RESUMEN
The hepatitis B Virus X (HBx) protein plays a crucial role in the HBV-induced hepatic steatosis. Fatty acid transport protein 2 (FATP2) is a key protein that is involved in hepatic lipogenesis, and it was found to be highly expressed in various metabolic diseases. However, Whether FATP2 is a key factor in the pathogenesis of HBx-induced hepatic steatosis remains unclear. In this study, we found that FATP2 was up-regulated by HBx in vitro and in vivo and participated in HBx-induced hepatic lipid accumulation. Treatment of HBx-expressing cell lines and mice with FATP2 inhibitor (FATP2i) lipofermata ameliorated HBx-induced lipid accumulation and reduced oxidative stress and inflammation caused by lipid accumulation. Moreover, the liver injury of mouse was restored after FATP2i treatment. In summary, our results reveal that FATP2 is a key driver factor for HBx-induced hepatic lipid accumulation, and inhibition of FATP2 can ameliorates lipid accumulation caused by HBx. This study provides new insights into the mechanism of HBV-induced hepatic steatosis.
Asunto(s)
Hígado Graso , Ratones , Animales , Regulación hacia Arriba , Hígado Graso/metabolismo , Línea Celular , Lípidos , Virus de la Hepatitis B/fisiologíaRESUMEN
INTRODUCTION AND OBJECTIVES: Blood glucose fluctuates severely in the diabetes (DM) and tumor microenvironment. Our previous works have found Hepatitis B virus X protein (HBx) differentially regulated metastasis and apoptosis of hepatoma cells depending on glucose concentration. We here aimed to explore whether HBx played dual roles in the angiogenesis of hepatocellular carcinoma varying on different glucose levels. MATERIALS AND METHODS: We collected conditioned medium from HBx-overexpressing cells cultured with two solubilities of glucose, and then applied to EA.hy926 cells. Alternatively, a co-culture cell system was established with hepatoma cells and EA.hy926 cells. We analyzed the angiogenesis of EA.hy926 cells with CCK8, wound-healing, transwell-migartion and tube formation experiment. ELISA was conducted to detect the secretion levels of angiogenesis-related factors. siRNAs were used to detect the P53-VEGF axis. RESULTS: HBx expressed in hepatoma cells suppressed VEGF secretion, and subsequently inhibited the proliferation, migration and tube formation of EA.hy926 cells in a high glucose condition, while attenuating these in the lower glucose condition. Furthermore, the p53-VEGF axis was required for the dual role of HBx in angiogenesis. Additionally, HBx mainly regulated the nuclear p53. CONCLUSIONS: These data suggest that the dual roles of HBx confer hepatoma cells to remain in a glucose-rich environment and escape from the glucose-low milieu through tumor vessels, promoting liver tumor progression overall. We exclusively revealed the dual role of HBx on the angiogenesis of liver tumors, which may shed new light on the mechanism and management strategy of HBV- and DM-related hepatocellular carcinoma.
Asunto(s)
Carcinoma Hepatocelular , Movimiento Celular , Proliferación Celular , Glucosa , Neoplasias Hepáticas , Neovascularización Patológica , Transducción de Señal , Transactivadores , Proteína p53 Supresora de Tumor , Factor A de Crecimiento Endotelial Vascular , Proteínas Reguladoras y Accesorias Virales , Humanos , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Glucosa/metabolismo , Línea Celular Tumoral , Células Hep G2 , Técnicas de Cocultivo , Virus de la Hepatitis B/genética , Microambiente Tumoral , AngiogénesisRESUMEN
Chronic hepatitis B virus (HBV) infections are one of the leading causes of cirrhosis and hepatocellular carcinoma. N6-methyladenosine (m6A) modification of cellular and viral RNAs is the most prevalent internal modification that occurs cotranscriptionally. Previously, we reported the dual functional role of m6A modification of HBV transcripts in the viral life cycle. Here, we show that viral HBV X (HBx) protein is responsible for the m6A modifications of viral transcripts. HBV genomes defective in HBx failed to induce m6A modifications of HBV RNAs during infection/transfection, while ectopic expression of HBx restores m6A modifications of the viral RNAs but not the mutant HBx carrying the nuclear export signal. Using chromatin immunoprecipitation assays, we provide evidence that HBx and m6A methyltransferase complexes are localized on the HBV minichromosome to achieve cotranscriptional m6A modification of viral RNAs. HBx interacts with METTL3 and 14 to carry out methylation activity and also modestly stimulates their nuclear import. This role of HBx in mediating m6A modification also extends to host phosphatase and tensin homolog (PTEN) mRNA. This study provides insight into how a viral protein recruits RNA methylation machinery to m6A-modify RNAs.
Asunto(s)
Virus de la Hepatitis B/genética , Hepatitis B Crónica/genética , Interacciones Huésped-Patógeno/genética , Metiltransferasas/genética , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales/genética , Adenosina/análogos & derivados , Adenosina/genética , Células Hep G2 , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/virología , Humanos , Fosfohidrolasa PTEN/genética , Procesamiento Postranscripcional del ARN/genética , ARN Viral/genética , Replicación Viral/genéticaRESUMEN
Neddylation is a post-translational modification that plays an important role not only in cancer development but also in regulating viral infection and replication. Upregulation of neddylation occurs in viral infections, and inhibition of neddylation can suppress viral replication. Neddylation is thought to enhance viral protein stability and replication. Neddylation has been reported to enhance the stability of the regulatory hepatitis B virus (HBV) X protein, modulate viral replication, and enhance hepatocarcinogenesis. Inhibition of neddylation using the NEDD8-activating enzyme E1 inhibitor MLN4924 inhibits viral replication, including that of HBV. Understanding of the role of neddylation in viral infections is critical for developing new therapeutic targets and potential treatment strategies. In this review, we discuss recent progress in the understanding of the effects of neddylation during viral infection, particularly in HBV infection, and strategies for curing viral infection by targeting the neddylation pathway.
Asunto(s)
Neoplasias , Virosis , Humanos , Proteína NEDD8/metabolismo , Ubiquitinas/genética , Procesamiento Proteico-Postraduccional , Virosis/tratamiento farmacológicoRESUMEN
Farnesoid X receptor (FXR) is a nuclear receptor known to play protective roles in anti-hepatocarcinogenesis and regulation of the basal metabolism of glucose, lipids, and bile acids. FXR expression is low or absent in HBV-associated hepatocarcinogenesis. Full-length HBx and HBx C-terminal truncation are frequently found in clinical HCC samples and play distinct roles in hepatocarcinogenesis by interacting with FXR or FXR signaling. However, the impact of C-terminal truncated HBx on the progression of hepatocarcinogenesis in the absence of FXR is unclear. In this study, we found that one known FXR binding protein, a C-terminal truncated X protein (HBx C40) enhanced obviously and promoted tumor cell proliferation and migration by altering cell cycle distribution and inducing apoptosis in the absence of FXR. HBx C40 enhanced the growth of FXR-deficient tumors in vivo. In addition, RNA-sequencing analysis showed that HBx C40 overexpression could affect energy metabolism. Overexpressed HSPB8 aggravated the metabolic reprogramming induced by down-regulating glucose metabolism-associated hexokinase 2 genes in HBx C40-induced hepatocarcinogenesis. Overall, our study suggests that C-terminal truncated HBx C40 synergizes with FXR deficiency by altering cell cycle distribution as well as disturbing glucose metabolism to promote HCC development.
Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinogénesis/genética , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica , Virus de la Hepatitis B/genética , Neoplasias Hepáticas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Lipid metabolic dysregulation and liver inflammation have been reported to be associated with nonalcoholic steatohepatitis (NASH), but the underlying mechanisms remain unclear. Hepatitis B virus x protein (HBx) is a risk factor for NASH. Based on metabolomic and transcriptomic screens and public database analysis, we found that HBx-expressing hepatocyte-derived prostaglandin E2 (PGE2) induced macrophage polarization imbalance via prostaglandin E2 receptor 4 (EP4) through in vitro, ex vivo, and in vivo models. Here, we revealed that the M1-type polarization of macrophages induced by endoplasmic reticulum oxidoreductase-1-like protein α (ERO1α)-dependent endoplasmic reticulum stress was associated with the HBx-related hepatic NASH phenotype. Mechanistically, HBx promoted Niemann-Pick type C1 (NPC1)/oxysterol-binding protein-related protein 5 (ORP5)-mediated cholesterol transport from the lysosome to the endoplasmic reticulum via mammalian target of rapamycin (mTOR) activation. This study provides a novel basis for screening potential biomarkers in the macrophage mTOR-cholesterol homeostasis-polarization regulatory signaling pathway and evaluating targeted interventions for HBx-associated NASH.
Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Oxiesteroles , Colesterol/metabolismo , Dinoprostona/metabolismo , Retículo Endoplásmico/metabolismo , Hepatocitos/metabolismo , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Proteína Niemann-Pick C1/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxidorreductasas/metabolismo , Oxiesteroles/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transactivadores , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatitis B virus (HBV) is a circular, and partially double-stranded DNA virus. Upon infection, the viral genome is translocated into the cell nucleus, generating the covalently closed circular DNA (cccDNA) intermediate, and forming a mini chromosome. HBV HBx is a small protein displaying multiple roles in HBV-infected cells, and in different subcellular locations. In the nucleus, the HBx protein is required to initiate and maintain viral transcription from the viral mini chromosome. In contrast, HBx also functions in the cytoplasm, where it is able to alter multiple cellular functions such as mitochondria metabolism, apoptosis and signal transduction pathways. It has been reported that in cultured cells, at low expression levels, the HBx protein is localized in the nucleus, whereas at high expression levels, it accumulates in the cytoplasm. This dynamic subcellular distribution of HBx might be essential to exert its multiple roles during viral infection. However, the mechanism that regulates different subcellular localizations of the HBx protein is unknown. We have previously taken a bioinformatics approach to investigate whether HBx might be regulated via post-translational modification, and we have proposed that the multiple nucleocytoplasmic functions of HBx might be regulated by an evolutionarily conserved mechanism via phosphorylation. In the current study, phylogenetically conserved amino acids of HBx with a high potential of phosphorylation were targeted for site-directed mutagenesis. Two conserved serine (Ser25 and Ser41), and one conserved threonine (Thr81) amino acids were replaced by either alanine or aspartic acid residues to simulate an unphosphorylated or phosphorylated state, respectively. Human hepatoma cells were transfected with increasing amounts of the HBx DNA constructs, and the cells were analyzed by fluorescence microscopy. Together, our results show that the nucleocytoplasmic distribution of the HBx protein could be regulated by phosphorylation since some of the modified proteins were mainly confined to distinct subcellular compartments. Remarkably, both HBx Ser41A, and HBx Thr81D proteins were predominantly localized within the nuclear compartment throughout the different expression levels of HBx mutants.
Asunto(s)
Carcinoma Hepatocelular/genética , Hepatitis B/genética , Neoplasias Hepáticas/genética , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales/genética , Secuencia de Aminoácidos/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Secuencia Conservada/genética , Regulación Viral de la Expresión Génica/genética , Genoma Viral/genética , Células Hep G2 , Hepatitis B/patología , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Fosforilación/genética , FilogeniaRESUMEN
Objective: To construct a recombinant HBV replication-type plasmid with liver-enriched transcription factor binding site mutation at proximal of HBV C promoter in order to elucidate the role of HBx-enhanced HBV replication. Methods: Site-directed mutagenesis technology was used to construct a recombinant plasmid with liver-enriched transcription factor binding site mutation at proximal of HBV C promoter on the basis of wild-type HBV replicating plasmid and HBV replicating plasmid lacking HBx expression. Subsequently, plasmid transfection was carried out in HBV liver cancer cell replication model and mouse replication model, and HBV replication intermediates of cells and mouse liver tissue were extracted for detection. Results: Based on the HBV replicating plasmid, the HBV replicating plasmid with liver-enriched transcription factor binding site mutation at proximal of HBV C promoter was successfully constructed. HBx-enhanced HBV replication were detected in both the HBV liver cancer replication model and the mouse replication model. After mutating liver-enriched transcription factor binding site mutation at proximal of HBV C promoter, the effect of HBx on the enhancement of HBV replication was not significantly affected. Conclusion: HBx may not enhance HBV replication through liver-enriched transcription factor binding site mutation at proximal of HBV C promoter. The role of other liver-enriched transcription factor binding sites in HBx-enhanced HBV replication needs further study.
Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Animales , Sitios de Unión , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Ratones , Mutación , Regiones Promotoras Genéticas , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales , Replicación ViralRESUMEN
The pathogenesis of hepatitis B virus ï¼HBVï¼-associated hepatocellular carcinoma ï¼HCCï¼ is complicated with the crosstalk of multiple factors and the multi-step processes. The main mechanisms underlying the HBV-induced HCC include:â integration of HBV DNA into the host hepatocyte genome to alter gene function at the insertion siteï¼resulting in host genome instability and expression of carcinogenic truncated proteins;â¡HBV gene mutations at Sï¼Cï¼and X coding regions in the genome;â¢HBV X gene-encoded HBx protein activates proto-oncogenes and inhibits tumor suppressor genesï¼leading to the HCC occurrence. In this articleï¼the recent research progress on the molecular mechanism of HBV-induced HCC is comprehensively reviewedï¼so as to provide insights into the preventionï¼early prediction and postoperative adjuvant therapy of HCC.
Asunto(s)
Carcinoma Hepatocelular , Hepatitis B , Neoplasias Hepáticas , Hepatitis B/complicaciones , Virus de la Hepatitis B/genética , Hepatocitos , HumanosRESUMEN
Hepatitis B virus (HBV) infection is the most common cause of hepatocellular carcinoma (HCC) worldwide, wherein the expression of alpha-fetoprotein (AFP) is reactivated to promote tumorgenesis. Hepatitis B virus X protein (HBx) protein encoded by the HBV virus X gene has been considered to be oncogenic and implicated in hepatocarcinogenesis. However, the relationship between HBx and abnormal AFP expression in HCC is yet to be fully understood. To explore the potential regulation of HBx on AFP re-expression in HCC, 97 HCC samples of different etiologies were analyzed, and extremely higher serum AFP levels were found in patients with HBsAg+ . Analyses of HBV-related HCC specimens showed that the expression of AFP was negatively correlated with the levels of miR-1236 and miR-329. Further analyses indicated that HBx promotes the expression of AFP by orchestrating the levels of miR-1236 and miR-329 both in vitro and in vivo. Specifically, miR-1236 and miR-329 bind to the potential target sequences in AFP mRNA 3'-untranslated region to suppress its expression. HBx transfection resulted in the significant decrement of these microRNAs and increment of AFP expression. Moreover, AFP promotes the proliferation of hepatoma cells and attenuates the proapoptotic effect of chemotherapy agents. These findings revealed a novel regulatory mechanism of HBx on the abnormal AFP expression in HCC, which may provide a therapeutic approach for combating HBV-related HCC by targeting the regulation of AFP expression.
Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , alfa-Fetoproteínas/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Proliferación Celular , Células Hep G2 , Hepatitis B/complicaciones , Hepatitis B/virología , Virus de la Hepatitis B/aislamiento & purificación , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Transactivadores/genética , Células Tumorales Cultivadas , Proteínas Reguladoras y Accesorias Virales/genética , alfa-Fetoproteínas/genéticaRESUMEN
Hepatitis B virus (HBV)-related hepatocarcinogenesis is not necessarily associated with the liver fibrotic stage and is occasionally seen at early fibrotic stages. MicroRNAs (miRNAs) are essentially 18- to 22-nucleotide-long endogenous noncoding RNAs. Aberrant miRNA expression is a common feature of various human cancers. The aberrant expression of specific miRNAs has been shown in hepatocellular carcinoma (HCC) tissue compared with nontumor tissue. Thus, we examined targetable miRNAs as a potential new biomarker related to the high risk of HBV-related hepatocarcinogenesis, toward the prevention of cancer-related deaths. HCC tissue samples from 29 patients who underwent hepatectomy at our hospital in 2002-2013 were obtained. We extracted the total RNA and analyzed it by microRNA array, real-time RT-PCR, and three comparisons: 1) HBV-related HCC and adjacent nontumor tissue, 2) HCV-related HCC and adjacent nontumor tissue, and 3) non-HBV-, non-HCV-related HCC and adjacent nontumor tissue. We also performed a functional analysis of miRNAs specific for HBV-related HCC by using HBV-positive HCC cell lines. MiR-210-3p expression was significantly increased only in the HBV-related HCC tissue samples. MiR-210-3p expression was upregulated, and the levels of its target genes were reduced in the HBV-positive HCC cells. The inhibition of miR-210-3p enhanced its target gene expression in the HBV-positive HCC cells. In addition, miR-210-3p regulated the HBx expression in HBV-infected Huh7/NTCP cells. The enhanced expression of miR-210-3p was detected specifically in HBV-related HCC and regulated various target genes, including HBx in the HBV-positive HCC cells. MiR-210-3p might, thus, be a new biomarker for the risk of HBV-related HCC.NEW & NOTEWORTHY Our present study demonstrated that miR-210-3p is the only microRNA with enhanced expression in HBV-related HCC, and the enhanced expression of miR-210-3p upregulates HBx expression. Therefore, miR-210-3p might be a pivotal biomarker of HBV-related hepatocarcinogenesis, and the inhibition of miR-210-3p could prevent inducing hepatocarcinogenesis related to HBV infection.
Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transformación Celular Viral , Virus de la Hepatitis B/metabolismo , Hepatitis B/virología , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Transactivadores/metabolismo , Anciano , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Hepatitis B/complicaciones , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Masculino , MicroARNs/genética , Transducción de Señal , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales , Replicación ViralRESUMEN
OBJECTIVE: Hepatitis B virus X protein (HBx) is a pivotal factor for HBV-induced hepatitis. Herein, we sought to investigate HBx-mediated NLR pyrin domain containing 3 (NLRP3) inflammasome activation and pyroptosis under oxidative stress. METHODS: The effect of HBx on the NLRP3 inflammasome was analyzed by enzyme-linked immunosorbent assays, quantitative reverse transcription-polymerase chain reaction, western blotting, and immunofluorescence in hepatic HL7702 cells. Pyroptosis was evaluated by western blotting, lactate dehydrogenase release, propidium iodide staining, and transmission electron microscopy. NLRP3 expression in the inflammasome from liver tissues was assessed by immunohistochemistry. RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1ß, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). HBx localized to the mitochondria, where it induced mitochondrial damage and production of mitochondrial reactive oxygen species (mitoROS). Treatment of HL7702 cells with a mitoROS scavenger attenuated HBx-induced NLRP3 activation and pyroptosis. Expression levels of NLRP3, ASC, and IL-1ß in liver tissues from patients were positively correlated with HBV DNA concentration. CONCLUSIONS: The NLRP3 inflammasome was activated by elevated mitoROS levels and mediated HBx-induced liver inflammation and hepatocellular pyroptosis under H2O2-stress conditions.
Asunto(s)
Hepatocitos/patología , Inflamasomas/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Estrés Oxidativo , Piroptosis/efectos de los fármacos , Transactivadores/farmacología , Proteínas Reguladoras y Accesorias Virales/farmacología , Proteínas Adaptadoras de Señalización CARD/sangre , Carcinoma Hepatocelular/virología , Línea Celular , ADN Viral/análisis , Expresión Génica , Virus de la Hepatitis B/genética , Hepatocitos/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias Hepáticas/virología , Mitocondrias Hepáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transactivadores/genética , Transfección , Proteínas Reguladoras y Accesorias Virales/genéticaRESUMEN
Objective: To investigate the role and mechanism of hepatitis B virus (HBV)-encoded X protein (HBx) on the regulation of lipid metabolism and proliferation of human hepatoma cell line HepG2. Methods: HepG2 cells were transiently transfected with HBx expressing plasmid, and the cell proliferation was detected by MTT assay. Lipid droplet accumulation condition was stained by Oil Red O. Western blot was used to detect the protein levels of lipid metabolism-related genes, such as CCAAT/enhancer binding protein α (C/EBPα), sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthetase (FASN) and acetyl-CoA carboxylase (ACC1). Methyl thiazolyl tetrazolium (MTT), Oil Red O staining and western blot were used to detect the effect of HBx on the regulation of lipid metabolism and proliferation of HepG2 cells under the conditions of overexpression and low expression of C/EBPα. Results: HBx had significantly promoted the proliferation of hepatoma cell line HepG2 in dose-and time-dependent manner (F = 32.82, P < 0.001; F = 58.21, P < 0.001). HBx had significantly promoted the lipid accumulation in HepG2 cells (F = 22.65, P < 0.001). Additionally, the protein levels of C/EBPα and SREBP-1 (key regulatory factors of lipid metabolism), and the rate-limiting enzymes FASN and ACC1 were significantly increased. C/EBPα overexpression had further strengthened the effect of HBx on HepG2 cell proliferation, lipid droplet accumulation, and lipid production-related gene expression. On the contrary, C/EBPα low expression had weakened HBx's promotional effect on cell proliferation, lipid droplet accumulation and lipid production-related gene expression. Conclusion: HBx may affect the lipid production and promote the proliferation of human hepatoma cell line HepG2 via the C/EBPa/SREBP-1 signaling pathway.
Asunto(s)
Metabolismo de los Lípidos , Neoplasias Hepáticas , Línea Celular , Proliferación Celular , Humanos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transactivadores , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatitis B virus (HBV) infection and aflatoxin B1 (AFB1) exposure have been recognized as independent risk factors for the occurrence and exacerbation of hepatic steatosis but their combined impacts and the potential mechanisms remain to be further elucidated. Here, we showed that exposure to AFB1 impaired mitochondrial dynamics and increased intracellular lipid droplets (LDs) in the liver of HBV-transgenic mice in vivo and the hepatitis B virus X protein (HBx)-expressing human hepatocytes both ex vivo and in vitro. HBx combined with AFB1 exposure also up-regulated receptor interaction protein 1 (RIP1), receptor interaction protein 3 (RIP3) and activated mixed lineage kinase domain like protein (MLKL), providing evidence of necrosome formation in the hepatocytes. The shift of the mitochondrial dynamics towards imbalance of fission and fusion was rescued when MLKL was inhibited in the HBx and AFB1 co-treated hepatocytes. Most importantly, based on siRNA or CRISPR/Cas9 system, we found that the combination of HBx and AFB1 exposure increased cyclooxygenase-2 (COX-2) to mediate up-regulation of RIP3 and dynamin-related protein 1 (Drp1), which in turn promoted location of RIP3-MLKL necrosome on mitochondria, subsequently exacerbated steatosis in hepatocytes. Taken together, these findings advance the understanding of mechanism associated with HBx and AFB1-induced hepatic necrosome formation, mitochondrial dysfunction and steatosis and make COX-2 a good candidate for treatment.
Asunto(s)
Aflatoxina B1/metabolismo , Ciclooxigenasa 2/metabolismo , Hígado Graso/patología , Hepatitis B/patología , Dinámicas Mitocondriales/fisiología , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Dinaminas/metabolismo , Células Hep G2 , Virus de la Hepatitis B , Hepatocitos/trasplante , Hepatocitos/virología , Humanos , Gotas Lipídicas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Trasplante HeterólogoRESUMEN
Hepatitis B virus X protein (HBx) is involved in the initiation and progression of hepatocellular carcinoma (HCC) by regulating the host protein-coding genes. In this study, we showed that HBx altered the expression of lncRNAs to promote the progression of HCC. lncRNA microarray and quantitative reverse-transcription polymerase chain reactions (qRT-PCRs) were performed to identify lncRNAs that were differentially regulated by HBx in HCC cells and tissues. Protein, mRNA, and lncRNA expression analyses; cell cycle and apoptosis analyses; loss/gain-of-function analysis were performed to delineate the consequences of WEE2-AS1 upregulation in HCC cells. WEE2-AS1 over-expressed in HCC and was positively correlated to hepatitis B virus (HBV) infection, hepatic vascular invasion, poor tumor differentiation and poor patient prognosis. WEE2-AS1 also accelerated the proliferation, migration, invasion and cell cycle progression of HCC cells. Fermitin family member 3 (FERMT3) was a downstream target of WEE2-AS1. In conclusion, there is a preliminary HBx-WEE2-AS1- FERMT3 pathway which may serve as a therapeutic target for HBV-associated HCC.
Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , ARN Largo no Codificante/genética , Transactivadores/genética , Transactivadores/metabolismo , Adulto , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/complicaciones , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , ARN sin Sentido/genética , ARN sin Sentido/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatocellular Carcinoma (HCC) is the fifth most prevalent cancer worldwide. Specially, Hepatitis B viurs X protein (HBx) is a leading factor in the progression of Hepatitis B viurs-related HCC. Nutrient-deprived tumor microenvironment also contributes to tumor development. However, the role of HBx in nutrient-deprived HCC has received little investigation. Here, we show that HBx elevates PINK1-Parkin mediating mitophagy in starvation. HBx not only increases the PINK1/Parkin gene expression but also accelerates Parkin recruitment to partial mitochondria. Further analysis indicates that, HBx either promotes mitochondrial unfolded protein response, with remarkable mitochondrial LONP1 increases, or reduces LONP1 expression in cytosol inducing LONP1-Parkin pathway, both consequently enhancing mitophagy. Moreover, the enhanced mitophagy lowers mitochondrial apoptosis in starved hepatoma cells, and Bax is implied in the machinery. In addition, we define differential centrifuge, 3000â¯g or 12,000â¯g to pellet mitochondria, as an effective method to obtain distinct mitochondria. In collect, HBx regulates diverse aspects of LONP1 and Parkin, enhancing mitophagy in starvation. This study may shed new insights into the machinery development of hepatocellular carcinoma.
Asunto(s)
Hepatitis B/virología , Neoplasias Hepáticas/virología , Mitocondrias/virología , Transactivadores/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular , Humanos , Mitofagia/fisiología , Péptido Hidrolasas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatitis B virus X protein (HBx) played a key role in the development of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). Emerging evidence has demonstrated that miR-181b and the inhibitor of growth protein 5 (ING5) participated in the pathophysiological process. However, the regulatory mechanism of HBx remained unknown. The expression of miR-181b and ING5 in HCC tissues and cell lines were examined using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Cell viability was determined using the MTT method following HCC cell lines transfection. The interaction between miR-181b and ING5 was assessed by luciferase reporter assay. The nude mice tumor model was well established to evaluate the role and biological functions of HBx on the progression of HBV-related HCC in vivo. MiR-181b was upregulated and ING5 was downregulated in HCC tissues and cell lines. As suggested by the results from in vitro and in vivo experiments, HBx downregulates the expression of the miR-181b target gene ING5, resulting in the promotion of HCC cell proliferation. HBx accelerates proliferation activity of HCC cells by increasing miR-181b expression via targeting ING5, thereby influencing the progression of HBV-related HCC.
Asunto(s)
Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , MicroARNs/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proliferación Celular , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatitis B virus X protein (HBx) acts as a multifunctional protein that regulates intracellular signalling pathways during HBV infection. It has mainly been studied in terms of its interaction with cellular proteins. Here, we show that HBx induces membrane permeabilization independently of the mitochondrial permeability transition pore complex. We generated mitochondrial outer membrane-mimic liposomes to observe the direct effects of HBx on membranes. We found that HBx induced membrane permeabilization, and the region comprising the transmembrane domain and the mitochondrial-targeting sequence was sufficient for this process. Membrane permeabilization was inhibited by nonselective channel blockers or by N-(n-nonyl)deoxynojirimycin (NN-DNJ), a viroporin inhibitor. Moreover, NN-DNJ inhibited HBx-induced mitochondrial depolarization in Huh-7 cells. Based on the results of this study, we can postulate that the HBx protein itself is sufficient to induce mitochondrial membrane permeabilization. Our finding provides important information for a strategy of HBx targeting during HBV treatment.
Asunto(s)
Virus de la Hepatitis B/fisiología , Hepatocitos/virología , Interacciones Huésped-Patógeno , Membranas Mitocondriales/fisiología , Permeabilidad , Transactivadores/metabolismo , Línea Celular , Humanos , Proteínas Reguladoras y Accesorias ViralesRESUMEN
Hepatitis B virus (HBV)-X protein (HBx) plays critical role in inducing the malignant transformation of liver cells. Alpha fetoprotein (AFP) expression is closely related to hepatocarcinogenesis. We report that Oct4, Klf4, Sox2 and c-myc expression positively associated with AFP(+)/HBV(+) hepatocellular carcinoma(HCC) tissues, and the expression of the stemness markers CD44, CD133 and EpCAM was significantly higher in AFP(+)/HBV(+) HCC tissues compared to normal liver tissues or AFP (-)/HBV(-) HCC tissues. AFP expression turned on prior to expression of Oct4, Klf4, Sox2 and c-myc, and the stemness markers CD44, CD133 and EpCAM in the normal human liver L-02 cell line or CHL cell lines upon transfection with MCV-HBx vectors. Stem-like cells generated more tumour colonies compared to primary cells, and xenografts induced tumourigenesis in nude mice. Expression of reprogramming-related proteins was significantly enhanced in HLE cells while transfected with pcDNA3.1-afp vectors. The specific PI3K inhibitor Ly294002 inhibited the effects of pcDNA3.1-afp vectors. AFP-siRNA vectors were able to inhibit tumour colony formation and reprogramming-related gene expression. Altogether, HBx stimulates AFP expression to induce natural reprogramming of liver cells, and AFP plays a critical role in promoting the initiation of HCC progenitor/stem cells. AFP may be a potential novel biotarget for combating HBV-induced hepatocarcinogenesis.
Asunto(s)
Carcinoma Hepatocelular/virología , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/fisiología , Neoplasias Hepáticas/virología , Proteínas de Neoplasias/fisiología , Transducción de Señal , Transactivadores/fisiología , alfa-Fetoproteínas/biosíntesis , Adulto , Anciano , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular , Transformación Celular Neoplásica/genética , Femenino , Hepatocitos/metabolismo , Hepatocitos/virología , Xenoinjertos , Humanos , Factor 4 Similar a Kruppel , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/virología , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/fisiología , Interferencia de ARN , Ensayo de Tumor de Célula Madre , Proteínas Reguladoras y Accesorias Virales , alfa-Fetoproteínas/genéticaRESUMEN
Hepatitis B virus X protein plays a crucial role in the pathogenesis of hepatocellular carcinoma. We previously showed that the tumor suppressor ARID2 inhibits hepatoma cell cycle progression and tumor growth. Here, we evaluated whether hepatitis B virus X protein was involved in the modulation of ARID2 expression and hepatocarcinogenesis associated with hepatitis B virus infection. ARID2 expression was downregulated in HBV-replicative hepatoma cells, HBV transgenic mice, and HBV-related clinical HCC tissues. The expression levels of HBx were negatively associated with those of ARID2 in hepatocellular carcinoma tissues. Furthermore, HBx suppressed ARID2 at transcriptional level. Mechanistically, the promoter region of ARID2 gene inhibited by HBx was located at nt-1040/nt-601 and contained potential ATOH1 binding elements. In addition, ectopic expression of ATOH1 or mutation of ATOH1 binding sites within ARID2 promoter partially abolished HBx-triggered ARID2 transcriptional repression. Functionally, ARID2 abrogated HBx-enhanced migration and proliferation of hepatoma cells, whereas depletion of ATOH1 enhanced tumorigenecity of HCC cells. Therefore, our findings suggested that deregulation of ARID2 by HBx through ATOH1 may be involved in HBV-related hepatocellular carcinoma development.