Brief Report Open Access
Copyright ©The Author(s) 2001. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 15, 2001; 7(4): 575-578
Published online Aug 15, 2001. doi: 10.3748/wjg.v7.i4.575
Differentially expressed genes in hepatocellular carcinoma induced by woodchuck hepatitis B virus in mice
Xiao-Yan Cao, Jie Liu, Jia-Lu Hu, Dai-Ming Fan, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an 710033, China
Zhao-Rui Lian, Marcy Clayton, Mark Feitelson, Department of Pathology & Cell Biology, Thomas Jefferson University, Philadelphia, PA19107 USA
Ming-Hua Zhu, Department of Pathology, Second Military Medical University, Shanghai 200433, China
Author contributions: All authors contributed equally to the work.
Supported by NSFC grant 30024002
Correspondence to: Jie Liu, Institute of Digestive Diseases, Xijing Hospital, Xi’an 710033, Shaanxi Province, China. jliu5@pub.xaonline.com
Telephone: 0086-29-3375229 Fax: 0086-29-2359041
Received: April 6, 2001
Revised: May 11, 2001
Accepted: May 18, 2001
Published online: August 15, 2001

Abstract
Key Words: liver neoplasm/virology, carcinoma, hepatocellular/virology, hepatitis B virus/genetics, genes, viral, gene expression



INTRODUCTION

Hepatocellular carcinoma (HCC) is one of the major causes of death in the world. The mechanism of carcinogenesis is unknown, although it is widely accepted that HBV and HCV are closely related to liver cancer[1-5]. Previously, a variety of studies have described the differences in gene expression which distinguished tumor from nontumor[6-11]. Cloning of the genes, especially the genes associated with HBV and HCV, is still very important to account for the development of liver cancer.

Traditionally, several methods were used to clone the new genes, which means to compare two population of mRNA and obtain clones of genes that expressed in one population but not in the other. Although these methods have been successful in some cases, they require many rounds of hybridization and are not well suited for the identification of rare messages. The suppression subtractive hybridization is a latest method employed in the gene cloning, which is a unique method based on selective amplification of differentially expressed sequences and overcomes technical limitation of traditional subtraction methods[12-14]. Hence, the purpose of our study is to find the differentially expressed genes in liver tumor and nontumor tissues induced by woodchuck hepatitis B virus using suppression subtractive hybridization.

MATERIALS AND METHODS
Patient samples

The tumor and nontumor tissues induced by woodchuck hepatitis B virus were obtained from Department of Pathology & Cell Biology, Thomas Jefferson University, Philadelphia, USA. The other HCC and surrounding nontumor liver tissues used for analysis were obtained from the patients who had undergone surgery for the removal of their tumors in Xijing Hospital. Fresh frozen blocks and -80 °C snap frozen paired liver and tumor samples from individual patients were collected, and were then made available for RNA extraction and in situ hybridization.

Total RNA and mRNA extraction

Total RNA and mRNA were extracted separately from tumor and nontumor tissues by using the Qiagen RNeasy Kit (Qiagen, Inc. Valencea, CA, USA) and the quality of extraction was determined by assaying 18S and 28S rRNA with agarose gel electrophoresis and ethidium bromide staining.

RT-PCR and adaptor ligation

The reverse transcriptase PCR was started with 2 μg poly-(A) + RNA isolated from tumor and nontumor tissues. Two adaptors were ligated to the fraction of Rsa I digested cDNA generated by RT-PCR. The sequence of two adaptors is as follows: Ad1: 5’-CTAATACGACTCAC-TATAGGGCTCGAGCGGCC-GCCCGGGCAGGT-3’ Ad2: 5’-TGTAGCGTGAAGACGACAGAAAGGGCGTGG-TGCGGAGGGCGGT-3’

cDNA subtraction and suppression PCR

The cDNA from tumor was referred to as tester, and the reference cDNA from nontumor as driver. The tester and driver cDNA were digested with Rsa I to obtain shorter, blunt-ended moleclules. The tester cDNA was then subdivided into two portions and each ligated with different cDNA adaptors. The driver cDNA had no adaptor. Two hybridizations were then performed. In the first hybridization, an excess of driver cDNA was added to each sample of tester for equalization and enrichment of differentially expressed gene. During the second hybridization, templates for PCR amplification were generated from differentially expressed sequence. The entire population of molecules was then subjected into PCR to amplify the desired differentially expressed genes. In the first PCR, only differentially expressed genes were amplified exponentially because of using suppression PCR. The second PCR was performed using nested primer which matched the sequence of adaptors to reduce the background and further enrich the differentially expressed genes.

Sequencing and GeneBank search of cloned genes

Following agarose gel electrophoresis, the unique fragments were eluted from the gels (using Qiagen gel extraction kit, Qiagen, Inc. Valencea, CA, USA) and cloned into pT7Blue (R) T vector (Novagen, Medison, WI, USA). Positive clones were selected by blue-white phenotype. Recombinant DNAs were isolated from minipreps of individual clones, and digested by Rsa I to check insert size, and then both strands were individually analysed by sequence analysis in the DNA sequence facility at the Kimmel Cancer Institute of Thomas Jefferson University in USA. The sequences obtained were compared with those in GeneBank using the FASTA command in the GCG software package for homology to known genes.

In situ hybridization (ISH)

The gene fragments obtained from PCR select cDNA subtraction were used as probes for in situ hybridization (ISH). ISH was conducted to verify that the subtraction hybridization procedure yielded probes whose expression was different between tumor and normal tissues. ISH was carried out using the Oncor ISH and digoxigenenin/biotin detection kits according the instruction provided by the manufacturer (Oncor, Gaithersburg, MD, USA).

RESULTS
PCR selected cDNA subtraction, cloning, sequencing and GeneBank search

PCR select cDNA subtraction generated totally 14 differentially expressed genes in tumors as compared with nontumors. Among them, 8 cDNA fragments from both tumor and nontumors had considerable homology with known genes in GeneBank (Table 1). Five genes from tumor and one gene from normal liver tissues had no homology as compared to those in the GeneBank, which implied that these may be new genes. PCR select cDNA subtraction was also performed with HBV virus X gene transfected HepG2 cells and control HepG2 cells. Ten genes were differentially expressed in HepG2X compared with HepG2 cells (data not shown). Interestingly, three genes cloned from the tumor tissue of woodchuck mouse liver shared considerable homology with sequences independently found to be upregulated in HBV-X[+] cells, suggesting that the different expressions of HBxAg effector can be independently observed in the tumor and nontumor tissues induced by woodchuck hepatitis B virus.

Table 1 Differentially expressed genes in tumor and nontumor liver induced by woodchuck hepatitis B virus.
CloneGeneBank search
Match% homology
Tumora
T8bHuman chromosome 1 (UT751,L1637).54% in 280 bp overlap
T18Unknown protein, uterine endometrium (x7723)60% in 151 bp overlap
T19Ribosomal protein L35A (x03475)88% in 91 bp overlap
T22Human T cell receptor beta chain (L166059)61% in 97 bp overlap
T6None
T7bNone
T11bNone
T24None
T25None
Nontumora
N7Human aminopeptidase N (x13276)93% in 54 bp overlap
N10Human IFN receptor gene (U10360)79% in 271 bp overlap
N11Human glutathione S-transferase (L02321)75% in 248 bp overlap
N13Beta-2 glycoprotein 1 from HepG2 (S80305)79% in 159 bp overlap
N8None
Validation and in vivo expression patterns of these genes

The cDNA fragments obtained from subtraction hybridization of tumor and nontumor tissue were then used as probes for in situ hybridization. In all cases, the probes from tumor showed transcripts that were preferentially expressed in tumor tissue compared with nontumor tissues. In contrast, the genes from nontumor tissue demonstrated strong hybridization in normal tissues, but little or no signal in tumor tissues.

DISCUSSION

Hepatocellular carcinoma is one of the major causes of the death in the world[15-20]. Although many researchers worked on HCC, the mechanism is still unclear[21-46]. It is widely accepted that HBV is closely associated with HCC, especially HBxAg. A common feature of HBV infection is the integration of HBV DNA, in whole or in part, into host chromatin[47-49]. The site of HBV integration is scattered throughout the host genome[50], making it unlikely that HBV brings about hepatocellular transformation by cis acting mechanisms in most cases. With regard to virus sequences, integration commonly occurs within a small region near the end of the virus genome[51], which is consistent with the hypothesis that transformation may be associated with the expression of one or more virus proteins from the integrated templates acting in trans. Integrated fragments of HBV DNA have been shown to make a truncated preS/S and or HBX polypeptides, both of which have trans-activting activities[52-56]. However only HBxAg transforms a mouse hepatocyte cell line in culture[57,58], and gives rise to liver tumors in at least one strain of transgenic mice[59-61]. Independent work has also shown that HBxAg stimulates the cell cycle, perhaps by the activation of a number of signal transduction pathways[62-66]. HBxAg is more consistently expressed than preS in the liver of infected patients. In addition, the findings that HBxAg binds to and inactivates the tumor suppressor p53 both in vitro and in vivo[67-69], and that it may bind to and alter the function of other transcriptional factors in the cells[70], implied that HBxAg function is important to the pathogenesis of HCC. There is some evidence that HBxAg naturally trans activates the insulin-like growth factor-1 (IGF-1) receptor[71], and may also stimulate the production of IGF-11[72], both of which may help sustain the survival and/or growth of tumor cells.

Because lots of factors are involved in the development of HCC induced by HBV and the mechanism need to be further elucidated, the new genes, especially the functional genes directly related with tumor are still worth being found in the liver tissues infected by HBV. Using the newly created method, which is the suppression subtractive hybridization, we identified the difference in gene expression which distinguished tumor from nontumor induced by woodchuck hepatitis B virus. The use of these fragments as probes for in situ hybridization of tumor and nontumor tissues verified that the PCR-selected cDNA subtraction actually yielded differences in the gene expression that distinguished tumor from nontumor, and that its differential expression may be relevant to the pathogenesis of HCC. Because of hepatitis B virus is closely associated with the development of chronic liver diseases, such as hepatitis and cirrhosis, as well as with the development of HCC, it is not known whether these differences are associated with HBxAg associated trans-activation[73-77], its inhibition of protesome function[60], its ribo/deoxy APTase[78], or AMP kinase activation[79], and/or its ability to alter signal transduction pathways[80]. However, experiments are in progress to firmly address these issues.

The results of this study showed that the up-regulation of multiple genes in tumor had considerable homology with known products from GeneBank, suggesting that the function of these genes is likely to positively regulate cell growth, while several genes generated from normal tissues suggests that these genes may be the negative regulators for cell growth. In addition, five genes from tumor and one gene from normal liver tissues had no homology as compared with entries in GeneBank, which implied that these may be new genes, and that it is very important to clone the full-length genes of these cDNA fragments to do the functional analysis. This kind of experiments are already on the way.

ACKNOWLEDGEMENTS

Prof. Bo Rong Pan in Oncology Center, Xijing Hospital has made great contributions to this article.

Footnotes

Edited by Wu XN and Ma JY

References
1.  Jiang RL, Lu QS, Luo KX. Cloning and expression of core gene cDNA of Chinese hepatitis C virus in cosmid pTM3. World J Gastroenterol. 2000;6:220-222.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Zhang SZ, Liang JJ, Qi ZT, Hu YP. Cloning of the non-structural gene 3 of hepatitis C virus and its inducible expression in cultured cells. World J Gastroenterol. 1999;5:125-127.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Yu MC, Gu CH. Mutation of hepatitis B virus and its association with liver diseases. Shijie Huaren Xiaohua Zazhi. 1999;7:978-979.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Zhou XP, Wang HY, Yang GS, Chen ZJ, Li BA, Wu MC. Cloning and expression of MXR7 gene in human HCC tissue. World J Gastroenterol. 2000;6:57-60.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Assy N, Gong Y, Zhang M, Minuk G. Appearance of an inhibitory cell nuclear antigen in rat and human serum during variable degrees of hepatic regenerative activity. World J Gastroenterol. 1999;5:103-106.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Begum NA, Mori M, Matsumata T, Takenaka K, Sugimachi K, Barnard GF. Differential display and integrin alpha 6 messenger RNA overexpression in hepatocellular carcinoma. Hepatology. 1995;22:1447-1455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
7.  Darabi AS, Gross M, Watabe M, Malafa M, Watabe K. Differen-tial gene expression in experimental hepatocellular carcinoma in-duced by woodchuck hepatitis B virus. Cancer Lett. 1995;95:153-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
8.  Inui Y, Higashiyama S, Kawata S, Tamura S, Miyagawa J, Taniguchi N, Matsuzawa Y. Expression of heparin-binding epidermal growth factor in human hepatocellular carcinoma. Gastroenterology. 1994;107:1799-1804.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Wu GS, Kar S, Carr BI. Identification of a human hepatocellular carcinoma-associated tumor suppressor gene by differential display polymerase chain reaction. Life Sci. 1995;57:1077-1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
10.  Yamashita N, Ishibashi H, Hayashida K, Kudo J, Takenaka K, Itoh K, Niho Y. High frequency of the MAGE-1 gene expression in hepatocellular carcinoma. Hepatology. 1996;24:1437-1440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 13]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
11.  Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology. 1997;25:862-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 162]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
12.  Diatchenko L, Lau YF, Campbell AP, Chenchik A, Moqadam F, Huang B, Lukyanov S, Lukyanov K, Gurskaya N, Sverdlov ED. Suppression subtractive hybridization: a method for generating differentially regulated or tissue-specific cDNA probes and libraries. Proc Natl Acad Sci USA. 1996;93:6025-6030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2262]  [Cited by in F6Publishing: 1997]  [Article Influence: 71.3]  [Reference Citation Analysis (0)]
13.  Liu J, Lian ZR, Pan JP, Hu JL, Zhu MH, Fan DM, Feitelson MA. Identification of differentially expressed genes in HBxAg trans-fected HepG2 cells by suppression subtractive hybridization. Zhonghua Xiaohua Zazhi. 2000;20:11-15.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Liu J, Lian ZR, Pan JP, Hu JL, Zhu MH, Fan DM, Feitelson MA. Function of one novel gene identified by SSH PCR differentially expressed in HBx transfected HepG2 cells. Zhonghua Yixue Zazhi. 2000;80:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Tang ZY. Advances in clinical research of hepatocellular carci-noma in China. World J Gastroenterol. 1998;4:4-7.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Wu GY, Wu CH. Gene therapy and liver diseases. World J Gastroenterol. 1998;4:18-19.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Roberts LR, LaRusso NF. Potential roles of tumor suppressor genes and microsatellite instability in hepatocellular carcinogenesis in southern African blacks. World J Gastroenterol. 2000;6:37-41.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Schmid R. Prospect of gastroenterology and hepatology in the next century. World J Gastroenterol. 1999;5:185-190.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Yip D, Findlay M, Boyer M, Tattersall MH. Hepatocellular carcinoma in central Sydney: a 10-year review of patients seen in a medical oncology department. World J Gastroenterol. 1999;5:483-487.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Lau GKK. Immunological approaches to the breakdown of hepa-titis B viral persistence. World J Gastroenterol. 1998;4:32.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Bian JC, Shen FM, Shen L, Wang TR, Wang XH, Chen GC, Wang JB. Susceptibility to hepatocellular carcinoma associated with null genotypes of GSTM1 and GSTT1. World J Gastroenterol. 2000;6:228-230.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Martins C, Kedda MA, Kew MC. Characterization of six tumor suppressor genes and microsatellite instability in hepatocellular carcinoma in southern African blacks. World J Gastroenterol. 1999;5:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Wei HS, Li DG, Lu HM. Hepatic cell apoptosis and fas gene. Shijie Huaren Xiaohua Zazhi. 1999;7:531-532.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Ning XY, Yang DH. Research and progress in vivo gene therapy for primary liver cancer. Shijie Huaren Xiaohua Zazhi. 2000;8:89-90.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Assy N, Minuk G. A comparison between previous and present histologic assessments of chronic hepatitis C viral infections in humans. World J Gastroenterol. 1999;5:107-110.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  He P, Tang ZY, Ye SL, Liu BB. Relationship between expression of alpha-fetoprotein messenger RNA and some clinical parameters of human hepatocellular carcinoma. World J Gastroenterol. 1999;5:111-115.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Sun HC, Li XM, Xue Q, Chen J, Gao DM, Tang ZY. Study of angiogenesis induced by metastatic and non-metastatic liver cancer by corneal micropocket model in nude mice. World J Gastroenterol. 1999;5:116-118.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Luo YQ, Wu MC, Cong WM. Gene expression of hepatocyte growth factor and its receptor in HCC and nontumorous liver tissues. World J Gastroenterol. 1999;5:119-121.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Wang YJ, Li MD, Wang YM, Nie QH, Chen GZ. Experimental study of bioartificial liver with cultured human liver cells. World J Gastroenterol. 1999;5:135-137.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Yang JM, Han DW, Liang QC, Zhao JL, Hao SY, Ma XH, Zhao YC. Effects of endotoxin on expression of ras, p53 and bcl-2 oncoprotein in hepatocarcinogenesis induced by thioacetamide in rats. China Natl J New Gastroenterol. 1997;3:213-217.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Yuan SL, Huang RM, Wang XJ, Song Y, Huang GQ. Reversing effect of Tanshinone on malignant phenotypes of human hepatocarcinoma cell line. World J Gastroenterol. 1998;4:317-319.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Zhao GQ, Xue L, Xu HY, Tang XM, Hu RD, Dong J. In situ hybridization assay of androgen receptor gene in hepatocarcinogenesis. World J Gastroenterol. 1998;4:503-505.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Ma ZY, Fan QS, Zhang DF. The effect of acupuncture on the IL2-IFN-NKC immunoregulatory system of mice with HAC grafting hepatocarcinoma. World J Gastroenterol. 2000;6:32.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sun HC, Li XM, Xue Q, Chen J, Gao DM, Tang ZY. Study of angiogenesis induced by metastatic and non-metastatic liver cancer by corneal micropocket model in nude mice. World J Gastroenterol. 1999;5:116-118.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Li WJ, Gao QX, Zhou GM, Wei ZQ. Micronuclei and cell survival in human liver cancer cells irradiated by 25MeV/u (40)Ar14(+). World J Gastroenterol. 1999;5:365-368.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Wu ZQ, Fan J, Qiu SJ, Zhou J, Tang ZY. The value of postoperative hepatic regional chemotherapy in prevention of recurrence after radical resection of primary liver cancer. World J Gastroenterol. 2000;6:131-133.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Yuan JH, Zhang RP, Zhang RG, Guo LX, Wang XW, Luo D, Xie Y, Xie H. Growth-inhibiting effects of taxol on human liver cancer in vitro and in nude mice. World J Gastroenterol. 2000;6:210-215.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Liu LX, Jiang HC, Zhu AL, Zhou J, Wang XQ, Wu M. Gene expression profiles in liver cancer and normal liver tissues. World J Gastroenterol. 2000;6:85.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Lin NF, Tang J, Ismael HS. Study on environmental etiology of high incidence areas of liver cancer in China. World J Gastroenterol. 2000;6:572-576.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Xu HY, Yang YL, Guan XL, Song G, Jiang AM, Shi LJ. Expression of regulating apoptosis gene and apoptosis index in primary liver cancer. World J Gastroenterol. 2000;6:721-724.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Gu GW, Zhou HG. Traditional Chinese Medicine in prevention of liver cancer. Shijie Huaren Xiaohua Zazhi. 1999;7:80-81.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Meng ZQ, Yu EX, Song MZ. Inhibition of telomerase activity of human liver cancer cell SMMC-7721 by chemotherapeutic drugs. Shijie Huaren Xiaohua Zazhi. 1999;7:252-254.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Wu YD, Song XQ, Zhou DN, Hu XH, Gan YQ, Li ZG, Liao P. Expermental and clinical study on targeting treatment of liver cancer using radionuclide-anti -AFP antibody-MMC double bomb. Shijie Huaren Xiaohua Zazhi. 1999;7:387-390.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Fu JM, Yu XF, Shao YF. Telomerase and primary liver cancer. Shijie Huaren Xiaohua Zazhi. 2000;8:461-463.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Mao H, Yuan AL, Zhao MF, Lai ZS, Zhang YL, Zhou DY. Effect of p38MAPK? signal pathway on ultrastructural change of liver cancer cells induced by VEGF. Shijie Huaren Xiaohua Zazhi. 2000;8:536-538.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Cui J, Yang DH. CSF-1 receptor/c-fms and liver cancer. Shijie Huaren Xiaohua Zazhi. 2000;8:696-697.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Okuda K. Hepatocellular carcinoma: recent progress. Hepatology. 1992;15:948-963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 372]  [Cited by in F6Publishing: 381]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
48.  Matsubara K, Tokino T. Integration of hepatitis B virus DNA and its implications for hepatocarcinogenesis. Mol Biol Med. 1990;7:243-260.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Feitelson MA, Duan LX. Hepatitis B virus X antigen in the pathogenesis of chronic infections and the development of hepatocellular carcinoma. Am J Pathol. 1997;150:1141-1157.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Tiollais P, Pourcel C, Dejean A. The hepatitis B virus. Nature. 1985;317:489-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 934]  [Cited by in F6Publishing: 919]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
51.  Dejean A, Sonigo P, Wain-Hobson S, Tiollais P. Specific hepatitis B virus integration in hepatocellular carcinoma DNA through a viral 11-base-pair direct repeat. Proc Natl Acad Sci USA. 1984;81:5350-5354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 129]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
52.  Caselmann WH, Meyer M, Kekulé AS, Lauer U, Hofschneider PH, Koshy R. A trans-activator function is generated by integration of hepatitis B virus preS/S sequences in human hepatocellular carcinoma DNA. Proc Natl Acad Sci USA. 1990;87:2970-2974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 113]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
53.  Kekulé AS, Lauer U, Meyer M, Caselmann WH, Hofschneider PH, Koshy R. The preS2/S region of integrated hepatitis B virus DNA encodes a transcriptional transactivator. Nature. 1990;343:457-461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 221]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
54.  Zahm P, Hofschneider PH, Koshy R. The HBV X-ORF encodes a transactivator: a potential factor in viral hepatocarcinogenesis. Oncogene. 1988;3:169-177.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Wollersheim M, Debelka U, Hofschneider PH. A transactivating function encoded in the hepatitis B virus X gene is conserved in the integrated state. Oncogene. 1988;3:545-552.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Schlüter V, Meyer M, Hofschneider PH, Koshy R, Caselmann WH. Integrated hepatitis B virus X and 3' truncated preS/S sequences derived from human hepatomas encode functionally active transactivators. Oncogene. 1994;9:3335-3344.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Höhne M, Schaefer S, Seifer M, Feitelson MA, Paul D, Gerlich WH. Malignant transformation of immortalized transgenic hepatocytes after transfection with hepatitis B virus DNA. EMBO J. 1990;9:1137-1145.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Seifer M, Höhne M, Schaefer S, Gerlich WH. In vitro tumorigenicity of hepatitis B virus DNA and HBx protein. J Hepatol. 1991;13 Suppl 4:S61-S65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 66]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
59.  Kim CM, Koike K, Saito I, Miyamura T, Jay G. HBx gene of hepatitis B virus induces liver cancer in transgenic mice. Nature. 1991;351:317-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 880]  [Cited by in F6Publishing: 855]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
60.  Koike K, Moriya K, Iino S, Yotsuyanagi H, Endo Y, Miyamura T, Kurokawa K. High-level expression of hepatitis B virus HBx gene and hepatocarcinogenesis in transgenic mice. Hepatology. 1994;19:810-819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 178]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
61.  Ueda H, Ullrich SJ, Gangemi JD, Kappel CA, Ngo L, Feitelson MA, Jay G. Functional inactivation but not structural mutation of p53 causes liver cancer. Nat Genet. 1995;9:41-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 250]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
62.  Benn J, Schneider RJ. Hepatitis B virus HBx protein activates Ras-GTP complex formation and establishes a Ras, Raf, MAP kinase signaling cascade. Proc Natl Acad Sci USA. 1994;91:10350-10354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 340]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
63.  Natoli G, Avantaggiati ML, Chirillo P, Costanzo A, Artini M, Balsano C, Levrero M. Induction of the DNA-binding activity of c-jun/c-fos heterodimers by the hepatitis B virus transactivator pX. Mol Cell Biol. 1994;14:989-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 88]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
64.  Benn J, Su F, Doria M, Schneider RJ. Hepatitis B virus HBx protein induces transcription factor AP-1 by activation of extracellular signal-regulated and c-Jun N-terminal mitogen-activated protein kinases. J Virol. 1996;70:4978-4985.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Cross JC, Wen P, Rutter WJ. Transactivation by hepatitis B virus X protein is promiscuous and dependent on mitogen-activated cellular serine/threonine kinases. Proc Natl Acad Sci USA. 1993;90:8078-8082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 139]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
66.  Benn J, Schneider RJ. Hepatitis B virus HBx protein deregulates cell cycle checkpoint controls. Proc Natl Acad Sci USA. 1995;92:11215-11219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 239]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
67.  Feitelson MA, Zhu M, Duan LX, London WT. Hepatitis B x antigen and p53 are associated in vitro and in liver tissues from patients with primary hepatocellular carcinoma. Oncogene. 1993;8:1109-1117.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Wang XW, Forrester K, Yeh H, Feitelson MA, Gu JR, Harris CC. Hepatitis B virus X protein inhibits p53 sequence-specific DNA binding, transcriptional activity, and association with transcription factor ERCC3. Proc Natl Acad Sci USA. 1994;91:2230-2234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 475]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
69.  Truant R, Antunovic J, Greenblatt J, Prives C, Cromlish JA. Direct interaction of the hepatitis B virus HBx protein with p53 leads to inhibition by HBx of p53 response element-directed transactivation. J Virol. 1995;69:1851-1859.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Henkler F F, Koshy R. Hepatitis B virus transcriptional activators: mechanisms and possible role in oncogenesis. J Viral Hepat. 1996;3:109-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
71.  Kim SO, Park JG, Lee YI. Increased expression of the insulin-like growth factor I (IGF-I) receptor gene in hepatocellular carcinoma cell lines: implications of IGF-I receptor gene activation by hepatitis B virus X gene product. Cancer Res. 1996;56:3831-3836.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Su Q, Liu YF, Zhang JF, Zhang SX, Li DF, Yang JJ. Expression of insulin-like growth factor II in hepatitis B, cirrhosis and hepatocellular carcinoma: its relationship with hepatitis B virus antigen expression. Hepatology. 1994;20:788-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
73.  Maguire HF, Hoeffler JP, Siddiqui A. HBV X protein alters the DNA binding specificity of CREB and ATF-2 by protein-protein interactions. Science. 1991;252:842-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 388]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
74.  Qadri I, Maguire HF, Siddiqui A. Hepatitis B virus transactivator protein X interacts with the TATA-binding protein. Proc Natl Acad Sci USA. 1995;92:1003-1007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 157]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
75.  Cheong JH, Yi M, Lin Y, Murakami S. Human RPB5, a subunit shared by eukaryotic nuclear RNA polymerases, binds human hepatitis B virus X protein and may play a role in X transactivation. EMBO J. 1995;14:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Antunović J, Lemieux N, Cromlish JA. The 17 kDa HBx protein encoded by hepatitis B virus interacts with the activation domains of Oct-1, and functions as a coactivator in the activation and repression of a human U6 promoter. Cell Mol Biol Res. 1993;39:463-482.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Huang J, Kwong J, Sun EC, Liang TJ. Proteasome complex as a potential cellular target of hepatitis B virus X protein. J Virol. 1996;70:5582-5591.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  de-Medina T, Haviv I, Noiman S, Shaul Y. The X protein of hepatitis B virus has a ribo/deoxy ATPase activity. Virology. 1994;202:401-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
79.  Dopheide TA, Azad AA. The hepatitis B virus X protein is a potent AMP kinase. J Gen Virol. 1996;77:173-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
80.  Kekulé AS, Lauer U, Weiss L, Luber B, Hofschneider PH. Hepatitis B virus transactivator HBx uses a tumour promoter signalling pathway. Nature. 1993;361:742-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 272]  [Cited by in F6Publishing: 303]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]