Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Exp Cell Res ; 320(2): 188-99, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24135225

ABSTRACT

Clinically aggressive prostate cancer (PCa) is linked to androgen resistance, metastasis, and expression of neuroendocrine markers. To understand mechanism(s) of neuroendocrine differentiation (NED) of PCa epithelia, we compared neuronal differentiation occurring during embryogenesis, in primary cultures of neural crest (NC) cells, and NED in PCa cell lines (LNCaP and PC3). We demonstrate, hypoxia promotes neuronal and neuroendocrine differentiation of NC cells and PCa cells, respectively, by inducing the miR-106 b~25 cluster. In turn, miR-106b~25 comprised of miR-106b, miR-93 and miR-25, down-regulates the transcriptional repressor REST, which represses neuron-specific protein-coding and miRNA genes. In prostate tumors of high Gleason score (≥ 8), an inverse trend was observed between REST and miR-106b~25 induction. Employing miRNA PCR arrays, we identified miRNAs up-regulated by hypoxia in LNCaP cells and REST-knockdown in NC cells. Significantly, a subset of miRNAs (miR-9, miR-25, miR-30d and miR302b) is up-regulated in high Gleason score (≥ 8) PCa, suggesting a mechanism by which NED contributes to PCa malignancy. We propose that loss of REST and induction of this set of microRNAs can serve as potential novel clinical markers of advanced PCa.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Animals , Cell Hypoxia/genetics , Cells, Cultured , Coturnix , Disease Progression , Down-Regulation , Gene Expression Regulation, Developmental , Humans , Male , Neural Crest/embryology , Neural Crest/physiology , Prostatic Neoplasms/pathology
2.
Hepatology ; 56(4): 1240-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22505317

ABSTRACT

UNLABELLED: Chronic hepatitis B virus (HBV) infection is a major risk factor for developing liver cancer, and the HBV X protein (pX) has been implicated as a cofactor in hepatocyte transformation. We have shown that HBV replication as well as in vitro transformation by pX are associated with induction of the mitotic polo-like kinase 1 (Plk1) and down-regulation of the chromatin remodeling components Suz12 and Znf198. Herein, we demonstrate the same inverse relationship between Plk1 and Suz12/Znf198 in liver tumors from X/c-myc bitransgenic mice and woodchuck hepatitis virus (WHV)-infected woodchucks. Employing these animal models and the HBV replicating HepAD38 cells we examined the effect of Suz12/Znf198 down-regulation on gene expression. Genes analyzed include hepatic cancer stem cell markers BAMBI, DKK1,2, DLK1, EpCAM, MYC, and proliferation genes CCNA1, CCND2, IGFII, MCM4-6, PLK1, RPA2, and TYMS. Suz12 occupancy at the promoters of BAMBI, CCND2, DKK2, DLK1, EpCAM, and IGFII was demonstrated by chromatin immunoprecipitation in untransformed hepatocytes, but was markedly reduced in pX-transformed and Suz12 knockdown cells. Accordingly, we refer to these genes as "Suz12 repressed" genes in untransformed hepatocytes. The Suz12 repressed genes and proliferation genes were induced in HBV-replicating HepAD38 cells and, interestingly, they exhibited distinct expression profiles during hepatocellular carcinoma (HCC) progression in X/c-myc bitransgenics. Specifically, CCND2, EpCAM, and IGFII expression was elevated at the proliferative and preneoplastic stages in X/c-myc bitransgenic livers, whereas BAMBI and PLK1 were overexpressed in hepatic tumors from X/c-myc bitransgenics and WHV-infected woodchucks. Importantly, most of these genes were selectively up-regulated in HBV-induced HCCs. CONCLUSION: The distinct expression profile of the identified Suz12 repressed genes in combination with the proliferation genes hold promise as biomarkers for progression of chronic HBV infection to HCC.


Subject(s)
Carcinoma, Hepatocellular/virology , Cell Cycle Proteins/genetics , Hepatitis B virus/genetics , Liver Neoplasms/virology , Polycomb Repressive Complex 2/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Viral , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/physiopathology , Hepatocytes/pathology , Liver Neoplasms/genetics , Marmota , Mice , Mice, Transgenic , Polycomb Repressive Complex 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Random Allocation , Sensitivity and Specificity , Trans-Activators/metabolism , Transcriptional Activation , Viral Regulatory and Accessory Proteins , Virus Replication/genetics , Polo-Like Kinase 1
3.
J Biol Chem ; 285(39): 30282-93, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20624918

ABSTRACT

Hepatitis B virus X protein (pX), implicated in hepatocarcinogenesis, induces DNA damage because of re-replication and allows propagation of damaged DNA, resulting in partial polyploidy and oncogenic transformation. The mechanism by which pX allows cells with DNA damage to continue proliferating is unknown. Herein, we show pX activates Polo-like kinase 1 (Plk1) in the G(2) phase, thereby attenuating the DNA damage checkpoint. Specifically, in the G(2) phase of pX-expressing cells, the checkpoint kinase Chk1 was inactive despite DNA damage, and protein levels of claspin, an adaptor of ataxia telangiectasia-mutated and Rad3-related protein-mediated Chk1 phosphorylation, were reduced. Pharmacologic inhibition or knockdown of Plk1 restored claspin protein levels, Chk1 activation, and p53 stabilization. Also, protein levels of DNA repair protein Mre11 were decreased in the G(2) phase of pX-expressing cells but not with Plk1 knockdown. Interestingly, in pX-expressing cells, Mre11 co-immunoprecipitated with transfected Plk1 Polo-box domain, and inhibition of Plk1 increased Mre11 stability in cycloheximide-treated cells. These results suggest that pX-activated Plk1 by down-regulating Mre11 attenuates DNA repair. Importantly, concurrent inhibition of Plk1, p53, and Mre11 increased the number of pX-expressing cells with DNA damage entering mitosis, relative to Plk1 inhibition alone. By contrast, inhibition or knockdown of Plk1 reduced pX-induced polyploidy while increasing apoptosis. We conclude Plk1, activated by pX, allows propagation of DNA damage by concurrently attenuating the DNA damage checkpoint and DNA repair, resulting in polyploidy. We propose this novel Plk1 mechanism initiates pX-mediated hepatocyte transformation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transformation, Viral , DNA Damage , DNA Repair , Hepatitis B virus/metabolism , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/genetics , Cell Line , Checkpoint Kinase 1 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , G2 Phase/genetics , Hepatitis B virus/genetics , Hepatocytes/metabolism , Hepatocytes/virology , Humans , MRE11 Homologue Protein , Phosphorylation/genetics , Protein Binding/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Stability , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Viral Regulatory and Accessory Proteins , Polo-Like Kinase 1
4.
Hepatology ; 50(2): 414-23, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19472310

ABSTRACT

UNLABELLED: Chronic hepatitis B virus (HBV) infection is linked to development of hepatocellular carcinoma (HCC). The HBV X protein (pX) is implicated in HCC pathogenesis acting as a weak oncogene or a cofactor in hepatocarcinogenesis. pX induces DNA re-replication, DNA damage, and partial polyploidy in a poorly differentiated, immortalized hepatocyte cell line. In this study we employed sorted, pX-induced polyploid cells to investigate their growth and oncogenic transformation potential over the course of 70 cell doublings. Immediately after live cell-sorting, nearly 40% of pX-induced polyploid cells undergo apoptosis, whereas the surviving cells exhibit proliferation sensitive to p53. After 40 cell generations the pX-expressing polyploid cultures exhibit loss of p53 function and become growth factor- and anchorage-independent, indicative of oncogenic transformation. The pX-induced polyploid cultures in the course of 70 cell generations undergo progressively increasing DNA damage, propagate damaged DNA to daughter cells, and display increased expression of a cluster of proliferation genes shown to be elevated in human HCC, including HBV-HCC. One of these genes is the mitotic kinase Polo-like kinase 1 (Plk1). Oncogenic transformation is suppressed in the absence of pX expression, and significantly, by inhibition of Plk1. These results identify Plk1 as crucial in pX-mediated oncogenic transformation. CONCLUSION: Partial polyploidy induced by pX is not immediately associated with oncogenic transformation. Continued DNA damage for 40 cell generations is reproducibly associated with loss of p53 function, enhanced expression of Plk1, and oncogenic transformation. Because Plk1 expression is also elevated in HBV-HCC tumors, this in vitro cellular model simulates liver cancer progression and pathogenesis in chronic HBV patients. Inhibition of Plk1 activity suppresses pX-mediated oncogenic transformation, identifying Plk1 as a promising therapeutic target for HBV-mediated HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic , Liver Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/virology , Cell Line , Cell Proliferation , DNA Damage , DNA Replication , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatitis B, Chronic/complications , Liver Neoplasms/virology , Mice , Polyploidy , Tumor Suppressor Protein p53/metabolism , Viral Regulatory and Accessory Proteins , Polo-Like Kinase 1
5.
J Biol Chem ; 283(42): 28729-40, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18693245

ABSTRACT

Hepatitis B virus X protein (pX) is implicated in hepatocellular carcinoma pathogenesis by an unknown mechanism. Employing the tetracycline-regulated pX-expressing 4pX-1 cell line, derived from the murine AML12 hepatocyte cell line, we demonstrate that pX induces partial polyploidy (>4N DNA). Depletion of p53 in 4pX-1 cells increases by 5-fold the polyploid cells in response to pX expression, indicating that p53 antagonizes pX-induced polyploidy. Dual-parameter flow cytometric analyses show pX-dependent bromodeoxyuridine (BrdUrd) incorporation in 4pX-1 cells containing 4N and >4N DNA, suggesting pX induces DNA re-replication. Interestingly, pX increases expression of endogenous replication initiation factors Cdc6 and Cdtl while suppressing geminin expression, a negative regulator of rereplication. In comparison to a geminin knockdown 4pX-1 cell line used as DNA re-replication control, the Cdt1/geminin ratio is greater in 4pX-1 cells expressing pX, indicating that pX promotes DNA re-replication. In support of this conclusion, pX-expressing 4pX-1 cells, similar to the geminin knockdown 4pX-1 cells, continue to incorporate BrdUrd in the G2 phase and exhibit nuclear Cdc6 and MCM5 co-localization and the absence of geminin. In addition, pX expression activates the ATR kinase, the sensor of DNA re-replication, which in turn phosphorylates RAD17 and H2AX. Interestingly, phospho-H2AX-positive and BrdUrd -positive cells progress through mitosis, demonstrating a link between pX-induced DNA re-replication and polyploidy. Our studies high-light a novel function of pX that likely contributes to hepatocellular carcinoma pathogenesis.


Subject(s)
Cell Cycle Proteins/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Nuclear Proteins/chemistry , Polyploidy , Trans-Activators/metabolism , Animals , Cell Line , Flow Cytometry/methods , Geminin , Hepatocytes/metabolism , Mice , Microscopy, Fluorescence , Mitosis , Models, Biological , Nocodazole/pharmacology , Polymerase Chain Reaction , Viral Regulatory and Accessory Proteins
6.
J Biol Chem ; 283(37): 25455-25467, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18606816

ABSTRACT

Hepatitis B virus (HBV) X protein (pX) is implicated in hepatocellular carcinoma (HCC) pathogenesis by an unknown mechanism. Deletions or mutations of genes involved in the p53 pathway are often associated with HBV-mediated HCC, indicating rescue from p53 apoptosis is a likely mechanism in HBV-HCC pathogenesis. Herein, we determined the mechanism by which pX sensitizes hepatocytes to p53-mediated apoptosis. Although it is well established that the Rb/E2F/ARF pathway stabilizes p53, and the DNA damage-activated ATM/ATR kinases activate p53, the mechanism that coordinates these two pathways has not been determined. We demonstrate that the p38MAPK pathway activated by pX serves this role in p53 apoptosis. Specifically, the activated p38MAPK pathway stabilizes p53 via E2F1-mediated ARF expression, and also activates the transcriptional function of p53 by activating ATR. Knockdown of p53, E2F1, ATR, or p38MAPKalpha abrogates pX-mediated apoptosis, demonstrating that E2F1, ATR, and p38MAPKalpha are all essential in p53 apoptosis in response to pX. Specifically, in response to pX expression, the p38MAPK pathway activates Cdk4 and Cdk2, leading to phosphorylation of Rb, release of E2F1, and transcription of ARF. The p38MAPK pathway also activates ATR, leading to phosphorylation of p53 on Ser-18 and Ser-23, transcription of pro-apoptotic genes Bax, Fas, and Noxa, and apoptosis. In conclusion, pX sensitizes hepatocytes to p53 apoptosis via activation of the p38MAPK pathway, which couples p53 stabilization and p53 activation, by E2F1 induction and ATR activation, respectively.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , E2F1 Transcription Factor/metabolism , MAP Kinase Signaling System , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cycloheximide/pharmacology , Mice , Models, Biological , Protein Synthesis Inhibitors/pharmacology , Retinoblastoma Protein/metabolism , Serine/chemistry , Transcription, Genetic , Viral Regulatory and Accessory Proteins
7.
J Biol Chem ; 281(5): 2969-81, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16330553

ABSTRACT

Combined BMP2 and cAMP signaling induces the catechola-minergic lineage in neural crest (NC) cultures by increasing expression of the proneural transcription factor Phox2a, in a cAMP response element (CRE)-binding protein (CREB)-mediated mechanism. To determine whether CREB acts directly on Phox2a transcription induced by BMP2+cAMP-elevating agent IBMX, transient transfections of hPhox2a-reporter constructs were performed in avian NC cultures and murine, catecholaminergic CAD cells. Although BMP2+IBMX increased endogenous Phox2a expression, the 7.5-kb hPhox2a reporters expressing either luciferase or DsRed1-E5 fluorescent protein were unresponsive to BMP2+IBMX, but active in both cell types. Cell sorting of fluorescence-positive NC cells expressing the 7.5-kb hPhox2a fluorescent timer reporter differentiated to equal numbers of catecholaminergic cells as fluorescence-negative cells, suggesting inappropriate transcription from the transfected hPhox2a promoter. NC or CAD cells treated with histone deacetylase inhibitor trichostatin A and BMP2+IBMX display increased endogenous Phox2a transcription and prolonged CREB phosphorylation, indicating Phox2a chromatin remodeling is linked to CREB activation. Chromatin immunoprecipitations employing CREB, CREB-binding protein, and acetylated H4 antibodies identified two CRE half-sites at -5.5 kb in the murine Phox2a promoter, which is also conserved in the human promoter. Proximal to the CRE half-sites, within a 170-bp region, are E-box and CCAAT binding sites, also conserved in mouse and human genes. This 170-bp promoter region confers cAMP, BMP2, and enhanced BMP2+cAMP regulation to Phox2a-luciferase reporters. We conclude these CREs are functional, with CREB directly activating Phox2a transcription. Because the E-box binds bHLH proteins like ASH1 induced in NC cells by BMP2, we propose this novel 170-bp cis-acting element is a composite site, mediating the synergistic regulation by BMP2+cAMP on Phox2a transcription.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cyclic AMP/metabolism , Homeodomain Proteins/genetics , Response Elements/physiology , Transcription, Genetic , Transforming Growth Factor beta/physiology , Animals , Binding Sites , Bone Morphogenetic Protein 2 , CREB-Binding Protein , Chromatin , Cyclic AMP/physiology , Mice , Phosphorylation , Promoter Regions, Genetic
8.
J Biol Chem ; 280(49): 41025-36, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16204240

ABSTRACT

Pluripotent neural crest (NC) cells differentiate to diverse lineages, including the neuronal, sympathoadrenal lineage. In primary NC cultures, bone morphogenetic protein 2 (BMP2) requires moderate activation of cAMP signaling for induction of the sympathoadrenal lineage. However, the mechanism by which cAMP signaling synergizes with BMP2 to induce the sympathodrenal lineage is unknown. Herein, we demonstrate that moderate activation of cAMP signaling induces both transcription and activity of proneural transcription factor Phox2a. In NC cultures inhibition of cAMP-response element-binding protein (CREB)-mediated transcription by expression of dominant-negative CREB suppresses Phox2a transcription and sympathoadrenal lineage development. Interestingly, the constitutively active CREB(DIEDML), despite inducing Phox2a transcription, is insufficient for sympathoadrenal lineage development, requiring activation of the cAMP pathway. Because CREB(DIEDML)-mediates cAMP-dependent transcription without requiring activation by the cAMP-dependent protein kinase A (PKA), these results identify PKA activation as necessary in sympathoadrenal lineage development. Treatment of NC cultures with the PKA inhibitor H89 or 1-10 nm okadaic acid (OA), a serine/threonine PP2A-like phosphatase inhibitor, suppresses sympathoadrenal lineage development. Likewise, OA treatment of the CNS-derived catecholaminergic CAD cell line inhibits cAMP-mediated neuronal differentiation. Specifically, OA inhibits cAMP-mediated Phox2a dephosphorylation, cAMP-dependent Phox2a DNA binding in vitro, and cAMP- and Phox2a-dependent dopamine-beta-hydroxylase-luciferase reporter expression. Together, these results support cAMP-dependent Phox2a dephosphorylation is required for its activation. We conclude that moderate activation of cAMP signaling has dual inputs in catecholaminergic, sympathoadrenal lineage development; that is, regulation of both Phox2a transcription and activity. These results provide the first mechanistic understanding of how moderate activation of the cAMP pathway in synergy with BMP2 promotes sympathoadrenal lineage development.


Subject(s)
Catecholamines/physiology , Central Nervous System/cytology , Cyclic AMP/physiology , Homeodomain Proteins/physiology , Neural Crest/cytology , Neurons/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Bone Morphogenetic Proteins/physiology , Cell Differentiation , Cell Line , Cells, Cultured , Chick Embryo , Coturnix/embryology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Synergism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts , Gene Expression , Genetic Vectors , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Isoquinolines/pharmacology , Molecular Sequence Data , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/physiology , Phosphorylation , RNA, Messenger/analysis , Sequence Alignment , Signal Transduction , Sulfonamides/pharmacology , Sympathetic Nervous System/cytology , Transcription, Genetic , Transfection , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology
9.
Mol Cell Biol ; 24(23): 10352-65, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542843

ABSTRACT

Activation of the cellular stress pathways (c-Jun N-terminal kinase [JNK] and p38 mitogen-activated protein [MAP] kinase) is linked to apoptosis. However, whether both pathways are required for apoptosis remains unresolved. Hepatitis B virus X protein (pX) activates p38 MAP kinase and JNK pathways and, in response to weak apoptotic signals, sensitizes hepatocytes to apoptosis. Employing hepatocyte cell lines expressing pX, which was regulated by tetracycline, we investigated the mechanism of apoptosis by p38 MAP kinase and JNK pathway activation. Inhibition of the p38 MAP kinase pathway rescues by 80% the initiation of pX-mediated apoptosis, whereas subsequent apoptotic events involve both pathways. pX-mediated activation of p38 MAP kinase and JNK pathways is sustained, inducing the transcription of the death receptor family genes encoding Fas/FasL and tumor necrosis factor receptor 1 (TNFR1)/TNF-alpha and the p53-regulated Bax and Noxa genes. The pX-dependent expression of Fas/FasL and TNFR1/TNF-alpha mediates caspase 8 activation, resulting in Bid cleavage. In turn, activated Bid, acting with pX-induced Bax and Noxa, mediates the mitochondrial release of cytochrome c, resulting in the activation of caspase 9 and apoptosis. Combined antibody neutralization of FasL and TNF-alpha reduces by 70% the initiation of pX-mediated apoptosis. These results support the importance of the pX-dependent activation of both the p38 MAP kinase and JNK pathways in pX-mediated apoptosis and suggest that this mechanism of apoptosis occurs in vivo in response to weak apoptotic signals.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cytochromes c/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Flow Cytometry , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Kinetics , MAP Kinase Kinase 4 , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tetracycline/pharmacology , Time Factors , Transcription, Genetic , Transfection , Tumor Suppressor Protein p53/metabolism , Viral Regulatory and Accessory Proteins
10.
J Virol ; 76(19): 9763-72, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12208955

ABSTRACT

Hepatitis B virus X protein (pX) is implicated in hepatocarcinogenesis by an unknown mechanism. Employing a cellular model linked to pX-mediated transformation, we investigated the role of the previously reported Stat3 activation by pX in hepatocyte transformation. Our model is composed of a differentiated hepatocyte (AML12) 3pX-1 cell line that undergoes pX-dependent transformation and a dedifferentiated hepatocyte (AML12) 4pX-1 cell line that does not exhibit transformation by pX. We report that pX-dependent Stat3 activation occurs only in non-pX-transforming 4pX-1 cells and conclude that Stat3 activation is not linked to pX-mediated transformation. Maximum Stat3 transactivation requires Ser727 phosphorylation, mediated by mitogenic pathway activation. Employing dominant negative mutants and inhibitors of mitogenic pathways, we demonstrate that maximum, pX-dependent Stat3 transactivation is inhibited by the p38 mitogen-activated protein kinase (MAPK)-specific inhibitor SB 203580. Using transient-transreporter and in vitro kinase assays, we demonstrate for the first time that pX activates the p38 MAPK pathway only in 4pX-1 cells. pX-mediated Stat3 and p38 MAPK activation is Ca(2+) and c-Src dependent, in agreement with the established cellular action of pX. Importantly, pX-dependent activation of p38 MAPK inactivates Cdc25C by phosphorylation of Ser216, thus initiating activation of the G(2)/M checkpoint, resulting in 4pX-1 cell growth retardation. Interestingly, pX expression in the less differentiated hepatocyte 4pX-1 cells activates signaling pathways known to be active in regenerating hepatocytes. These results suggest that pX expression in the infected liver effects distinct mitogenic pathway activation in less differentiated versus differentiated hepatocytes.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Trans-Activators/metabolism , Trans-Activators/physiology , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Transformation, Neoplastic , DNA/metabolism , Enzyme Activation , G2 Phase , Hepatocytes/pathology , Mitosis , Phosphorylation , STAT3 Transcription Factor , Tyrosine/metabolism , Viral Regulatory and Accessory Proteins , cdc25 Phosphatases/metabolism , p38 Mitogen-Activated Protein Kinases
11.
J Biol Chem ; 277(10): 8730-40, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11756437

ABSTRACT

Hepatitis B virus (HBV) X protein (pX) is implicated in hepatocarcinogenesis of chronically infected HBV patients. To understand mechanism(s) of pX-mediated cellular transformation, we employed two tetracycline-regulated, pX-expressing cell lines, constructed in AML12 immortalized hepatocytes: one a differentiated (3pX-1) and the other a de-differentiated (4pX-1) hepatocyte cell line. Only 3pX-1 cells undergo pX-mediated transformation, via sustained Ras-Raf-mitogen-activated protein kinase pathway activation. pX-nontransforming 4pX-1 cells display sustained, pX-dependent JNK pathway activation. To understand how pX mediates different growth characteristics in 3pX-1 and 4pX-1 cells, we report, herein, comparative cell cycle analyses. pX-transforming 3pX-1 cells display pX-dependent G(1), S, and G(2)/M progression evidenced by cyclin D(1), A, and B(1) induction, and Cdc2 kinase activation. pX-nontransforming 4pX-1 cells display pX-dependent G(1) and S phase entry, followed by S phase pause and absence of Cdc2 kinase activation. Interestingly, 4pX-1 cells exhibit selective pX-induced expression of cyclin-dependent kinase inhibitor p21(Cip1), tumor suppressor p19(ARF), and proapoptotic genes bax and IGFBP-3. Despite the pX-mediated induction of growth arrest and apoptotic genes and the absence of pX-dependent Cdc2 activation, 4pX-1 cells do not undergo pX-dependent G(2)/M arrest or apoptosis. Nocodazole-treated, G(2)/M-arrested 4pX-1 cells exhibit pX-dependent formation of multinucleated cells, similar to human T-cell lymphotropic virus type I Tax-expressing cells. We propose that in 4pX-1 cells, pX deregulates the G(2)/M checkpoint, thus rescuing cells from pX-mediated apoptosis.


Subject(s)
Hepatocytes/metabolism , Proto-Oncogene Proteins c-bcl-2 , Trans-Activators/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , CDC2 Protein Kinase/metabolism , Cell Cycle , Cell Line , Cell Line, Transformed , Cyclin A/metabolism , Cyclin B/metabolism , Cyclin B1 , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Nocodazole/pharmacology , Promoter Regions, Genetic , Protein Binding , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetracycline/pharmacology , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF/metabolism , Viral Regulatory and Accessory Proteins , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...