelzl, R. Foulle et al., “A directed protein interaction network for investigating intracellular signal transduction,” Science Signaling, vol. 4, no. 189, p. rs8, 2011.
[26]  W.-W. Lei, K.-H. Zhang, X.-C. Pan et al., “Histone deacetylase 1 and 2 differentially regulate apoptosis by opposing effects on extracellular signal-regulated kinase 1/2,” Cell Death and Disease, vol. 1, no. 5, article e44, 2010.
[27]  J. H. Noh, K. H. Jung, J. K. Kim et al., “Aberrant regulation of HDAC2 mediates proliferation of hepatocellular carcinoma cells by deregulating expression of G1/S cell cycle proteins,” PLoS One, vol. 6, no. 11, Article ID e28103, 2011.
[28]  E. Wurmbach, Y.-B. Chen, G. Khitrov et al., “Genome-wide molecular profiles of HCV-induced dysplasia and hepatocellular carcinoma,” Hepatology, vol. 45, no. 4, pp. 938–947, 2007.
[29]  M. E. Nemergut, C. A. Mizzen, T. Stukenberg, C. D. Allis, and I. G. Macara, “Chromatin docking and exchange activity enhancement of RCC1 by histones H2A and H2B,” Science, vol. 292, no. 5521, pp. 1540–1543, 2001.
[30]  A. C. Kimmelman, A. F. Hezel, A. J. Aguirre, et al., “Genomic alterations link Rho family of GTPases to the highly invasive phenotype of pancreas cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 49, pp. 19372–19377, 2008.
[31]  J. Xing, D. D. Ginty, and M. E. Greenberg, “Coupling of the RAS-MAPK pathway to gene activation by RSK2, a growth factor-regulated CREB kinase,” Science, vol. 273, no. 5277, pp. 959–963, 1996.
[32]  S. Kakugawa, M. Shimojima, H. Goto et al., “Mitogen-activated protein kinase-activated kinase RSK2 plays a role in innate immune responses to influenza virus infection,” Journal of Virology, vol. 83, no. 6, pp. 2510–2517, 2009.
[33]  C. Guichard, G. Amaddeo, S. Imbeaud et al., “Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma,” Nature Genetics, vol. 44, no. 6, pp. 694–698, 2012.
[34]  L. Homesley, M. Lei, Y. Kawasaki, S. Sawyer, T. Christensen, and B. K. Tye, “Mcm10 and the MCM2-7 complex interact to initiate DNA synthesis and to release replication factors from origins,” Genes and Development, vol. 14, no. 8, pp. 913–926, 2000.
[35]  K. A. Honeycutt, Z. Chen, M. I. Koster et al., “Deregulated minichromosomal maintenance protein MCM7 contributes to oncogene driven tumorigenesis,” Oncogene, vol. 25, no. 29, pp. 4027–4032, 2006.
[36]  J. Liu, S.-K. Yao, and F. Yin, “Expression and significance of minichromosome maintenance protein7 in hepatocellular carcinoma,” Zhonghua Gan Zang Bing Za Zhi, vol. 16, no. 1, pp. 55–56, 2008.
[37]  R. W. Furlanetto, B. R. Dey, W. Lopaczynski, and S. P. Nissley, “14-3-3 proteins interact with the insulin-like growth factor receptor but not the insulin receptor,” Biochemical Journal, vol. 327, part 3, pp. 765–771, 1997.
[38]  M. Niemantsverdriet, K. Wagner, M. Visser, and C. Backendorf, “Cellular functions of 14-3-3ζ in apoptosis and cell adhesion emphasize its oncogenic character,” Oncogene, vol. 27, no. 9, pp. 1315–1319, 2008.
[39]  J. E. Choi, W. Hur, C. K. Jung et al., “Silencing of 14-3-3ζ over-expression in hepatocellular carcinoma inhibits tumor growth and enhances chemosensitivity to cis-diammined dichloridoplatium,” Cancer Letters, vol. 303, no. 2, pp. 99–107, 2011.
[40]  C. Sardet, M. Vidal, D. Cobrinik et al., “E2F-4 and E2F-5, two members of the E2F family, are expressed in the early phases of the cell cycle,” Proceedings of the National Academy of Sciences of the United States of America, vol. 92, no. 6, pp. 2403–2407, 1995.
[41]  N. Kothandaraman, V. B. Bajic, P. N. K. Brendan et al., “E2F5 status significantly improves malignancy diagnosis of epithelial ovarian cancer,” BMC Cancer, vol. 10, article 64, 2010.
[42]  G. V. Petukhova, R. J. Pezza, F. Vanevski, M. Ploquin, J.-Y. Masson, and R. D. Camerini-Otero, “The Hop2 and Mnd1 proteins act in concert with Rad51 and Dmc1 in meiotic recombination,” Nature Structural and Molecular Biology, vol. 12, no. 5, pp. 449–453, 2005.
[43]  J. He, C.-C. Mao, A. Reyes et al., “The AAA+ protein ATAD3 has displacement loop binding properties and is involved in mitochondrial nucleoid organization,” Journal of Cell Biology, vol. 176, no. 2, pp. 141–146, 2007.
[44]  L. Zhang, Z. Smit-McBride, X. Pan, J. Rheinhardt, and J. W. B. Hershey, “An oncogenic role for the phosphorylated h-subunit of human translation initiation factor eIF3,” Journal of Biological Chemistry, vol. 283, no. 35, pp. 24047–24060, 2008.
[45]  H. Okamoto, K. Yasui, C. Zhao, S. Arii, and J. Inazawa, “PTK2 and EIF3S3 genes may be amplification targets at 8q23-q24 and are associated with large hepatocellular carcinomas,” Hepatology, vol. 38, no. 5, pp. 1242–1249, 2003.
[46]  M. Saegusa, Y. Takano, H. Kishimoto, G. Wakabayashi, K. Nohga, and M. Okudaira, “Comparative analysis of p53 and c-myc expression and cell proliferation in human hepatocellular carcinomas—an enhanced immunohistochemical approach,” Journal of Cancer Research and Clinical Oncology, vol. 119, no. 12, pp. 737–744, 1993.
[47]  Y. Hoshida, S. M. B. Nijman, M. Kobayashi et al., “Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma,” Cancer Research, vol. 69, no. 18, pp. 7385–7392, 2009.
[48]  F. Simon, M. Bockhorn, C. Praha et al., “Deregulation of HIF1-alpha and hypoxia-regulated pathways in hepatocellular carcinoma and corresponding non-malignant liver tissue-influence of a modulated host stroma on the prognosis of HCC,” Langenbeck's Archives of Surgery, vol. 395, no. 4, pp. 395–405, 2010.
[49]  Z.-G. Chang, L.-Y. Yang, W. Wang et al., “Determination of high mobility group A1 (HMGA1) expression in hepatocellular carcinoma: a potential prognostic marker,” Digestive Diseases and Sciences, vol. 50, no. 10, pp. 1764–1770, 2005.
[50]  B. Shen, E. S. H. Chu, G. Zhao et al., “PPARgamma inhibits hepatocellular carcinoma metastases in vitro and in mice,” British Journal of Cancer, vol. 106, no. 9, pp. 1486–1494, 2012.
[51]  A. Csepregi, M. P. A. Ebert, C. R?cken et al., “Promoter methylation of CDKN2A and lack of p16 expression characterize patients with hepatocellular carcinoma,” BMC Cancer, vol. 10, Article ID 3171471, 2010.
[52]  Y. Matsuda, T. Ichida, J. Matsuzawa, K. Sugimura, and H. Asakura, “p16(INK4) is inactivated by extensive CpG methylation in human hepatocellular carcinoma,” Gastroenterology, vol. 116, no. 2, pp. 394–400, 1999.
[53]  Y. Liu, J. Binz, M. J. Numerick et al., “Hepatoprotection by the farnesoid X receptor agonist GW4064 in rat models of intra- and extrahepatic cholestasis,” Journal of Clinical Investigation, vol. 112, no. 11, pp. 1678–1687, 2003.
[54]  V. R. Mas, D. G. Maluf, K. J. Archer et al., “Genes involved in viral carcinogenesis and tumor initiation in hepatitis C virus-induced hepatocellular carcinoma,” Molecular Medicine, vol. 15, no. 3-4, pp. 85–94, 2009.
[55]  D. L. Diamond, A. L. Krasnoselsky, K. E. Burnum, et al., “Proteome and computational analyses reveal new insights into the mechanisms of hepatitis C virus-mediated liver disease posttransplantation,” Hepatology, vol. 56, no. 1, pp. 28–38, 2012.
[56]  K. Furuta, S. Sato, T. Yamauchi, and S. Kakumu, “Changes in intrahepatic gene expression profiles from chronic hepatitis to hepatocellular carcinoma in patients with hepatitis C virus infection,” Hepatology Research, vol. 38, no. 7, pp. 673–682, 2008.
[57]  J. Jiang, L. Yu, X. Huang et al., “Identification of two novel human dynein light chain genes, DNLC2A and DNLC2B, and their expression changes in hepatocellular carcinoma tissues from 68 Chinese patients,” Gene, vol. 281, no. 1-2, pp. 103–113, 2001.
[58]  S. Boyault, D. S. Rickman, A. de Reyniès et al., “Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets,” Hepatology, vol. 45, no. 1, pp. 42–52, 2007.
[59]  C. Schlaeger, T. Longerich, C. Schiller et al., “Etiology-dependent molecular mechanisms in human hepatocarcinogenesis,” Hepatology, vol. 47, no. 2, pp. 511–520, 2008.
[60]  M. W. Welker, W. O. Bechstein, S. Zeuzem, and J. Trojan, “Recurrent hepatocellular carcinoma after liver transplantation—an emerging clinical challenge,” Transplant International, vol. 26, no. 2, pp. 109–118, 2013.
[61]  E. R. McDonald III and W. S. El-Deiry, “Suppression of caspase-8- and -10-associated RING proteins results in sensitization to death ligands and inhibition of tumor cell growth,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 16, pp. 6170–6175, 2004.
[62]  C. H. Chen, S. M. Chuang, M. F. Yang, J. W. Liao, S. L. Yu, and J. J. Chen, “A novel function of YWHAZ/beta-catenin axis in promoting epithelial-mesenchymal transition and lung cancer metastasis,” Molecular Cancer Research, vol. 10, no. 10, pp. 1319–1331, 2012.
[63]  A. Budhu, M. Forgues, Q.-H. Ye et al., “Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment,” Cancer Cell, vol. 10, no. 2, pp. 99–111, 2006.
[64]  T. Seki, N. Hayashi, and T. Nishimoto, “RCC1 in the Ran pathway,” Journal of Biochemistry, vol. 120, no. 2, pp. 207–214, 1996.
[65]  T. Tachibana, N. Imamoto, H. Seino, T. Nishimoto, and Y. Yoneda, “Loss of RCC1 leads to suppression of nuclear protein import in living cells,” Journal of Biological Chemistry, vol. 269, no. 40, pp. 24542–24545, 1994.
[66]  H.-F. Yuen, K.-K. Chan, C. Grills et al., “Ran is a potential therapeutic target for cancer cells with molecular changes associated with activation of the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways,” Clinical Cancer Research, vol. 18, no. 2, pp. 380–391, 2012.
[67]  U. Giri, C. L. Ashorn, L. Ramdas et al., “Molecular signatures associated with clinical outcome in patients with high-risk head-and-neck squamous cell carcinoma treated by surgery and radiation,” International Journal of Radiation Oncology Biology Physics, vol. 64, no. 3, pp. 670–677, 2006.
[68]  A. Mortazavi, B. A. Williams, K. McCue, L. Schaeffer, and B. Wold, “Mapping and quantifying mammalian transcriptomes by RNA-Seq,” Nature Methods, vol. 5, no. 7, pp. 621–628, 2008.
[69]  J. D. Morlan, K. Qu, and D. V. Sinicropi, “Selective depletion of rRNA enables whole transcriptome profiling of archival fixed tissue,” PLoS One, vol. 7, no. 8, Article ID e42882, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413