Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(17): 2435-2445, 2017 04 27.
Article in English | MEDLINE | ID: mdl-27797380

ABSTRACT

Sal-like protein 4 (SALL4), an embryonic stem cell transcriptional regulator, is re-expressed by an unknown mechanism in poor prognosis hepatocellular carcinoma (HCC), often associated with chronic hepatitis B virus (HBV) infection. Herein, we investigated the mechanism of SALL4 re-expression in HBV-related HCCs. We performed bisulfite sequencing PCR of genomic DNA isolated from HBV-related HCCs and HBV replicating cells, and examined DNA methylation of a CpG island located downstream from SALL4 transcriptional start site (TSS). HBV-related HCCs expressing increased SALL4 exhibited demethylation of specific CpG sites downstream of SALL4 TSS. Similarly, SALL4 re-expression and demethylation of these CpGs was observed in HBV replicating cells. SALL4 is also re-expressed in poor prognosis HCCs of other etiologies. Indeed, increased SALL4 expression in hepatitis C virus-related HCCs correlated with demethylation of these CpG sites. To understand how CpG demethylation downstream of SALL4 TSS regulates SALL4 transcription, we quantified by chromatin immunoprecipitation (ChIP) assays RNA polymerase II occupancy of SALL4 gene, as a function of HBV replication. In absence of HBV replication, RNA polymerase II associated with SALL4 exon1. By contrast, in HBV replicating cells RNA polymerase II occupancy of all SALL4 exons increased, suggesting CpG demethylation downstream from SALL4 TSS influences SALL4 transcriptional elongation. Intriguingly, demethylated CpGs downstream from SALL4 TSS are within binding sites of octamer-binding transcription factor 4 (OCT4) and signal transducer and activator of transcription3 (STAT3). ChIP assays confirmed occupancy of these sites by OCT4 and STAT3 in HBV replicating cells, and sequential ChIP assays demonstrated co-occupancy with chromatin remodeling BRG1/Brahma-associated factors. BRG1 knockdown reduced SALL4 expression, whereas BRG1 overexpression increased SALL4 transcription in HBV replicating cells. We conclude demethylation of CpGs located within OCT4 and STAT3 cis-acting elements, downstream of SALL4 TSS, enables OCT4 and STAT3 binding, recruitment of BRG1, and enhanced RNA polymerase II elongation and SALL4 transcription.


Subject(s)
Carcinoma, Hepatocellular/pathology , DNA Methylation , Hepacivirus/physiology , Hepatitis B virus/physiology , Liver Neoplasms/pathology , Transcription Factors/genetics , Amino Acid Sequence , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , CpG Islands/genetics , DNA Helicases/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/virology , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Virus Replication
2.
Oncogene ; 35(6): 715-26, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-25893293

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major risk factor for developing hepatocellular carcinoma (HCC), and HBV X protein (HBx) acts as cofactor in hepatocarcinogenesis. In liver tumors from animals modeling HBx- and HBV-mediated hepatocarcinogenesis, downregulation of chromatin regulating proteins SUZ12 and ZNF198 induces expression of several genes, including epithelial cell adhesion molecule (EpCAM). EpCAM upregulation occurs in HBV-mediated HCCs and hepatic cancer stem cells, by a mechanism not understood. Herein we demonstrate HBx induces EpCAM expression via active DNA demethylation. In hepatocytes, EpCAM is silenced by polycomb repressive complex 2 (PRC2) and ZNF198/LSD1/Co-REST/HDAC1 chromatin-modifying complexes. Cells with stable knockdown of SUZ12, an essential PRC2 subunit, upon HBx expression demethylate a CpG dinucleotide located adjacent to NF-κB/RelA half-site. This NF-κB/RelA site is in a CpG island downstream from EpCAM transcriptional start site (TSS). Chromatin immunoprecipitation (ChIP) assays demonstrate HBx-dependent RelA occupancy of NF-κB half-site, whereas RelA knockdown suppresses CpG demethylation and EpCAM expression. Tumor necrosis factor-α activates RelA, propagating demethylation to nearby CpG sites, shown by sodium bisulfite sequencing. RelA-dependent demethylation occurring upon HBx expression requires methyltrasferase EZH2, TET2 a key factor in cytosine demethylation and inactive DNMT3L, shown by knockdown assays and sodium bisulfite sequencing. Co-immunoprecipitations and sequential ChIP assays demonstrate that RelA in the presence of HBx forms a complex with EZH2, TET2 and DNMT3L, although the role of DNMT3L remains to be understood. Interestingly, the human EpCAM gene also has a CpG island downstream from its TSS, and a NF-κB-binding site flanked by CpGs. HepG2 cells derived from human HCC exhibit demethylation of these NF-κB-flanking CpG sites, and HBV replication propagates demethylation to nearby CpG sites. DLK1, another PRC2 target gene, also upregulated in HBV-mediated HCCs, is demethylated in liver tumors at CpG dinucleotides flanking the NF-κB-binding sequence, supporting that this active DNA demethylation mechanism functions during oncogenic transformation.


Subject(s)
Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/physiology , Transcription Factor RelA/metabolism , Animals , Antigens, Neoplasm/metabolism , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dioxygenases , Enhancer of Zeste Homolog 2 Protein , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Hep G2 Cells , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Molecular Sequence Data , Multiprotein Complexes/metabolism , Promoter Regions, Genetic , Protein Binding , Viral Regulatory and Accessory Proteins
3.
Pigment Cell Res ; 14(5): 328-36, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11601654

ABSTRACT

Cells of the vertebrate neural crest (crest cells) differentiate in vitro to melanocytes and sympathoadrenal (SA) progenitor cells. We have shown previously, using primary J. quail neural crest cultures, the combinatorial effect of bone morphogenetic protein-2 (BMP-2) and cAMP signaling on SA cell development. Herein, we report that in primary J. quail neural crest cultures, BMP-2 and cAMP signaling similarly exert a combinatorial effect on melanocyte development. We demonstrate that BMP-2 treatment of neural crest cells increases melanogenesis by promoting the synthesis of melanin. This increased melanin synthesis by BMP-2 is effected by the selective increase in the transcription of the tyrosinase gene, encoding the rate-limiting enzyme of the melanin biosynthetic pathway. By contrast, BMP-2 exerts no effect on the expression of the tyrosine-related proteins 1 and 2 (Tyrpl and Dct), also involved in the melanin biosynthetic process, or on the expression of microphalmia (Mitf) gene, supporting the fact that BMP-2 does not affect melanocyte differentiation. Employing transient transfection analysis of tyrosinase-reporter constructs in B16 melanoma cells, we demonstrate that the BMP-2 response-element is localized between 900 and 1,100 bp upstream from the tyrosinase transcriptional start site. These studies support a role for BMP-2 in melanogenesis by selectively targeting the expression of the tyrosinase gene involved in melanin biosynthesis.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Gene Expression Regulation, Enzymologic , Melanins/biosynthesis , Melanocytes/physiology , Monophenol Monooxygenase/genetics , Neural Crest/drug effects , Transforming Growth Factor beta , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Bone Morphogenetic Protein 2 , Cell Differentiation , Cells, Cultured , Colforsin/pharmacology , Coturnix , Cyclic AMP/metabolism , Melanocytes/drug effects , Melanocytes/enzymology , Melanocytes/ultrastructure , Mice , Monophenol Monooxygenase/metabolism , Neural Crest/cytology , Neural Crest/enzymology , Neural Crest/physiology , Phosphodiesterase Inhibitors/pharmacology , Pigmentation/physiology , Signal Transduction/physiology
4.
J Biol Chem ; 276(37): 34671-80, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11461911

ABSTRACT

The hepatitis B virus (HBV) X protein (pX) is implicated in hepatocarcinogenesis of chronic HBV patients by an unknown mechanism. Activities of pX likely relevant to hepatocyte transformation include activation of the mitogenic RAS-RAF-MAPK and JNK pathways. To assess the importance of mitogenic pathway activation by pX in transformation, we employed a cellular model system composed of two tetracycline-regulated, pX-expressing cell lines, constructed in AML12-immortalized hepatocytes. This system includes the differentiated 3pX-1 and the de-differentiated 4pX-1 hepatocytes. Our studies have demonstrated that conditional pX expression transforms only 3pX-1 cells. Here, comparative in vitro kinase assays and various in vivo analyses demonstrate that pX affects an inverse activation of RAS-RAF-MAPK and JNK pathways in 3pX-1 versus 4pX-1 cells. Sustained pX-dependent RAS-RAF-MAPK pathway activation is observed in pX-transforming 3pX-1 cells, whereas sustained pX-dependent JNK pathway activation is observed in pX non-transforming 4pX-1 cells. This differential, pX-dependent mitogenic pathway activation affects differential activation of cAMP-response element-binding protein and c-Jun and determines the proliferative response of 3pX-1 and 4pX-1 cells. Furthermore, tetracycline-regulated, pX-NLS-expressing cell lines demonstrate that expression of the nuclear pX-NLS variant minimally activates the RAS-RAF-MAPK pathway and results in markedly reduced transformation. These results link sustained, pX-mediated activation of RAS-RAF-MAPK pathway to hepatocyte transformation.


Subject(s)
Cell Transformation, Neoplastic , Hepatocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Trans-Activators/toxicity , ras Proteins/metabolism , Cell Division , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin A/genetics , Hepatocytes/pathology , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Viral Regulatory and Accessory Proteins
5.
Oncogene ; 20(26): 3323-31, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11423982

ABSTRACT

Chronic hepatitis B virus infection is strongly associated with the development of hepatocellular carcinoma (HCC). Epithelial tumors are frequently characterized by loss of cadherin expression or function. Cadherin-dependent adhesion prevents the acquisition of a migratory and invasive phenotype, and loss of its function is itself enough for the progression from adenoma to carcinoma. The HBx protein of hepatitis B virus is thought to contribute to the development of the carcinoma, however, its role in the oncogenic and metastatic processes is far from being fully understood. We report herein the ability of HBx to disrupt intercellular adhesion in three different cell lines stably transfected with an inducible HBx expression vector. The linkage between the actin cytoskeleton and cadherin complex, which is essential for its function, is disrupted in the presence of HBx, as indicated by detergent solubility and immunoprecipitation experiments. In addition, beta-catenin was tyrosine phosphorylated in HBx-expressing cells. Inhibition of the src family of tyrosine kinases resulted in the prevention of the disruption of adherens junctions. These results suggest that HBx is able to disrupt intercellular adhesion in a src-dependent manner, and provide a novel mechanism by which HBx may contribute to the development of HCC.


Subject(s)
Adherens Junctions/drug effects , Carcinoma, Hepatocellular/etiology , Cell Transformation, Viral/physiology , Hepatitis B virus/physiology , Hepatitis B/complications , Liver Neoplasms/etiology , Trans-Activators/physiology , src-Family Kinases/physiology , Adherens Junctions/ultrastructure , Animals , Benzoquinones , Cadherins/metabolism , Cell Adhesion , Cell Line , Cell Transformation, Viral/genetics , Cocarcinogenesis , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells , Hepatitis B virus/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lactams, Macrocyclic , Mice , Neoplasm Metastasis , Phosphorylation , Protein Processing, Post-Translational , Quinones/pharmacology , Recombinant Fusion Proteins/physiology , Rifabutin/analogs & derivatives , Trans-Activators/genetics , Transfection , Viral Regulatory and Accessory Proteins , beta Catenin , src-Family Kinases/antagonists & inhibitors
6.
In Vitro Cell Dev Biol Anim ; 37(3): 185-92, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11370813

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) promotes the development of primary neural crest cells grown in tissue culture to the sympathoadrenal (SA) lineage. Independent studies have characterized the expression patterns of SA-lineage genes in developing chicken embryo; however, studies using cultured primary neural crest cells have characterized only the expression patterns of the catecholaminergic markers, tyrosine hydroxylase (TH) and catecholamines (CAs). To further explore the molecular mechanisms that control SA-cell development using the in vitro model system, it is crucial to define the expression patterns of both the catecholaminergic markers and the genes regulating SA-lineage determination. Accordingly, we defined, in the absence and presence of BMP-2, the temporal expression patterns of TH and CA, the SA lineage-determining genes ASH-1, Phox2a, and Phox2b, the GATA-2 gene, and the pan-neuronal SCG10 gene. Comparison of these data with the reported temporal and spatial patterns of expression in vivo demonstrate that the inductive steps of SA-lineage determination, including the specification of neurotransmitter identity and neuronal fate, are recapitulated in the neural-crest culture system.


Subject(s)
Adrenal Glands/cytology , Cell Differentiation/genetics , Gene Expression , Neural Crest/cytology , Phenotype , Sympathetic Nervous System/cytology , Transforming Growth Factor beta , Animals , Biomarkers/analysis , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Catecholamines/genetics , Cells, Cultured , Chick Embryo , Coturnix , DNA-Binding Proteins/genetics , GATA2 Transcription Factor , Gene Expression/drug effects , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Humans , Nerve Tissue Proteins , Neurons/cytology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/genetics
7.
Mol Cell Biol ; 20(9): 3004-14, 2000 May.
Article in English | MEDLINE | ID: mdl-10757785

ABSTRACT

Cells of the vertebrate neural crest (crest cells) are an invaluable model system to address cell fate specification. Crest cells are amenable to tissue culture, and they differentiate to a variety of neuronal and nonneuronal cell types. Earlier studies have determined that bone morphogenetic proteins (BMP-2, -4, and -7) and agents that elevate intracellular cyclic AMP (cAMP) stimulate the development of the sympathoadrenal (SA, adrenergic) lineage in neural crest cultures. To investigate whether interactive mechanisms between signaling pathways influence crest cell differentiation, we characterized the combinatorial effects of BMP-2 and cAMP-elevating agents on the development of quail trunk neural crest cells in primary culture. We report that the cAMP signaling pathway modulates both positive and negative signals influencing the development of SA cells. Specifically, we show that moderate activation of cAMP signaling promotes, in synergy with BMP-2, SA cell development and the expression of the SA lineage-determining gene Phox2a. By contrast, robust activation of cAMP signaling opposes, even in the presence of BMP-2, SA cell development and the expression of the SA lineage-determining ASH-1 and Phox2 genes. We conclude that cAMP signaling acts as a bimodal regulator of SA cell development in neural crest cultures.


Subject(s)
Adrenal Glands/embryology , Bone Morphogenetic Proteins/metabolism , Cyclic AMP/physiology , Drosophila Proteins , Neural Crest/metabolism , Transforming Growth Factor beta , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Bone Morphogenetic Protein 2 , Cell Division/drug effects , Cell Lineage , Cells, Cultured , Colforsin/pharmacology , Coturnix , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Models, Biological , Nerve Tissue Proteins , Norepinephrine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcription Factors/metabolism
8.
J Virol ; 74(1): 83-90, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10590094

ABSTRACT

The hepatitis B virus X protein (pX) interacts directly with the bZip transactivator CREB and the bZip repressors ICERIIgamma and ATF3, increasing their DNA-binding affinity in vitro and their transcriptional efficacy in vivo. However, the mechanism of bZip-pX interaction and of the pX-mediated increase in the bZip transcriptional efficacy remains to be understood. In this study with deletion mutants of pX, we delineated a 67-amino-acid region spanning residues 49 to 115 required for direct CREB, ATF3, and ICER IIgamma interaction in vitro and in vivo and increased bZip/CRE binding in vitro. Transient transfections of the pX deletion mutants in AML12 hepatocytes demonstrate that pX(49-115) is as effective as the full-length pX in enhancing the ATF3- and ICERIIgamma-mediated transrepression. However, this pX region is inactive in increasing the transactivation efficacy of CREB; additional amino acid residues present in pX(49-140) are required to mediate the increased transactivation efficacy of CREB in vivo. This requirement for different regions of pX in affecting CREB transactivation suggests that amino acid residues 115 to 140 integrate additional events in effecting pX-mediated transactivation, such as concomitant interactions with select components of the basal transcriptional apparatus.


Subject(s)
DNA-Binding Proteins/physiology , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factors/physiology , Transcriptional Activation/genetics , Basic-Leucine Zipper Transcription Factors , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , G-Box Binding Factors , Liver/cytology , Liver/metabolism , Mutation , Sequence Deletion , Transcriptional Activation/physiology , Viral Regulatory and Accessory Proteins
9.
Int J Oncol ; 15(2): 373-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10402250

ABSTRACT

The hepatitis B virus (HBV) encodes a 16.5 kDa multifunctional protein termed pX or HBx, required for transcription of the viral genome and implicated in the development of hepatocellular carcinoma (HCC) in chronic HBV-infected patients. However, the mechanism of pX-mediated hepatocarcinogenesis remains unknown. pX is a multifunctional protein exhibiting a number of activities affecting transcription, cell growth, and apoptotic cell death. Although pX does not directly bind DNA, pX is regarded as a promiscuous transactivator, acting via a dual mechanism: in the cytoplasm, pX activates mitogenic signaling cascades; in the nucleus, pX interacts directly with members of the bZip class of transcription factors and with specific components of the basal transcriptional apparatus. The focus of this review is to describe the transactivation function of pX and its role in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Trans-Activators/physiology , Transcription, Genetic , Animals , Basal Metabolism , Carcinoma, Hepatocellular/genetics , Genome, Viral , Humans , Leucine Zippers , Liver Neoplasms/genetics , Signal Transduction/physiology , Viral Regulatory and Accessory Proteins
10.
Crit Rev Eukaryot Gene Expr ; 9(1): 19-32, 1999.
Article in English | MEDLINE | ID: mdl-10200909

ABSTRACT

cAMP-response-element-binding protein, CREB, is a 43-kDa leucine zipper transcription factor identified and cloned via the study of cAMP-regulated genes. In the last decade, numerous studies have contributed much to our understanding of CREB structure, function, and CREB-mediated transcription. CREB binds to the cAMP-response-element (CRE) as a homodimer formed via the leucine zipper motif present at its C-terminus; its transcriptional activity is regulated by phosphorylation at Ser133, located within the N-terminal transactivation domain. Active, Ser133-phosphorylated CREB effects transcription of CRE-dependent genes via interaction with the 265-kDa co-activator protein CREB-binding-protein, CBP, which bridges the CRE/CREB complex to components of the basal transcriptional apparatus. This mechanism of CREB activation is effected by diverse signals, including those regulating the intracellular levels of cAMP and Ca+2, growth factors, and cellular stress. Accordingly, CREB-mediated transcription regulates diverse cellular responses, including intermediary metabolism, neuronal signaling, cell proliferation, and apoptosis. In addition to the regulation of CREB by phosphorylation, the viral oncoproteins HBV pX and HTLVI Tax regulate CREB transcriptional efficacy by an alternative mechanism, by increasing its DNA-binding affinity for viral and/or cellular CRE sites. In this review I describe key experiments that have defined the mechanism of CREB activation, with primary emphasis on emerging evidence linking CREB to cellular growth and development.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Developmental/genetics , Transcription, Genetic/genetics , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Nucleus/metabolism , Cloning, Molecular , Cyclic AMP Response Element-Binding Protein/metabolism , Molecular Sequence Data , Phosphorylation , Signal Transduction/genetics
11.
J Biol Chem ; 274(4): 2327-36, 1999 Jan 22.
Article in English | MEDLINE | ID: mdl-9890999

ABSTRACT

We report construction and characterization of tetracycline-controlled hepatitis B virus pX-expressing hepatocyte (AML12) cell lines. These cell lines were constructed in AML12 clonal isolates (clones 3 and 4), which express constitutively the tetracycline-controlled transactivator. Since pX is implicated in HCC, this immortalized hepatocyte model system was used to investigate the mechanism of pX in transformation. Clonal isolates of 3pX and 4pX lineages display conditional synthesis of pX mRNA and protein and a 2-fold increase in growth saturation density following tetracycline removal, implicating pX in monolayer overgrowth. Interestingly, only 3pX clones display pX-dependent anchorage independence. Clone 3 lineages express hepatocyte nuclear factor-1alpha and hepatocyte-specific marker genes; clone 4 lineages express hepatocyte nuclear factor-1beta and reduced levels of hepatocyte-specific marker genes, suggesting the importance of the differentiated hepatocyte in pX-mediated oncogenic transformation. Importantly, 3pX and 4pX lineages display differential expression of immediate early genes c-fos and ATF3. The pX-transforming 3pX lineage displays early, pX-dependent induction of ATF3 and prolonged induction of c-fos. The nontransforming 4pX cells display an absence of pX-dependent ATF3 induction and transient induction of c-fos. Our results support the direct link of pX expression to oncogenic transformation in 3pX lineage clones and underscore the advantage of this conditional cellular model system for studying mechanisms of pX-mediated oncogenesis.


Subject(s)
Genes, Immediate-Early , Hepatitis B Antigens/physiology , Hepatitis B virus/physiology , Liver/metabolism , Trans-Activators/physiology , Activating Transcription Factor 3 , Blotting, Northern , Cell Adhesion , Cell Division , Cell Line , Cell Transformation, Viral , Clone Cells , Gene Expression Regulation, Viral/physiology , Hepatitis B virus/immunology , Liver/cytology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , Signal Transduction , Transcription Factors/genetics , Viral Regulatory and Accessory Proteins
12.
Anim Biotechnol ; 9(2): 101-20, 1998.
Article in English | MEDLINE | ID: mdl-9713676

ABSTRACT

Transcriptional regulation of the porcine alpha-skeletal actin gene was investigated by comparative transient transfection assays in cultured mammalian cells and by direct DNA injection in skeletal muscle. Intron I sequences were necessary to direct high-level, cell-specific porcine alpha-skeletal actin expression in C2C12 myotubes, but they inhibited transcription in skeletal muscle. A 5' distal sequence (-1929 to -550), had enhancer-like activity in C2C12 myotubes and directly injected muscle, and inhibited transcription in Hela cells. In contrast, a central region (-550 to -388) enhanced basal transcription in directly injected muscle, but not in C2C12 myotubes. A distal regulatory element localized to the 3' untranslated region modulated SV40 promoter activity only in cell culture studies. These results suggest that the intragenic and 3' distal regulatory element may be differentially utilized during differentiation and maturation of skeletal muscle. All three regions decreased SV40 promoter activity in Hela cells, suggesting that they play a role in defining tissue-specific expression of porcine alpha-skeletal actin. Furthermore, different regulatory programs of alpha-skeletal actin expression appear to exist in these two experimental systems.


Subject(s)
Actins/genetics , Gene Expression Regulation , Swine/genetics , Transcription, Genetic , Actins/biosynthesis , Animals , HeLa Cells , Humans , In Vitro Techniques , Introns , Muscle, Skeletal/physiology , Promoter Regions, Genetic
13.
J Biol Chem ; 272(33): 20684-90, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9252388

ABSTRACT

The hepatitis B virus X protein interacts with the basic-region, leucine zipper protein (bZip) domain of cAMP response element-binding protein increasing its affinity for the cAMP response element site in vitro and its transcriptional efficacy in vivo (Williams, J. S., and Andrisani, O. M. (1995) Proc. Natl. Acad. Sci. U. S. A. 92, 3819-3823). Here we examine pX interactions with bZip transcription factors ATF3, gadd153/Chop10, ICER IIgamma, and NF-IL6. We demonstrate direct interactions in vitro between pX and the bZip proteins tested. In contrast MyoD and Gal4(1-147) fail to interact with pX. We also demonstrate by the mammalian two-hybrid assay the direct interaction of pX with cAMP response element- binding protein, ICER IIgamma, ATF3, and NF-IL6 in hepatocytes. In addition, pX increases the DNA binding potential of bZip proteins for their cognate DNA-binding site in vitro. In transient transfections in hepatocytes (AML12 cell line), pX increases the transcriptional efficacy of the bZip transcription factors. NF-IL6-mediated transcriptional activation is enhanced 3-fold by pX. Most interestingly, pX augments the repression mediated by bZip repressors ATF3 and ICER IIgamma, by 6- and 7-fold, respectively, demonstrating for the first time the involvement of pX in gene repression. We conclude that pX is an enhancer of the DNA binding potential of bZip transcription factors, thereby increasing the transactivation or repression efficacy of bZip-responsive genes.


Subject(s)
DNA-Binding Proteins/physiology , DNA/metabolism , Leucine Zippers , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Transcription, Genetic , Animals , Basic-Leucine Zipper Transcription Factors , Cyclic AMP Response Element-Binding Protein/metabolism , G-Box Binding Factors , Viral Regulatory and Accessory Proteins
14.
Gene ; 159(2): 281-2, 1995 Jul 04.
Article in English | MEDLINE | ID: mdl-7622066

ABSTRACT

Here we report the nucleotide sequence of the canine interleukin-2 (IL-2)-encoding cDNA. Cloning of the canine IL-2 cDNA was achieved by the polymerase chain reaction employing, as a template, a cDNA derived from mitogen-stimulated canine splenic lymphocyte mRNA. The deduced amino acid (aa) sequence of canine IL-2 consists of 155 aa and displays 84% sequence similarity to human IL-2.


Subject(s)
Dogs/genetics , Interleukin-2/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Species Specificity
15.
Proc Natl Acad Sci U S A ; 92(9): 3819-23, 1995 Apr 25.
Article in English | MEDLINE | ID: mdl-7731990

ABSTRACT

The X gene product encoded by the hepatitis B virus, termed pX, is a promiscuous transactivator of a variety of viral and cellular genes under the control of diverse cis-acting elements. Although pX does not appear to directly bind DNA, pX-responsive elements include the NF-kappa B, AP-1, and CRE (cAMP response element) sites. Direct protein-protein interactions occur between viral pX and the CRE-binding transcription factors CREB and ATF. Here we examine the mechanism of the protein-protein interactions occurring between CREB and pX by using recombinant proteins and in vitro DNA-binding assays. We demonstrate that pX interacts with the basic region-leucine zipper domain of CREB but not with the DNA-binding domain of the yeast transactivator protein Gal4. The interaction between CREB and pX increases the affinity of CREB for the CRE site by an order of magnitude, although pX does not alter the rate of CREB dimerization. Methylation interference footprinting reveals differences between the CREB DNA and CREB-pX DNA complexes. These experiments demonstrate that pX titers the way CREB interacts with the CRE DNA and suggest that the basic, DNA-binding region of CREB is the target of pX. Transfection assays in PC12 cells with the CREB-dependent somatostatin promoter demonstrate a nearly 15-fold transcriptional induction after forskolin stimulation in the presence of pX. These results support the significance of the CREB-pX protein-protein interactions in vivo.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Hepatitis B virus/metabolism , Leucine Zippers , Trans-Activators/metabolism , Animals , Baculoviridae , Cell Line , Cloning, Molecular , Cyclic AMP Response Element-Binding Protein/isolation & purification , Kinetics , Methylation , Mutagenesis, Site-Directed , PC12 Cells , Point Mutation , Protein Kinases/isolation & purification , Protein Kinases/metabolism , Rats , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Trans-Activators/isolation & purification , Transfection , Viral Regulatory and Accessory Proteins
16.
J Biol Chem ; 269(51): 32187-93, 1994 Dec 23.
Article in English | MEDLINE | ID: mdl-7798217

ABSTRACT

The cAMP-dependent protein kinase (PKA) phosphorylates CREB327/341 at a single serine residue, Ser119/133, respectively. Phosphorylation at this site creates the sequence motif SXXXS(P), a consensus site of the glycogen synthase kinase-3 (GSK-3) enzyme (Fiol, C.J., Mahrenholz, A.M., Wang, Y., Roeske, R.W., and Roach, P.J. (1987) J. Biol. Chem. 262, 14042-14048). We examined the phosphorylation of CREB at the SXXXS(P) consensus site and its role in CREB transactivation to cAMP induction. Neither isoform of the GSK-3 enzyme (GSK-3 alpha or beta) utilizes CREB as its substrate unless CREB is already phosphorylated at Ser119/133. A 13-amino acid peptide containing the sequence surrounding Ser119/133 was phosphorylated by GSK-3, at Ser115/129, only after the primary phosphorylation of the peptide by PKA (at Ser119/133), suggesting that Ser115/129 is a GSK-3 phosphoacceptor site. Mutant CREB327/341 proteins containing Ser-->Ala substitutions confirmed Ser115/129 as the only GSK-3 phosphorylation site. Transfection assays of wild type and mutant Gal4-CREB fusion proteins in PC12 cells demonstrated that Ser-->Ala substitution of residue 129 of CREB341 impairs the transcriptional response to cAMP induction. Analogous mutation in CREB327 results in 70% decrease in its transactivation response to cAMP. In undifferentiated F9 cells, which are refractory to cAMP induction, transfected GSK-3 beta kinase induces a 60-fold increase in cyclic AMP response element-dependent transcription, mediated via the endogenous CREB protein. We propose that the hierarchical phosphorylation at the PKA and GSK-3 sites of CREB are essential for cAMP control of CREB.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/physiology , Gene Expression Regulation , Serine/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/chemistry , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Molecular Sequence Data , Mutation , PC12 Cells , Peptide Mapping , Phosphorylation , Plasmids , Rabbits , Rats , Transcriptional Activation , Transfection
17.
Protein Sci ; 2(9): 1461-71, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8401230

ABSTRACT

In this paper we describe the expression and purification from bacteria of the recombinant basic leucine zipper (bZip) domain of the cAMP response element binding protein, CREB327. The bZip peptide, CREB259-327, purified to near homogeneity, maintains the sequence-specific CRE site recognition demonstrated by in vitro competition assays. Alkylation of the three cysteine residues of CREB259-327 was employed to prevent aggregation of the peptide due to cysteine oxidation. The Kd of the purified native and modified CREB259-327 for the CRE site was determined by gel retardation assays to be on the order of 10(-7) M. We employed CD spectroscopy to study the folding properties of the native and modified CREB259-327. The CD analyses of the native/modified CREB259-327 peptide demonstrated a 20% increase in the alpha-helical content upon binding to the cAMP response-element. Only a 5% increase in the alpha-helical content of CREB259-327 is observed upon binding to the AP-1 site. This observation contrasts with CREB from the GCN4 protein (Weiss, M.A., et al., 1990, Nature 347, 575-578). In addition, the two-dimensional (2D) 1H-NMR studies of the bZip CREB peptide further support the distinct features of the CREB protein, in comparison to GCN4. Analysis by CD and 2D NMR of the dimerization domain of CREB suggests that the distinct DNA binding characteristics of CREB reside in the basic portion of the bZip module.


Subject(s)
Circular Dichroism , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/genetics , Escherichia coli/genetics , Gene Expression , Leucine Zippers , Magnetic Resonance Spectroscopy , Amino Acid Sequence , Base Sequence , Chromatography, High Pressure Liquid , Cyclic AMP Response Element-Binding Protein/metabolism , Cysteine/metabolism , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Methylation , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Structure, Secondary , Transformation, Bacterial
18.
DNA Cell Biol ; 12(2): 183-90, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8471166

ABSTRACT

In this study, we report the binding constants (Kd) of the cAMP-responsive element binding protein (delta-CREB) for various cAMP-response element (CRE) motifs. We utilized purified recombinant delta CREB protein in binding reactions with natural CRE motifs found in the promoter of two neuropeptide hormone genes and with several variant CRE motifs. The Kd of delta CREB for the perfectly palindromic CRE, TGACG-TCA, found within the somatostatin promoter is estimated to be 5.0 x 10(-9) M. The Kd of delta CREB for the variant CRE motif TG_CGTCA found within the enkephalin promoter is calculated to be in the 3 x 10(-8) M. These studies provide an in vitro quantitative assessment of the binding affinity of delta CREB for various CRE motifs.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Amino Acid Sequence , Base Sequence , Cyclic AMP Response Element-Binding Protein/isolation & purification , Kinetics , Molecular Sequence Data , Protein Binding , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Biochem Cell Biol ; 70(10-11): 1277-82, 1992.
Article in English | MEDLINE | ID: mdl-1338414

ABSTRACT

Phosphorylation of the cAMP response element binding protein (CREB) by the catalytic subunit of cAMP-dependent protein kinase (cAK) has been implicated in the cAMP-dependent stimulation of gene transcription. delta-CREB, a spliced variant of CREB, and CREBtide (KRREILSRRPSYR), a synthetic peptide based on the phosphorylation sequence in delta-CREB, were tested as substrates of cAK. Phosphorylation of delta-CREB (0.17 microM) was stoichiometric within 30 s when using a concentration of cAK which approximated the intracellular level (0.2 microM). The rate of phosphorylation of delta-CREB was comparable to the rates of the best physiological substrates of cAK tested. The rate of CREBtide phosphorylation was at least as great as that of delta-CREB, indicating that the peptide retained the determinants of delta-CREB which were responsible for substrate efficacy. The apparent Km of CREBtide phosphorylation by cAK was 3.9 microM, which is 10-fold lower than that of kemptide (Km = 39 microM), the synthetic peptide substrate most often employed for cAK measurement. The Vmax values were 12.4 mumol/(min.mg) for CREBtide and 9.8 mumol/(min.mg) for kemptide. The apparent Km of CREBtide phosphorylation by cGMP-dependent protein kinase (cGK) was 2.9 microM and the Vmax value was 3.2 mumol/(min.mg). Both delta-CREB and CREBtide were phosphorylated at a much slower rate by cGK as compared with cAK, implying that the high cAK/cGK specificity exhibited by delta-CREB was retained by the peptide. Taken together, the results indicated that delta-CREB and CREBtide are among the best substrates tested for cAK and suggested that phosphorylation of CREB by this enzyme could occur in intact cells.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/physiology , Cyclic GMP/physiology , Peptides/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Kinetics , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational , Substrate Specificity
20.
Mol Mar Biol Biotechnol ; 1(4-5): 366-75, 1992.
Article in English | MEDLINE | ID: mdl-1285009

ABSTRACT

Gene constructs consisting of human growth hormone (hGH) gene driven by promoter/regulatory sequence of mouse metallothionein (mMT), viral thymidine kinase (vTK), rat cholecystokinin (rCCK), or chicken beta-actin (cBA) gene were injected into the cytoplasm of fertilized medaka eggs via the micropyle. More than 49% of the injected embryos survived at hatching. Up to 26% of the survivors showed integration of the introduced gene construct, as determined by polymerase chain reaction analysis and subsequent confirmation by Southern blot hybridization of the genomic DNA. A significant fraction of F1 progeny, derived from crosses between transgenic founders and the nontransgenic individuals, inherited the transgene. Expression of hGH gene was also observed in some of the P1 founders and F1 transgenic progeny carrying mMT-hCG or cBA-hGH gene. Furthermore, the growth performance of the P1 mMT-hGH and cBA-hGH transgenic founders and F1 cBA-hGH F1 transgenic progeny was significantly greater than their full sibling, nontransgenic individuals. In addition to the microinjection experiment, a gene construct containing the long-terminal repeat (LTR) sequence of avian Rous sarcoma virus (RSV) and rainbow trout (rt) GH2 cDNA was introduced into embryos of medaka by electroporation using an exponential decay electroporator. Approximately 70% of the electroporated embryos survived at hatching, and 20% of the survived individuals integrated RSVLTR-rtGH2 cDNA into their genomes. These two techniques will greatly enhance the ability to study regulation of gene expression in transgenic animals during differentiation and development.


Subject(s)
Animals, Genetically Modified/genetics , Gene Expression Regulation , Growth Hormone/genetics , Oryzias/genetics , Transfection/methods , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/growth & development , Base Sequence , Blotting, Southern , Cytological Techniques , DNA/analysis , DNA/chemistry , Female , Growth Hormone/biosynthesis , Immunoblotting , Male , Microinjections , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Oryzias/embryology , Oryzias/growth & development , Polymerase Chain Reaction , RNA/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...