Cellular homeostasis is regulated tightly by the activities of many cellular proteins. The subcellular localization and stability of these proteins are critical for their activities. Examples include proteins that are transported into or out of the nucleus by specific receptors through the recognition of nuclear-localization signals (NLS) and/or nuclear export signals (NES). One recently identified NES is a short, hydrophobic, leucine-rich motif that is necessary and sufficient to mediate nuclear export of large carrier proteins and mRNAs. The activity of many cellular transcription factors, oncoproteins, cell cycle regulators and tumor suppressor proteins has been reported to be regualted by their NES. Interestingly, many viral proteins also utilize the Crm1/Ran-mediated pathway, even though some of these viral proteins are thought to be small enough to passively diffuse through the nuclear pore complex (NPC). The fact that these oncogenic viral proteins have acquired NES activity and modified nuclear export implies that nuclear export may be an efficiet target for viral-mediated oncogenesis.In this study, we are examining the hypothesis that both the X protein (HBx) of hepatitis B virus (HBV) and core protein (HC-core) of hepatitis C virus (HCV), two major risk factors for hepatocellular carcinoma, may induce neoplastic transformation by disregulating the Crm1/Ran-mediated pathways. Recently, we discovered that the HBx and HC-core proteins contain functional NESs. Unlike other cellular NES-containing proteins, HBx binds to and sequesters Crm1 in the cytoplasm, thereby modulating Crm1-mediated nuclear export of other cellular proteins including the NFkB/IkBa complex. Similarly, HC-core-mediated activation of the NFkB/IkBa complex also depends on the presence of NESs. These findings suggest that multiple cellular functions associated with HBx or HC-core may be due, in part, to their influence on the Crm1/Ran-mediated pathway. Because Crm1 and its cofactor Ran GTPase also play a key role in mitosis initiation, the inactivation of Crm1 by HBx or HC-core also may induce genomic instability. Our initial results indicate that HBx induces centrosome amplification and aberrant mitosis. These results led us to generate a novel testable hypothesis that Crm1 may be a common target for viral hepatitis-mediated oncogenicity and provide a foundation for a possible involvement of Crm1 in human carcinogenesis.

Agency
National Institute of Health (NIH)
Institute
Division of Basic Sciences - NCI (NCI)
Type
Intramural Research (Z01)
Project #
1Z01BC005793-06
Application #
6435175
Study Section
(LHC)
Project Start
Project End
Budget Start
Budget End
Support Year
6
Fiscal Year
2000
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Zhao, Xuelian; Ji, Junfang; Yu, Li-Rong et al. (2015) Cell cycle-dependent phosphorylation of nucleophosmin and its potential regulation by peptidyl-prolyl cis/trans isomerase. J Mol Biochem 4:95-103
Ji, Junfang; Wang, Xin Wei (2010) A Yin-Yang balancing act of the lin28/let-7 link in tumorigenesis. J Hepatol :
Budhu, A; Chen, Y; Kim, J W et al. (2007) Induction of a unique gene expression profile in primary human hepatocytes by hepatitis C virus core, NS3 and NS5A proteins. Carcinogenesis 28:1552-60
Lim, Mi Jung; Wang, Xin Wei (2006) Nucleophosmin and human cancer. Cancer Detect Prev 30:481-90
Zhang, Yuexing; Wang, Xin-Wei; Jelovac, Danijela et al. (2005) The ErbB3-binding protein Ebp1 suppresses androgen receptor-mediated gene transcription and tumorigenesis of prostate cancer cells. Proc Natl Acad Sci U S A 102:9890-5
Wang, Wei; Budhu, Anuradha; Forgues, Marshonna et al. (2005) Temporal and spatial control of nucleophosmin by the Ran-Crm1 complex in centrosome duplication. Nat Cell Biol 7:823-30
Budhu, Anuradha S; Wang, Xin W (2005) Loading and unloading: orchestrating centrosome duplication and spindle assembly by Ran/Crm1. Cell Cycle 4:1510-4
Forgues, Marshonna; Difilippantonio, Michael J; Linke, Steven P et al. (2003) Involvement of Crm1 in hepatitis B virus X protein-induced aberrant centriole replication and abnormal mitotic spindles. Mol Cell Biol 23:5282-92
Staib, Frank; Hussain, S Perwez; Hofseth, Lorne J et al. (2003) TP53 and liver carcinogenesis. Hum Mutat 21:201-16
Linke, Steven P; Sengupta, Sagar; Khabie, Nissim et al. (2003) p53 interacts with hRAD51 and hRAD54, and directly modulates homologous recombination. Cancer Res 63:2596-605