Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Hepatology ; 67(4): 1378-1391, 2018 04.
Article in English | MEDLINE | ID: mdl-29059467

ABSTRACT

Telomeres protect chromosomal ends from deterioration and have been shown to be susceptible to shortening by reactive oxygen species (ROS)-induced damage. ROS levels increase during the progression from early to advanced hepatocellular carcinoma (HCC). An independent study found that the telomeres in most HCC tissues lengthened during carcinogenic advancement. Activated telomerase has been hypothesized to elongate telomeres during the progression of malignant HCC, but it remains unclear which signaling pathway is necessary for telomerase activation in HCC. Here, we showed using cell lines derived from human HCC that H2 O2 , which is a major component of ROS in living organisms, elongates telomeres by increasing telomerase activity through protein kinase B (AKT) activation. The AKT inhibitor, perifosine, decreased telomere length, cellular viability, and H2 O2 -mediated migration and invasion capacity in HCC cells while also inhibiting AKT activation, telomere maintenance, and tumor growth in nude mice. Advanced HCC tissues showed a positive correlation among ROS levels, phosphorylated AKT (pAKT) levels, and telomere length. Furthermore, patients with HCC tumors that have high ROS levels and long telomeres displayed poorer survival rates. These data demonstrate the significant utilities of ROS levels, pAKT levels, and telomere length for predicting a poor prognosis in patients with HCC. Taken together, AKT activation could be essential for telomere maintenance in advanced HCC tumors as well as being an important contributor to malignant HCC progression. CONCLUSION: We showed that H2 O2 contributes to telomere elongation through AKT activation in advanced HCC, suggesting that an AKT inhibitor such as perifosine may be useful for treating patients with malignant HCC. (Hepatology 2018;67:1378-1391).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Telomere Homeostasis/genetics , Telomere/metabolism , Adult , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Migration Assays , Female , Heterografts , Humans , In Situ Hybridization, Fluorescence , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Recurrence, Local , Reactive Oxygen Species/pharmacology , Real-Time Polymerase Chain Reaction , Risk Factors , Survival Rate
2.
Hepatology ; 68(5): 1851-1864, 2018 11.
Article in English | MEDLINE | ID: mdl-29672884

ABSTRACT

Dyskerin pseudouridine synthase 1 (DKC1) is a conserved gene encoding the RNA-binding protein dyskerin, which is an essential component of the telomerase holoenzyme. DKC1 up-regulation is frequently observed in many different human cancers including hepatocellular carcinoma (HCC); however, its regulatory mechanisms remain unclear. Thus, we investigated the regulatory mechanism of DKC1 in HCC progression. We found that protein-disulfide isomerase-associated 3 (PDIA3) interacted with the DKC1 regulatory DNA in HCC cells but not in HCC cells with elevated reactive oxygen species (ROS) levels, using liquid chromatographic-tandem mass spectrometric analysis after isolating the DKC1 regulatory region binding proteins. PDIA3 repressed DKC1 expression in HCC cells by recognizing the G-quadruplex DNA at the DKC1 location. However, oxidative modification of PDIA3 induced by ROS redistributed this protein into the cytosolic regions, which stimulated DKC1 expression. We also identified Met338 in PDIA3 as the oxidatively modified residue and validated the effect of oxidative modification using an ectopic expression system, a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 knock-in system, and a xenograft mouse model. We observed that oxidatively modified PDIA3 promoted DKC1-mediated malignancy and survival of HCC cells in vitro and in vivo. HCC tissues showed a positive association with ROS, cytoplasmic PDIA3, and nuclear DKC1 levels. HCC patients with high PDIA3 protein and DKC1 mRNA levels also displayed reduced recurrence-free survival rates. Cumulatively, the results showed that cytoplasmic PDIA3 activity could be essential in raising DKC1 expression in HCC progression and predicting poor prognoses in HCC patients. Conclusion: Our study indicates that the elevated ROS levels in HCC modulate cytoplasmic PDIA3 levels, resulting in HCC cell survival through DKC1 up-regulation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/mortality , Mice , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Survival Rate
3.
Hepatology ; 68(6): 2285-2300, 2018 12.
Article in English | MEDLINE | ID: mdl-30300952

ABSTRACT

Class I phosphoinositide 3-kinase (PI3K) signaling is a major pathway in human cancer development and progression. Among the four PI3K isoforms, PI3Kα and PI3KƟ are ubiquitously expressed, whereas PI3KƎĀ³ and PI3KƎĀ“ are found primarily in leukocytes. Until now, PI3K targeting in solid tumors has focused on inhibiting PI3Kα-mediated and PI3KƟ-mediated cancer cell-intrinsic PI3K activity. The role of PI3KƎĀ“ in solid tumors is unknown. Here, we evaluated the effects of PI3KƎĀ“ using established hepatocellular carcinoma (HCC) cells, malignant hepatocytes derived from patients with advanced HCC, murine models, and HCC tissues using RNA sequencing, quantitative PCR, immunoblotting, immunofluorescence, microarray, liquid chromatography-tandem mass spectrometry, and kinase assay. We established a chemical carcinogenesis model of liver malignancy that reflects the malignant phenotype and the in vivo environment of advanced HCC. In this in vivo advanced HCC-mimic system using HCC cells treated with hydrogen peroxide (H2 O2 ), we showed that H2 O2 selectively increases PI3KƎĀ“ activity while decreasing that of other class I PI3Ks. Blocking PI3KƎĀ“ activity with a PI3KƎĀ“ inhibitor or small interfering RNA-mediated PI3KƎĀ“ gene silencing inhibited HCC-cell proliferation and dampened key features of malignant HCC, including the up-regulation of telomerase reverse transcriptase (TERT). Mechanistically, H2 O2 induced oxidative modification of the serpin peptidase inhibitor, serpin peptidase inhibitor (SERPINA3), blocking its ubiquitin-dependent degradation and enhancing its activity as a transcriptional activator of PI3KƎĀ“ and TERT. High PI3KƎĀ“ levels in HCC were found to correlate with poor survival rates, with human advanced HCC showing positive correlations between the protein levels of oxidized SERPINA3, PI3KƎĀ“, and TERT. Thus, PI3KƎĀ“ plays significant roles in malignant liver tumors. Conclusion: Our data identify PI3KƎĀ“ inhibition, recently approved for the treatment of human B-cell malignancies, as a potential treatment for HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Liver Neoplasms/metabolism , Purines/therapeutic use , Quinazolinones/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Screening Assays, Antitumor , Humans , Hydrogen Peroxide , Liver Neoplasms/drug therapy , Mice , Molecular Targeted Therapy , Purines/pharmacology , Quinazolinones/pharmacology , Serpins/metabolism , Telomerase/metabolism
4.
Proc Natl Acad Sci U S A ; 113(31): 8771-6, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27439875

ABSTRACT

The role of cereblon (CRBN) in T cells is not well understood. We generated mice with a deletion in Crbn and found cereblon to be an important antagonist of T-cell activation. In mice lacking CRBN, CD4(+) T cells show increased activation and IL-2 production on T-cell receptor stimulation, ultimately resulting in increased potassium flux and calcium-mediated signaling. CRBN restricts T-cell activation via epigenetic modification of Kcna3, which encodes the Kv1.3 potassium channel required for robust calcium influx in T cells. CRBN binds directly to conserved DNA elements adjacent to Kcna3 via a previously uncharacterized DNA-binding motif. Consequently, in the absence of CRBN, the expression of Kv1.3 is derepressed, resulting in increased Kv1.3 expression, potassium flux, and CD4(+) T-cell hyperactivation. In addition, experimental autoimmune encephalomyelitis in T-cell-specific Crbn-deficient mice was exacerbated by increased T-cell activation via Kv1.3. Thus, CRBN limits CD4(+) T-cell activation via epigenetic regulation of Kv1.3 expression.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Epigenesis, Genetic , Kv1.3 Potassium Channel/genetics , Lymphocyte Activation/genetics , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , CD4-Positive T-Lymphocytes/cytology , Calcium/metabolism , Cells, Cultured , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling/methods , Kv1.3 Potassium Channel/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Potassium/metabolism
5.
Biochem Biophys Res Commun ; 503(4): 2892-2898, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30115382

ABSTRACT

Hepatitis B virus (HBV) infection can cause chronic liver diseases, cirrhosis, and hepatocellular carcinoma (HCC). Heat shock proteins (Hsps) are important factors in the formation of the HBV capsid and in genome replication during the viral life cycle. Hsp90 is known to promote capsid assembly. However, the functional roles of Hsp70 in HBV capsid assembly with Hsp90 have not been studied so far. Using microscale thermophoresis analyses and inĀ vitro nucleocapsid formation assays, we found that Hsp70 bound to a HBV core protein dimer and facilitated HBV capsid assembly. Inhibition of Hsp70 by methylene blue (MB) led to a decrease in capsid assembly. Moreover, Hsp70 inhibition reduced intracellular capsid formation and HBV virus particle number in HepG2.2.15Ć¢Ā€ĀÆcells. Furthermore, we examined synergism between Hsp70 and Hsp90 on HBV capsid formation inĀ vitro. Our results clarify the role of Hsp70 in HBV capsid formation via an interaction with core dimers and in synergistically promoting capsid assembly with Hsp90.


Subject(s)
Capsid/metabolism , HSP70 Heat-Shock Proteins/physiology , HSP90 Heat-Shock Proteins/physiology , Hepatitis B virus/ultrastructure , Capsid Proteins/metabolism , Genome, Viral , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Hep G2 Cells , Hepatitis B virus/physiology , Humans , Viral Proteins/metabolism , Virus Assembly , Virus Replication
6.
Article in English | MEDLINE | ID: mdl-29353039

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.

7.
Biochem Biophys Res Commun ; 457(3): 328-33, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25576869

ABSTRACT

Hepatitis B virus (HBV) infection induces reactive oxygen species (ROS) production and has been associated with the development of hepatocellular carcinoma (HCC). ROS are also an important factor in HCC because the accumulated ROS leads to abnormal cell proliferation and chromosome mutation. In oxidative stress, heat shock protein 90 (Hsp90) and glutathione (GSH) function as part of the defense mechanism. Hsp90 prevents cellular component from oxidative stress, and GSH acts as antioxidants scavenging ROS in the cell. However, it is not known whether molecules regulated by oxidative stress are involved in HBV capsid assembly. Based on the previous study that Hsp90 facilitates HBV capsid assembly, which is an important step for the packing of viral particles, here, we show that ROS enrich Hsp90-driven HBV capsid formation. In cell-free system, HBV capsid assembly was facilitated by ROS with Hsp90, whereas it was decreased without Hsp90. In addition, GSH inhibited the function of Hsp90 to decrease HBV capsid assembly. Consistent with the result of cell-free system, ROS and buthionine sulfoximine (BS), an inhibitor of GSH synthesis, increased HBV capsid formation in HepG2.2.15 cells. Thus, our study uncovers the interplay between ROS and Hsp90 during HBV capsid assembly.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , Hepatitis B virus/physiology , Reactive Oxygen Species/metabolism , Virus Assembly/physiology , Buthionine Sulfoximine/pharmacology , Capsid/physiology , Cell-Free System , DNA, Viral/metabolism , Glutathione/metabolism , HSP90 Heat-Shock Proteins/chemistry , Hep G2 Cells , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Host-Pathogen Interactions , Humans , Oxidative Stress , Protein Conformation , Virus Assembly/drug effects
8.
Biochem Biophys Res Commun ; 447(2): 358-63, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24732358

ABSTRACT

Invasion, the representative feature of malignant tumors, leads to an increase in mortality. The malignant liver tumor - hepatocellular carcinoma (HCC) - has an enhanced invasive capacity that results in increased patient mortality. Moreover, this enhanced invasive capacity is due to the up-regulation of invasion promoters such as zinc finger protein SNAI1 (Snail) and matrix metalloproteinases (MMPs), and the down-regulation of invasion suppressor molecules such as E-cadherin. Telomerase reverse transcriptase (TERT), which encodes the catalytic subunit of telomerase, is highly expressed in a variety of invasive cancers, including HCC. Telomerase activation induces telomere elongation, thereby leading to cell immortalization during malignant tumor progression. However, the relationship between telomere length and invasion is yet to be experimentally corroborated. In this paper, we revealed that invasive HCC cells passing through the Matrigel display significantly longer telomeres than non-invasive HCC cells. Moreover, we established a method that can distinguish and sort cells containing long telomeres and short telomeres. Using this system, we observed that the HCC cells containing long telomeres had a high-level expression of invasion-promoting genes and a low-level expression of invasion-suppressing E-cadherin. Furthermore, HCC cells containing long telomeres exhibited a higher invasive capacity than HCC cells containing short telomeres. Taken together, our findings suggest that long telomeres are positively associated with the invasive capacity of HCC cells and may be a potent target for malignant liver cancer treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Telomere Homeostasis , Telomere/physiology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Neoplasm Invasiveness
9.
Biochem Biophys Res Commun ; 433(1): 6-10, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23454378

ABSTRACT

Notch1 intracellular domain (NICD) is the transcription factor which controls cell fate and differentiation in embryonic and tumor cells. Snail has a critical role which increases invasion and metastasis of cancer cell as a transcription factor and epigenetic regulator. Recently, we discovered NICD induced Snail degradation by direct binding interaction with Snail. In this experiment, we found that Snail suppressed transcriptional activity of the protein complex formed with NICD and RBPJk in nucleus. Moreover, Snail decreased transcription of NICD target genes via competing with MAML1, co-activator, in NICD complex. In conclusion, Snail inhibited NICD-mediated transcriptional activation of target genes by physical interaction with NICD.


Subject(s)
DNA-Binding Proteins/metabolism , Receptor, Notch1/antagonists & inhibitors , Transcription Factors/metabolism , Binding, Competitive , Cell Line , DNA-Binding Proteins/genetics , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Structure, Tertiary , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation
10.
J Enzyme Inhib Med Chem ; 28(5): 916-25, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22803663

ABSTRACT

Virus capsid structure is essential in virion maturation and durability, so disrupting capsid assembly could be an effective way to reduce virion count and cure viral diseases. However, currently there is no known antiviral which affects capsid inhibition, and only a small number of assembly inhibitors were experimentally successful. In this present study, we aimed to find hepatitis B virus (HBV) capsid assembly inhibitor which binds to the HBV core protein and changes protein conformation. Several candidate molecules were found to bind to certain structure in core protein with high specificity. Furthermore, these molecules significantly changed the protein conformation and reduced assembly affinity of core protein, leading to decrease of the number of assembled capsid or virion, both in vitro and in vivo. In addition, prediction also suggests that improvements in inhibition efficiency could be possible by changing functional groups and ring structures.


Subject(s)
Capsid/drug effects , Capsid/metabolism , Drug Design , Hepatitis B virus/chemistry , Hepatitis B virus/drug effects , Sulfanilamides/chemistry , Sulfanilamides/pharmacology , Capsid/chemistry , Hepatitis B virus/metabolism , Models, Molecular , Molecular Structure , Sulfanilamide , Sulfanilamides/chemical synthesis , Virus Assembly/drug effects
12.
Hepatology ; 53(4): 1352-62, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21480340

ABSTRACT

UNLABELLED: The tumor suppressor p53 is a key prognostic factor in hepatocellular carcinoma (HCC), yet only 35% of grade III tumors exhibit mutation of p53. Several other pathways have been implicated in HCC and, among these, the role of the Notch1/Snail pathway remains unclear. Therefore, we investigated the expression of p53, Notch1, and Snail proteins in HCC with regard to both clinical grade and p53 mutational status. Immunoblotting for p53 revealed that, whereas in many tumors increased p53 was a result of p53 mutation, wildtype p53 (p53WT) expression was also frequently elevated in HCCs. Coordinated evaluation of p53, Notch1, and Snail expression suggests that grade III HCC can be subdivided based on the expression of these three proteins. We found that Notch1 expression in HCC tissues and cell lines is differentially affected by p53WT and mutant p53 (p53Mut). Notch1 expression was correlated with p53 expression in cells expressing p53WT, but was not elevated in p53Mut-expressing cells. Virally mediated expression or silencing of p53WT or p53Mut confirmed that p53WT overexpression causes Notch1 up-regulation in HCC. Surprisingly, the consequence of Notch1 overexpression for the proliferative and invasive capacity of HCC cells depends on both the p53 mutational status and activation of the Snail pathway. CONCLUSION: In the presence of p53WT, Snail/Notch1 activation increased the invasiveness of HCC cells. In contrast, in the absence of p53WT, Notch1 decreased the invasiveness of HCC. Taken together, these findings shed new light on the complex role of the Notch1/Snail axis in HCC and provide a framework for further classifying HCC based on the expression and mutational status of p53 and the expression of Notch1 and Snail.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Receptor, Notch1/physiology , Tumor Suppressor Protein p53/biosynthesis , Adult , Animals , Carcinoma, Hepatocellular/classification , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/genetics
13.
BMC Biol ; 9: 83, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-22128911

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a common, highly invasive malignant tumor associated with a high mortality rate. We previously reported that the aberrant expression of Snail via activation of reactive oxygen species contributes to the invasive property of HCC, in part by downregulation of E-cadherin through both transcriptional repression and epigenetic modification of the E-cadherin promoter. Having demonstrated the ability of Snail to bind and recruit histone deacetylase 1 and DNA methyltransferase 1 in this context, we set out to look for other interactions that could affect its ability to promote oncogenic transformation and cancer cell invasion. RESULTS: Using cells that stably expressed Snail, we characterized Snail protein interactors by tandem affinity purification and mass spectrometry. Immunoprecipitation and subcellular colocalization studies were performed to confirm our identification of the Notch1 intracellular domain (NICD) as a novel Snail-binding partner. NICD interaction with Snail was found to induce ubiquitination and MDM2-dependent degradation of Snail. Interestingly, NICD inhibited Snail-dependent invasive properties in both HCC cells and mouse embryonic fibroblasts. CONCLUSIONS: Our study demonstrates that NICD can oppose Snail-dependent HCC cell invasion by binding and inducing proteolytic degradation of Snail. Although Notch signaling and Snail are both widely considered tumor-promoting factors, our findings indicate that the individual oncogenic contribution of Notch1 and Snail in malignant systems should be interpreted carefully, particularly when they are conjointly expressed.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Receptor, Notch1/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Liver Neoplasms/pathology , Mice , Neoplasm Invasiveness , Protein Binding , Protein Structure, Tertiary , Receptor, Notch1/analysis , Snail Family Transcription Factors , Transcription Factors/chemistry , Ubiquitination , Zinc Fingers
14.
J Hepatol ; 54(5): 939-47, 2011 May.
Article in English | MEDLINE | ID: mdl-21145824

ABSTRACT

BACKGROUND & AIMS: The concept of multistep hepatocarcinogenesis has been well-established, and an accumulation of methylating events has recently been demonstrated; however, the methylation status of low-grade dysplastic nodules (LGDN), high-grade dysplastic nodules (HGDN), and the recently introduced early hepatocellular carcinoma (eHCC) in hepatitis B virus (HBV)-related hepatocarcinogenesis has not yet been studied. METHODS: One hundred thirty-three DNA samples (45 cirrhotic nodules, 29 LGDNs, 13 HGDNs, 14 eHCCs, and 32 progressed HCCs (pHCCs)) from HBV-infected resected livers were subjected to MethyLight analysis for nine CpG island loci (APC, RASSF1A, SOCS1, P16, COX2, SPRY2, PTEN, GNMT, and ERK), and COX2, RASSF1A, and SOCS1 protein expression status was analyzed by immunohistochemistry. The methylation status of each sample was correlated with the clinicopathological features. RESULTS: APC, RASSF1A, and SOCS1 were methylated in 20 (44.4%), 25 (55.6%), and 13 (28.9%) of 45 cirrhosis samples, and APC (p=0.0008) and SOCS1 (p=0.0187) methylation were more frequent in dysplastic nodules and HCCs. APC (p=0.001) and RASSF1A (p=0.019) methylation levels were significantly increased from cirrhosis to LGDN. SOCS1 methylation gradually increased along multistep hepatocarcinogenesis, peaked at eHCC and decreased significantly in pHCCs (p=0.039). By contrast, p16 and COX2 was only methylated in dysplastic nodules and HCCs, with a stepwise increase up to pHCCs. As a whole, the frequency of methylation was highest in eHCCs. A stepwise decrease in COX2, RASSF1A, and SOCS1 protein expression was demonstrated. CONCLUSIONS: A general stepwise increase in methylating events is seen during HBV-related multistep hepatocarcinogenesis, and epigenetic changes may occur predominantly in the earlier stages of HCC development.


Subject(s)
Carcinoma, Hepatocellular , CpG Islands/genetics , DNA Methylation/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic , Liver Neoplasms , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Adult , Aged , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/virology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Male , Middle Aged , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Korean J Hepatol ; 17(2): 120-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21757983

ABSTRACT

BACKGROUND/AIMS: Combination treatment consisting of hepatic arterial infusion chemotherapy with epirubicin and cisplatin (HAIC-EC) and systemic infusion of low-dose 5-fluorouracil (5-FU) are sometimes effective against advanced hepatocellular carcinoma (HCC). However, there is no effective treatment for advanced HCCs with arterioportal shunts (APS) or arteriovenous shunts (AVS). METHODS: We investigated a response and adverse events of a new combination protocol of repeated HAIC-EC and percutaneous intratumoral injection chemotherapy with a mixture of recombinant interferon-gamma (IFN-ƎĀ³) and 5-FU (PIC-IF) in patients with far-advanced HCCs with large APSs or AVSs. RESULTS: There was a complete response (CR) for the large vascular shunts in all three patients and for all tumor burdens in two patients. Significant side effects were flu-like symptoms (grade 2) and bone marrow suppression (grade 2 or 3) after each cycle, but these were well-tolerated. CONCLUSIONS: These results suggest that the combination of HAIC-EC and PIC-IF is a new and promising approach for advanced HCC accompanied by a large APS or AVS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Aged , Angiography , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/pathology , Cisplatin/administration & dosage , Epirubicin/administration & dosage , Fluorouracil/administration & dosage , Hepatic Artery , Humans , Infusions, Intra-Arterial , Injections, Intramuscular , Interferon-gamma/administration & dosage , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Male , Recombinant Proteins , Tomography, X-Ray Computed , Tumor Burden
16.
Nat Struct Mol Biol ; 27(6): 581-588, 2020 06.
Article in English | MEDLINE | ID: mdl-32451488

ABSTRACT

TENT4 enzymes generate 'mixed tails' of diverse nucleotides at 3' ends of RNAs via nontemplated nucleotide addition to protect messenger RNAs from deadenylation. Here we discover extensive mixed tailing in transcripts of hepatitis B virus (HBV) and human cytomegalovirus (HCMV), generated via a similar mechanism exploiting the TENT4-ZCCHC14 complex. TAIL-seq on HBV and HCMV RNAs revealed that TENT4A and TENT4B are responsible for mixed tailing and protection of viral poly(A) tails. We find that the HBV post-transcriptional regulatory element (PRE), specifically the CNGGN-type pentaloop, is critical for TENT4-dependent regulation. HCMV uses a similar pentaloop, an interesting example of convergent evolution. This pentaloop is recognized by the sterile alpha motif domain-containing ZCCHC14 protein, which in turn recruits TENT4. Overall, our study reveals the mechanism of action of PRE, which has been widely used to enhance gene expression, and identifies the TENT4-ZCCHC14 complex as a potential target for antiviral therapeutics.


Subject(s)
Cytomegalovirus/genetics , Hepatitis B virus/genetics , Host-Pathogen Interactions/physiology , RNA, Viral/metabolism , Cell Line , Cytomegalovirus/pathogenicity , Hepatitis B virus/pathogenicity , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phylogeny , RNA Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/metabolism , RNA, Viral/chemistry
17.
J Hepatol ; 51(5): 853-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726098

ABSTRACT

BACKGROUND/AIMS: The nonstructural 5A (NS5A) protein of hepatitis C virus (HCV) has been implicated in HCV-induced liver pathogenesis. Wnt/beta-catenin signaling has also been involved in tumorigenesis. To elucidate the molecular mechanism of HCV pathogenesis, we examined the potential effects of HCV NS5A protein on Wnt/beta-catenin signal transduction cascades. METHODS: The effects of NS5A protein on beta-catenin signaling cascades in hepatic cells were investigated by luciferase reporter gene assay, confocal microscopy, immunoprecipitation assay, and immunoblot analysis. RESULTS: beta-Catenin-mediated transcriptional activity is elevated by NS5A protein, in the context of HCV replication, and by infection of cell culture-produced HCV. NS5A protein directly interacts with endogenous beta-catenin and colocalizes with beta-catenin in the cytoplasm. NS5A protein inactivates glycogen synthase kinase 3beta and increases subsequent accumulation of beta-catenin in HepG2 cells. beta-Catenin was also accumulated in HCV patients' liver tissues. In addition, the accumulation of beta-catenin in HCV replicon cells requires both activation of phosphatidylinositol 3-kinase and inactivation of GSK3beta. CONCLUSIONS: NS5A activates beta-catenin signaling cascades through increasing the stability of beta-catenin. This modulation is accomplished by the protein interplay between viral and cellular signaling transducer. These data suggest that NS5A protein may directly be involved in Wnt/beta-catenin-mediated liver pathogenesis.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Liver Neoplasms/etiology , Viral Nonstructural Proteins/metabolism , beta Catenin/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , COS Cells , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytosol/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepacivirus/genetics , Humans , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replicon , Sequence Deletion , Signal Transduction , Transcription Factor 4 , Transcription Factors/metabolism , Transfection , Viral Nonstructural Proteins/genetics , Virus Replication , Wnt Proteins/metabolism , beta Catenin/chemistry , beta Catenin/genetics
18.
Gastroenterology ; 135(6): 2128-40, 2140.e1-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801366

ABSTRACT

BACKGROUND & AIMS: In addition to genetic alterations, epigenetic changes underlie tumor progression and metastasis. Promoter methylation can silence tumor suppressor genes, and reactive oxygen species (ROS) promote DNA damage, although the relationship between ROS and epigenetic changes in cancer cells is not clear. We sought to determine whether ROS promote hypermethylation of the promoter region of E-cadherin, a regulator of the epithelial-to-mesenchymal transition, in hepatocellular carcinoma (HCC) cells. METHODS: HCC cells were exposed to H(2)O(2) or stably transfected to express Snail, a transcription factor that down-regulates E-cadherin expression. E-cadherin and Snail expression levels were examined by real-time reverse-transcriptase polymerase chain reaction and immunoblot analyses. The methylation status of E-cadherin was examined by methyl-specific polymerase chain reaction, bisulfite sequencing, and chromatin immunoprecipitation. The interactions between Snail, histone deacetylase 1, and DNA methyltransferase 1 were assessed by immunoprecipitation/immunoblot and immunofluorescence analyses. ROS-induced stress, E-cadherin expression, Snail expression, and E-cadherin promoter methylation were confirmed in HCC tissues by immunoblot, immunohistochemistry, and methyl-specific polymerase chain reaction analyses. RESULTS: We demonstrated that ROS induce hypermethylation of the E-cadherin promoter by increasing Snail expression. Snail induced DNA methylation of the E-cadherin promoter by recruiting histone deacetylase 1 and DNA methyltransferase 1. In human HCC tissues, we observed a correlation among ROS induction, E-cadherin down-regulation, Snail up-regulation, and E-cadherin promoter methylation. CONCLUSIONS: These findings provide novel mechanistic insights into epigenetic modulations induced by ROS in the process of carcinogenesis. They are potentially relevant to understanding the activity of ROS in silencing various tumor suppressor genes and in subsequent tumor progression and metastasis.


Subject(s)
Cadherins/genetics , Carcinoma, Hepatocellular/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Promoter Regions, Genetic/drug effects , Reactive Oxygen Species/pharmacology , Adult , Aged , Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Chromatin Immunoprecipitation , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Methylation/drug effects , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured
19.
Mol Cell Biol ; 26(8): 3048-59, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581780

ABSTRACT

Hepatitis C virus (HCV) NS5B protein is a membrane-associated phosphoprotein that possesses an RNA-dependent RNA polymerase activity. We recently reported that NS5A protein interacts with TRAF2 and modulates tumor necrosis factor alpha (TNF-alpha)-induced NF-kappaB and Jun N-terminal protein kinase (JNK). Since NS5A and NS5B are the essential components of the HCV replication complex, we examined whether NS5B could modulate TNF-alpha-induced NF-kappaB and JNK activation. In this study, we have demonstrated that TNF-alpha-induced NF-kappaB activation is inhibited by NS5B protein in HEK293 and hepatic cells. Furthermore, NS5B protein inhibited both TRAF2- and IKK-induced NF-kappaB activation. Using coimmunoprecipitation assays, we show that NS5B interacts with IKKalpha. Most importantly, NS5B protein in HCV subgenomic replicon cells interacted with endogenous IKKalpha, and then TNF-alpha-mediated IKKalpha kinase activation was significantly decreased by NS5B. Using in vitro kinase assay, we have further found that NS5B protein synergistically activated TNF-alpha-mediated JNK activity in HEK293 and hepatic cells. These data suggest that NS5B protein modulates TNF-alpha signaling pathways and may contribute to HCV pathogenesis.


Subject(s)
Hepacivirus/metabolism , I-kappa B Kinase/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/physiology , Animals , COS Cells , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation/drug effects , Genes, Reporter , Hepacivirus/genetics , Humans , I-kappa B Kinase/analysis , JNK Mitogen-Activated Protein Kinases/analysis , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/pathology , Luciferases/metabolism , MAP Kinase Kinase 4/metabolism , Precipitin Tests , Protein Binding , Tumor Necrosis Factor-alpha/pharmacology , Viral Nonstructural Proteins/genetics
20.
Biochem J ; 416(1): 47-54, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18605987

ABSTRACT

The HBV (hepatitis B virus) core is a phosphoprotein whose assembly, replication, encapsidation and localization are regulated by phosphorylation. It is known that PKC (protein kinase C) regulates pgRNA (pregenomic RNA) encapsidation by phosphorylation of the C-terminus of core, which is a component packaged into capsid. Neither the N-terminal residue phosphorylated by PKC nor the role of the C-terminal phosphorylation have been cleary defined. In the present study we found that HBV Cp149 (core protein C-terminally truncated at amino acid 149) expressed in Escherichia coli was phosphorylated by PKC at Ser(106). PKC-mediated phosphorylation increased core affinity, as well as assembly and capsid stability. In vitro phosphorylation with core mutants (S26A, T70A, S106A and T114A) revealed that the Ser(106) mutation inhibited phosphorylation of core by PKC. CD analysis also revealed that PKC-mediated phosphorylation stabilized the secondary structure of capsid. When either pCMV/FLAG-Cp149[WT (wild-type)] or pCMV/FLAG-S106A Cp149 was transfected into Huh7 human hepatoma cells, mutant capsid level was decreased by 2.06-fold with the S106A mutant when compared with WT, although the same level of total protein was expressed in both cases. In addition, when pUC1.2x and pUC1.2x/S106A were transfected, mutant virus titre was decreased 2.31-fold compared with WT virus titre. In conclusion, PKC-mediated phosphorylation increased capsid assembly, stability and structural stability.


Subject(s)
Capsid/physiology , Hepatitis B Core Antigens/metabolism , Hepatitis B virus/physiology , Protein Kinase C/metabolism , Carcinoma, Hepatocellular/metabolism , Circular Dichroism , Hepatitis B Core Antigens/chemistry , Hepatitis B Core Antigens/genetics , Hepatitis B virus/genetics , Humans , Phosphorylation , Protein Structure, Secondary , Surface Plasmon Resonance , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL