Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Physiol Biochem ; 51(1): 80-96, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30448843

RESUMEN

BACKGROUND/AIMS: Chronic hepatitis B virus (HBV) infection markedly increases the risk of development of hepatocellular carcinoma (HCC). Among the seven viral proteins that HBV encodes, HBV X protein (HBx) appears to have the most oncogenic potential. The mitochondria-associated HBx can induce oxidative stress in hepatocytes, leading to the production of abundant reactive oxygen species (ROS). High levels of ROS usually induce oxidative DNA damage and 8-hydroxy-2-deoxyguanosine (8-OHdG), also known as 8-oxo-7,8-dihydro-2-deoxyguanosine (8-oxodG), which is one of the major products of DNA oxidation and an important biomarker for oxidative stress and carcinogenesis. Cells have evolved a mechanism to prevent oxidized nucleotides from their incorporation into DNA through nucleotide pool sanitization enzymes of MTH1 (NUDT1), MTH2 (NUDT15), MTH3 (NUDT18) and NUDT5. However, little is known as to whether HBx can regulate the expression of those enzymes and modulate the formation and accumulation of 8-oxodG in hepatocytes. METHODS: The level of 8-oxodG was assessed by ELISA in stable HBV-producing hepatoma cell lines, an HBV infectious mouse model, HBV and HBx transgenic mice and HBV-infected patients versus their respective controls. Expression of MTH1, MTH2, MTH3 and NUDT5 was determined by a real-time quantitative PCR and western blot analysis. Transcriptional regulation of MTH1 and MTH2 expression by HBx and the effect of HBx on MTH1 and MTH2 promoter hypermethylation were examined using a luciferase reporter assay and bisulfite sequencing analysis. RESULTS: In comparison with controls, significantly higher levels of 8-oxodG were detected in the genome and culture supernatant of stable HBV-producing HepG2.2.15 cells, in the sera and liver tissues of HBV infectious mice and HBV or HBx transgenic mice, and in the sera of HBV-infected patients. Expression of HBx in hepatocytes significantly increased 8-oxodG level and reduced the expression of MTH1 and MTH2 at both mRNA and protein levels. It was also demonstrated that HBx markedly attenuated the MTH1 or MTH2 promoter activities through hypermethylation. Furthermore, enhancement of 8-oxodG production by HBx was reversible by overexpression of MTH1 and MTH2. CONCLUSION: Our data show that HBx expression results in the accumulation of 8-oxodG in hepatocytes through inhibiting the expression of MTH1 and MTH2. This may implicate that HBx may act as a tumor promoter through facilitating the mutational potential of 8-oxodG thus connecting a possible link between HBV infection and liver carcinogenesis.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Desoxiguanosina/análogos & derivados , Monoéster Fosfórico Hidrolasas/metabolismo , Pirofosfatasas/metabolismo , Transactivadores/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Metilación de ADN , Enzimas Reparadoras del ADN/genética , Desoxiguanosina/metabolismo , Hepatitis B/metabolismo , Hepatitis B/patología , Hepatitis B/virología , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/patología , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monoéster Fosfórico Hidrolasas/genética , Regiones Promotoras Genéticas , Pirofosfatasas/genética , Especies Reactivas de Oxígeno/metabolismo , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales
2.
Cell Physiol Biochem ; 44(6): 2243-2255, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29258109

RESUMEN

BACKGROUND/AIMS: Liver fatty acid-binding protein (FABP1) is a key regulator of hepatic lipid metabolism. MicroRNAs (miRNAs) are thought to be involved in nonalcoholic fatty liver disease (NAFLD), and the underlying mechanism is largely unclear. We investigated whether miRNAs influence hepatocyte steatosis by regulating the FABP1 gene. METHODS: Candidate FABP1-targeting miRNAs were evaluated using luciferase reporter assay. FABP1 expression was measured using western blotting and quantitative reverse transcription-PCR. Intracellular lipid accumulation was measured based on Oil Red O staining and intracellular triglyceride content. Hepatocyte injury was evaluated based on culture supernatant levels of alanine aminotransferase, aspartate aminotransferase, and intracellular adenosine triphosphate, and mitochondrial membrane potential. RESULTS: Dicer1 knockdown significantly elevated FABP1 expression. In total, 68 miRNAs potentially targeting FABP1 were selected; of these, miR-3941, miR-4517, and miR-4672 directly targeted the FABP1 3' untranslated region. Mimics of the three miRNAs substantially repressed FABP1 expression at translational level and led to HepG2 cell resistance to steatosis and cell injury induced by free fatty acids mixture, which rescue of FABP1 overexpression reversed. CONCLUSION: Our findings identify a novel mechanism by which miRNAs protect against hepatocyte steatosis and injury by downregulating FABP1 expression.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/genética , Regulación de la Expresión Génica , Hepatocitos/patología , MicroARNs/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Regiones no Traducidas 3' , Regulación hacia Abajo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Enfermedad del Hígado Graso no Alcohólico/patología
3.
Sci Rep ; 7: 40246, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28074857

RESUMEN

Hepatitis B virus X protein (HBx) plays important roles in viral replication and the development of hepatocellular carcinoma. HBx is a rapid turnover protein and ubiquitin-proteasome pathway has been suggested to influence HBx stability as treatment with proteasome inhibitors increases the levels of HBx protein and causes accumulation of the polyubiquitinated forms of HBx. Deubiquitinases (DUBs) are known to act by removing ubiquitin moieties from proteins and thereby reverse their stability and/or activity. However, no information is available regarding the involvement of DUBs in regulation of ubiquitylation-dependent proteasomal degradation of HBx protein. This study identified the deubiquitylating enzyme USP15 as a critical regulator of HBx protein level. USP15 was found to directly interact with HBx via binding to the HBx region between amino acid residues 51 and 80. USP15 increased HBx protein levels in a dose-dependent manner and siRNA-mediated knockdown of endogenous USP15 reduced HBx protein levels. Increased HBx stability and steady-state level by USP15 were attributable to reduced HBx ubiquitination and proteasomal degradation. Importantly, the transcriptional transactivation function of HBx is enhanced by overexpression of USP15. These results suggest that USP15 plays an essential role in stabilizing HBx and subsequently affects the biological function of HBx.


Asunto(s)
Transactivadores/metabolismo , Activación Transcripcional , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación , Línea Celular , Humanos , Proteínas Reguladoras y Accesorias Virales
4.
J Virol ; 90(4): 1729-40, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26637457

RESUMEN

UNLABELLED: Hepatitis B virus (HBV) has been implicated as a potential trigger of hepatic steatosis although molecular mechanisms involved in the pathogenesis of HBV-associated hepatic steatosis still remain elusive. Our prior work has revealed that the expression level of liver fatty acid binding protein 1 (FABP1), a key regulator of hepatic lipid metabolism, was elevated in HBV-producing hepatoma cells. In this study, the effects of HBV X protein (HBx) mediated FABP1 regulation on hepatic steatosis and the underlying mechanism were determined. mRNA and protein levels of FABP1 were measured by quantitative RT-PCR (qPCR) and Western blotting. HBx-mediated FABP1 regulation was evaluated by luciferase assay, coimmunoprecipitation, and chromatin immunoprecipitation. Hepatic lipid accumulation was measured by using Oil-Red-O staining and the triglyceride level. It was found that expression of FABP1 was increased in HBV-producing hepatoma cells, the sera of HBV-infected patients, and the sera and liver tissues of HBV-transgenic mice. Ectopic overexpression of HBx resulted in upregulation of FABP1 in HBx-expressing hepatoma cells, whereas HBx abolishment reduced FABP1 expression. Mechanistically, HBx activated the FABP1 promoter in an HNF3ß-, C/EBPα-, and PPARα-dependent manner, in which HBx increased the gene expression of HNF3ß and physically interacted with C/EBPα and PPARα. On the other hand, knockdown of FABP1 remarkably blocked lipid accumulation both in long-chain free fatty acids treated HBx-expressing HepG2 cells and in a high-fat diet-fed HBx-transgenic mice. Therefore, FABP1 is a key driver gene in HBx-induced hepatic lipid accumulation via regulation of HNF3ß, C/EBPα, and PPARα. FABP1 may represent a novel target for treatment of HBV-associated hepatic steatosis. IMPORTANCE: Accumulating evidence from epidemiological and experimental studies has indicated that chronic HBV infection is associated with hepatic steatosis. However, the molecular mechanism underlying HBV-induced pathogenesis of hepatic steatosis still remains to be elucidated. In this study, we found that expression of liver fatty acid binding protein (FABP1) was dramatically increased in the sera of HBV-infected patients and in both sera and liver tissues of HBV-transgenic mice. Forced expression of HBx led to FABP1 upregulation, whereas knockdown of FABP1 remarkably diminished lipid accumulation in both in vitro and in vivo models. It is possible that HBx promotes hepatic lipid accumulation through upregulating FABP1 in the development of HBV-induced nonalcoholic fatty liver disease. Therefore, inhibition of FABP1 might have therapeutic value in steatosis-associated chronic HBV infection.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/biosíntesis , Hígado Graso/patología , Hígado Graso/virología , Hepatitis B/complicaciones , Hepatitis B/patología , Interacciones Huésped-Patógeno , Transactivadores/metabolismo , Animales , Fusión Artificial Génica , Western Blotting , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Perfilación de la Expresión Génica , Genes Reporteros , Células Hep G2 , Hepatocitos/patología , Hepatocitos/virología , Humanos , Inmunoprecipitación , Luciferasas/análisis , Luciferasas/genética , Masculino , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Reguladoras y Accesorias Virales
5.
Biochimie ; 112: 1-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25726912

RESUMEN

Apolipoprotein F (ApoF) inhibits cholesteryl ester transfer protein (CETP) activity and plays an important role in lipid metabolism. In the present study, the full-length human ApoF promoter was cloned, and the molecular mechanism of the regulation of ApoF was investigated. The ApoF promoter displayed higher activities in hepatoma cell lines, and the -198 nt to +79 nt promoter region contained the maximum promoter activity. In the promoter region of -198 nt to -2 nt there were four putative binding sites for transcription factors ETS-1/ETS-2 (named EBS-1 to EBS-4) and one for C/EBP. Mutation of EBS-2, EBS4 and the C/EBP binding site abolished the promoter activity, and ETS-1/ETS-2 and C/EBPα could interact with corresponding binding sites. In addition, overexpression of ETS-1/2 or C/EBPα enhanced, while dominant-negative mutants of ETS-1/2 and knockdown of C/EBPα decreased, ApoF promoter activities. ETS-1 and C/EBPα associated physically, and acted synergistically to activate ApoF transcription. These results demonstrated combined activation of the ApoF promoter by liver-enriched and ubiquitous transcription factors. Direct interactions between C/EBPα and ETS-1 were important for high liver-specific expression of ApoF.


Asunto(s)
Apolipoproteínas/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína Proto-Oncogénica c-ets-2/metabolismo , Transcripción Genética , Apolipoproteínas/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-2/genética , Elementos de Respuesta
6.
FASEB J ; 29(3): 1113-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466893

RESUMEN

Hepatitis B virus core protein (HBc) has been implicated in hepatocarcinogenesis through several mechanisms. Resistance of hepatitis B virus (HBV)-infected hepatocytes to apoptosis is considered one of the major contributors to the progression of chronic hepatitis to cirrhosis and ultimately to hepatocellular carcinoma. The Fas receptor/ligand (Fas/FasL) system plays a prominent role in hepatocyte death during HBV infection. Here we report that HBc mediates resistance of hepatoma cells to agonistic anti-Fas antibody (CH11)-induced apoptosis. When HBc was introduced into human hepatoma cells, the cells became resistant to CH11 cytotoxicity in a p53-dependent manner. HBc significantly down-regulated the expression of p53, total Fas, and membrane-bound Fas at the mRNA and protein levels and reduced FasL mRNA expression. In contrast, HBc up-regulated the expression of soluble forms of Fas by increasing Fas alternative mRNA splicing. Mechanistically, HBc-mediated Fas alternative mRNA splicing was associated with up-regulation of polypyrimidine tract-binding protein 1 and down-regulation of Fas-activated serine/threonine kinase. These results indicated that HBc may prevent hepatocytes from Fas-induced apoptosis by the dual effects of reducing the expression of the proapoptotic form of Fas and enhancing the expression of the antiapoptotic form of the receptor, which may contribute to the survival and persistence of infected hepatocytes during chronic infection.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/patología , Proteína Ligando Fas/metabolismo , Antígenos del Núcleo de la Hepatitis B/metabolismo , Hepatitis B/patología , Neoplasias Hepáticas/patología , Receptor fas/metabolismo , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Proteína Ligando Fas/genética , Citometría de Flujo , Hepatitis B/metabolismo , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Receptor fas/genética
7.
BMC Cancer ; 14: 282, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24758376

RESUMEN

BACKGROUND: The risk of hepatocellular carcinoma (HCC) increases in chronic hepatitis B surface antigen (HBsAg) carriers who often have concomitant increase in the levels of benzo[alpha]pyrene-7,8-diol-9,10-epoxide(±) (BPDE)-DNA adduct in liver tissues, suggesting a possible co-carcinogenesis of Hepatitis B virus (HBV) and benzo[alpha]pyrene in HCC; however the exact mechanisms involved are unclear. METHODS: The interaction between hepatitis B spliced protein (HBSP) and microsomal epoxide hydrolase (mEH) was confirmed using GST pull-down, co-immunoprecipitation and mammalian two-hybrid assay; the effects of HBSP on mEH-mediated B[alpha]P metabolism was examined by high performance liquid chromatography (HPLC); and the influences of HBSP on B[alpha]P carcinogenicity were evaluated by bromodeoxyuridine cell proliferation, anchorage-independent growth and tumor xenograft. RESULTS: HBSP could interact with mEH in vitro and in vivo, and this interaction was mediated by the N terminal 47 amino acid residues of HBSP. HBSP could greatly enhance the hydrolysis activity of mEH in cell-free mouse liver microsomes, thus accelerating the metabolism of benzo[alpha]pyrene to produce more ultimate carcinnogen, BPDE, and this effect of HBSP requires the intact HBSP molecule. Expression of HBSP significantly increased the formation of BPDE-DNA adduct in benzo[alpha]pyrene-treated Huh-7 hepatoma cells, and this enhancement was blocked by knockdown of mEH. HBSP could enhance the cell proliferation, accelerate the G1/S transition, and promote cell transformation and tumorigenesis of B[alpha]P-treated Huh-7 hepatoma cells. CONCLUSIONS: Our results demonstrated that HBSP could promote carcinogenic effects of B[alpha]P by interacting with mEH and enhancing its hydrolysis activity.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular/genética , Epóxido Hidrolasas/metabolismo , Neoplasias Hepáticas/genética , Proteínas Virales/metabolismo , Animales , Benzopirenos/toxicidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Aductos de ADN/metabolismo , Epóxido Hidrolasas/genética , Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Humanos , Hidrólisis/efectos de los fármacos , Neoplasias Hepáticas/patología , Ratones , Microsomas/efectos de los fármacos , Microsomas/enzimología , Proteínas Virales/genética
8.
Free Radic Biol Med ; 65: 632-644, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23920313

RESUMEN

NAD(P)H:quinone oxidoreductase 1 (NQO1) is a phase II enzyme that participates in the detoxification of dopamine-derived quinone molecules and reactive oxygen species. Our prior work using a proteomic approach found that NQO1 protein levels were significantly decreased in stable hepatitis B virus (HBV)-producing hepatoma cells relative to the empty-vector-transfected controls. However, the mechanism and biological significance of the NQO1 suppression remain elusive. In this study we demonstrate that HBV X protein (HBx) induces epigenetic silencing of NQO1 in hepatoma cells through promoter hypermethylation via recruitment of DNA methyltransferase DNMT3A to the promoter region of the NQO1 gene. In HBV-related hepatocellular carcinoma (HCC) specimens, HBx expression was correlated negatively to NQO1 transcripts but positively to NQO1 promoter hypermethylation. Downregulation of NQO1 by HBx reduced intracellular glutathione levels, impaired mitochondrial function, and increased susceptibility of hepatoma cells to oxidative stress-induced cell injury. These results suggest a novel mechanism for HBV-mediated pathogenesis of chronic liver diseases, including HCC.


Asunto(s)
Carcinoma Hepatocelular/virología , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Hepáticas/virología , NAD(P)H Deshidrogenasa (Quinona)/genética , Transactivadores/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilación de ADN , Epigénesis Genética , Silenciador del Gen , Hepatitis B/genética , Humanos , Mitocondrias/patología , Estrés Oxidativo/genética , Reacción en Cadena de la Polimerasa , Proteínas Reguladoras y Accesorias Virales
9.
J Virol ; 86(24): 13533-41, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23035214

RESUMEN

Hepatitis B spliced protein (HBSP) is involved in the pathogenicity and/or persistence of hepatitis B virus (HBV). Chronic HBV infection is one of the most important risk factors for the development of hepatocellular carcinoma (HCC). However, whether or not HBSP contributes to the progression of HBV-associated HCC remains unknown. This study reports that overexpression of HBSP in human hepatoma cells increased cell invasion and motility. Conversely, small interfering RNA (siRNA)-mediated knockdown of HBSP expression inhibited migration and invasion. By glutathione S-transferase (GST) pulldown, coimmunoprecipitation, and a mammalian two-hybrid assay, HBSP was found to directly interact with cathepsin B (CTSB). Similar to HBSP knockdown, knocking down CTSB also reduced cell migration and invasion. Furthermore, the HBSP-overexpressing hepatoma cells were shown to have increased expression and activity of matrix metalloproteinase-9 (MMP-9) and urokinase-type plasminogen activator (uPA), and overexpression of HBSP significantly enhanced tumor-induced vascularization of endothelial cells. In contrast, knockdown of either HBSP or CTSB by siRNA resulted in inhibition of the two proteolytic enzymes and of the in vitro angiogenesis. Expression of HBSP in the hepatoma cells appeared to activate the mitogen-activated protein kinase (MAPK) and Akt signaling pathway, as evidenced by increases in phosphorylation of p38, Jun N-terminal protein kinase (JNK), extracellular signal-regulated kinase (ERK), and Akt. Taken together, these findings imply that interaction of HBSP with CTSB may promote hepatoma cell motility and invasion and highlight new molecular mechanisms for HBSP-induced HCC progression that involve the secretion and activation of proteolytic enzymes, increased tumor-induced angiogenesis, and activation of the MAPK/Akt signaling, thereby leading to the aggressiveness of hepatoma cells.


Asunto(s)
Carcinoma Hepatocelular/patología , Catepsina B/metabolismo , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Virales/metabolismo , Secuencia de Bases , Carcinoma Hepatocelular/irrigación sanguínea , Línea Celular Tumoral , Cartilla de ADN , Humanos , Inmunoprecipitación , Neoplasias Hepáticas/irrigación sanguínea , Neovascularización Patológica , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
J Med Virol ; 84(6): 871-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22499008

RESUMEN

Hepatitis B virus (HBV)-encoded X protein (HBx protein) is a multi-functional regulatory protein. It functions by protein-protein interaction and plays a pivotal role in the pathogenesis of HBV-related diseases. However, the partners in hepatocytes interacting with HBx protein are far from understood fully. In this study, immunoprecipitation was employed to screen for binding partners for the HBx protein from huh-7 hepatoma cells infected with recombinant adenovirus expressing HBx protein, and five cellular proteins including eukaryotic translation elongation factor 1 alpha 1 (eEF1A1), were identified. The interaction between HBx protein and eEF1A1 was confirmed further using a GST pull-down assay and co-immunoprecipitation, respectively. In Huh-7 hepatoma cells, the HBx protein inhibits dimer formation of eEF1A1, hence blocks filamentous actin bundling. These findings provide new insights into the molecular mechanisms involved in the functions of the HBx protein.


Asunto(s)
Actinas/antagonistas & inhibidores , Virus de la Hepatitis B/patogenicidad , Interacciones Huésped-Patógeno , Factor 1 de Elongación Peptídica/metabolismo , Multimerización de Proteína , Transactivadores/metabolismo , Adenoviridae/genética , Línea Celular , Vectores Genéticos , Hepatocitos/virología , Humanos , Inmunoprecipitación , Unión Proteica , Proteínas Reguladoras y Accesorias Virales
11.
Biochimie ; 94(2): 384-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21856370

RESUMEN

The human liver fatty acid binding protein (hFABP1) participates in cellular long-chain fatty acid trafficking and regulation of lipid metabolism and changes in hFABP1 are associated with an increased risk for type 2 diabetes, cardiovascular disease (CVD), and metabolic syndromes. Gene regulation of hFABP1 is not fully understood. Therefore, in the present study, the full length hFABP1 promoter (nucleotides -2125 to +51) and a series of truncated promoter regions were cloned. A luciferase reporter assay revealed that nucleotides -255 to +50 in the promoter region contained full of maximum hFABP1 promoter activity compared with the full length promoter. Furthermore high activity was shown when the plasmid was transfected into liver-derived cells such as the human hepatoblastoma cell line HepG2 and the hepatoma cell line Huh7. TFSEARCH and TESS programs were used to predict potential transcription factor binding sites. Two putative binding sites for the liver-enriched transcription factors hepatocyte nuclear factor 3ß (HNF3ß) and CCAAT/enhancer binding protein α (C/EBPα) were identified in the -255 nt to -155 nt hFABP1 promoter region. Site-directed mutagenesis of these two sites reduced dramatically hFABP1 promoter activity. In addition, the electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP) revealed that these binding sites were recognized by HNF3ß and C/EBPα respectively. Overexpression of HNF3ß and C/EBPα enhanced the transcription of hFABP1 and consequently improved the protein level of hFABP1 in HepG2 cells, while knockdown of HNF3ß and C/EBPα showed the inverse effects. Taken together, the hFABP1 gene is highly transcribed in liver-derived cells, and regulated predominantly by liver-enriched transcription factors HNF3ß and C/EBPα.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Activación Transcripcional/genética , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral , Clonación Molecular , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Unión a Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Genes Reporteros , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Transfección
12.
J Med Virol ; 82(12): 2019-26, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20981788

RESUMEN

Hepatitis B spliced protein (HBSP) encoded by a 2.2 kb singly spliced hepatitis B virus (HBV) pre-genomic RNA (spliced between positions 2447 and 489 nt) is involved in the pathogenesis of HBV infection, whereas the exact mechanism is far from being fully elucidated. In this study, a yeast two-hybrid system using HBSP as bait was employed to screen binding partners for HBSP from a human liver cDNA library. The interaction between HBSP and fibrinogen γ chain (FGG) was further confirmed in vitro using a GST pull-down assay and confirmed in vivo using a mammalian two-hybrid assay and co-immunoprecipitation. It was identified that this interaction is mediated by the N terminal 47 amino acid residues of HBSP. HBSP could inhibit fibrin polymerization, factor XIIIa-mediated fibrin cross-linking, adhesion of platelets to fibrinogen and ADP-stimulated platelet aggregation. However, the interaction-mediating fragment 1-47 of HBSP is not sufficient for the inhibitory activity on fibrinogen function. The findings suggested that HBSP may participate in the hemostatic abnormality in patients with HBV-related liver diseases.


Asunto(s)
Fibrina/metabolismo , Fibrinógeno/metabolismo , Virus de la Hepatitis B/patogenicidad , Empalme del ARN , Proteínas Virales/metabolismo , Adulto , Secuencia de Aminoácidos , Línea Celular Tumoral , Factor XIIIa/metabolismo , Biblioteca de Genes , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Humanos , Hígado/metabolismo , Datos de Secuencia Molecular , Adhesividad Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Técnicas del Sistema de Dos Híbridos , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA