Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Br J Pharmacol ; 181(19): 3717-3742, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38852992

ABSTRACT

BACKGROUND AND PURPOSE: Only limited therapeutic agents have been developed for non-alcoholic steatohepatitis (NASH). Glabridin, a promising anti-obesity candidate, has only limited druggability due to its low in vivo chemical stability and bioavailability. Therefore, we developed vutiglabridin (VUTI), which is based on a glabridin backbone, and investigated its mechanism of action in treating NASH in animal models. EXPERIMENTAL APPROACH: Anti-NASH effects of VUTI were determined in in vitro fatty liver models, spheroids of primary human hepatocytes and L02 normal liver cell lines. To identify VUTI possible cellular target/s, biotin-labelled VUTI was synthesized and underwent chemical proteomic analysis. Further, the evaluation of VUTI therapeutic efficacy was carried out using an amylin-NASH and high-fat (HF) diet-induced obese (DIO) mouse models. This was carried out using transcriptomic, lipidomic and proteomic analyses of the livers from the amylin-NASH mouse model. KEY RESULTS: VUTI treatment markedly reduces hepatic steatosis, fibrosis and inflammation by promoting lipid catabolism, activating autophagy and improving mitochondrial dysfunction, all of which are hallmarks of effective NASH treatment. The cellular target of VUTI was identified as paraoxonase 2 (PON2), a newly proposed protein target for the treatment of NASH, VUTI enhanced PON2 activity. The results using PON2 knockdown cells demonstrated that PON2 is important for VUTI- activation of autophagy, promoting mitochondrial function, decreasing oxidative stress and alleviating lipid accumulation under lipotoxic condition. CONCLUSION AND IMPLICATIONS: Our data demonstrated that VUTI is a promising therapeutic for NASH. Targeting PON2 may be important for improving liver function in various immune-metabolic diseases including NASH.


Subject(s)
Aryldialkylphosphatase , Autophagy , Non-alcoholic Fatty Liver Disease , Animals , Humans , Male , Mice , Aryldialkylphosphatase/metabolism , Autophagy/drug effects , Diet, High-Fat , Disease Models, Animal , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism
2.
Exp Mol Med ; 56(1): 192-209, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38200153

ABSTRACT

Tumor necrosis factor superfamily (TNFSF) resistance contributes to the development and progression of tumors and resistance to various cancer therapies. Tumor-intrinsic alterations involved in the adaptation to the TNFSF response remain largely unknown. Here, we demonstrate that protein kinase C substrate 80K-H (PRKCSH) abundance in lung cancers boosts oncogenic IGF1R activation, leading to TNFSF resistance. PRKCSH abundance is correlated with IGF1R upregulation in lung cancer tissues. Specifically, PRKCSH interacts with IGF1R and extends its half-life. The PRKCSH-IGF1R axis in tumor cells impairs caspase-8 activation, increases Mcl-1 expression, and inhibits caspase-9, leading to an imbalance between cell death and survival. PRKCSH deficiency augmented the antitumor effects of natural killer (NK) cells, representative TNFSF effector cells, in a tumor xenograft IL-2Rg-deficient NOD/SCID (NIG) mouse model. Our data suggest that PRKCSH plays a critical role in TNFSF resistance and may be a potential target to improve the efficacy of NK cell-based cancer therapy.


Subject(s)
Lung Neoplasms , Animals , Mice , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Half-Life , Cell Line, Tumor , Mice, Inbred NOD , Mice, SCID , Tumor Necrosis Factors/metabolism , Calcium-Binding Proteins , Glucosidases/metabolism , Receptor, IGF Type 1/metabolism
3.
Sci Rep ; 12(1): 21483, 2022 12 12.
Article in English | MEDLINE | ID: mdl-36509805

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is an increasingly prevalent immuno-metabolic disease that can progress to hepatic cirrhosis and cancer. NAFLD pathogenesis is extremely complex and is characterized by oxidative stress, impaired mitochondrial function and lipid metabolism, and cellular inflammation. Thus, in-depth research on its underlying mechanisms and subsequent investigation into a potential drug target that has overarching effects on these features will help in the discovery of effective treatments for NAFLD. Our study examines the role of endogenous paraoxonase-2 (PON2), a membrane protein with reported antioxidant activity, in an in vitro cell model of NAFLD. We found that the hepatic loss of PON2 activity aggravated steatosis and oxidative stress under lipotoxic conditions, and our transcriptome analysis revealed that the loss of PON2 disrupts the activation of numerous functional pathways closely related to NAFLD pathogenesis, including mitochondrial respiratory capacity, lipid metabolism, and hepatic fibrosis and inflammation. We found that PON2 promoted the activation of the autophagy pathway, specifically the mitophagy cargo sequestration, which could potentially aid PON2 in alleviating oxidative stress, mitochondrial dysfunction, lipid accumulation, and inflammation. These results provide a mechanistic foundation for the prospect of PON2 as a drug target, leading to the development of novel therapeutics for NAFLD.


Subject(s)
Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/pathology , Mitochondria/metabolism , Autophagy , Liver/metabolism , Oxidative Stress , Inflammation/pathology
4.
Nat Commun ; 10(1): 3776, 2019 Aug 16.
Article in English | MEDLINE | ID: mdl-31420557

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nat Commun ; 10(1): 3185, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320625

ABSTRACT

Unfolded protein response (UPR) is an adaptive mechanism that aims at restoring ER homeostasis under severe environmental stress. Malignant cells are resistant to environmental stress, which is largely due to an activated UPR. However, the molecular mechanisms by which different UPR branches are selectively controlled in tumor cells are not clearly understood. Here, we provide evidence that PRKCSH, previously known as glucosidase II beta subunit, functions as a regulator for selective activation of the IRE1α branch of UPR. PRKCSH boosts ER stress-mediated autophosphorylation and oligomerization of IRE1α through mutual interaction. PRKCSH contributes to the induction of tumor-promoting factors and to tumor resistance to ER stress. Increased levels of PRKCSH in various tumor tissues are positively correlated with the expression of XBP1-target genes. Taken together, our data provide a molecular rationale for selective activation of the IRE1α branch in tumors and adaptation of tumor cells to severe environmental stress.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Transformation, Neoplastic/pathology , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/metabolism , Glucosidases/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , Animals , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Survival/genetics , Endoribonucleases/genetics , Glucosidases/genetics , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics
6.
EMBO J ; 38(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30979777

ABSTRACT

The multifunctional influenza virus protein PB1-F2 plays several roles in deregulation of host innate immune responses and is a known immunopathology enhancer of the 1918 influenza pandemic. Here, we show that the 1918 PB1-F2 protein not only interferes with the mitochondria-dependent pathway of type I interferon (IFN) signaling, but also acquired a novel IFN antagonist function by targeting the DEAD-box helicase DDX3, a key downstream mediator in antiviral interferon signaling, toward proteasome-dependent degradation. Interactome analysis revealed that 1918 PB1-F2, but not PR8 PB1-F2, binds to DDX3 and causes its co-degradation. Consistent with intrinsic protein instability as basis for this gain-of-function, internal structural disorder is associated with the unique cytotoxic sequences of the 1918 PB1-F2 protein. Infusing mice with recombinant DDX3 protein completely rescued them from lethal infection with the 1918 PB1-F2-producing virus. Alongside NS1 protein, 1918 PB1-F2 therefore constitutes a potent IFN antagonist causative for the severe pathogenicity of the 1918 influenza strain. Our identification of molecular determinants of pathogenesis should be useful for the future design of new antiviral strategies against influenza pandemics.


Subject(s)
DEAD-box RNA Helicases/metabolism , Influenza, Human/virology , Interferons/metabolism , Orthomyxoviridae/pathogenicity , Viral Proteins/physiology , A549 Cells , Animals , Dogs , Female , HEK293 Cells , History, 20th Century , Humans , Influenza, Human/epidemiology , Influenza, Human/history , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae/metabolism , Pandemics , Proteolysis , Signal Transduction , U937 Cells , Viral Proteins/metabolism , Virulence/physiology
7.
J Hepatol ; 70(6): 1093-1102, 2019 06.
Article in English | MEDLINE | ID: mdl-30794889

ABSTRACT

BACKGROUND & AIMS: Tenofovir disoproxil fumarate (TDF) is one the most potent nucleot(s)ide analogues for treating chronic hepatitis B virus (HBV) infection. Phenotypic resistance caused by genotypic resistance to TDF has not been reported. This study aimed to characterize HBV mutations that confer tenofovir resistance. METHODS: Two patients with viral breakthrough during treatment with TDF-containing regimens were prospectively enrolled. The gene encoding HBV reverse transcriptase was sequenced. Eleven HBV clones harboring a series of mutations in the reverse transcriptase gene were constructed by site-directed mutagenesis. Drug susceptibility of each clone was determined by Southern blot analysis and real-time PCR. The relative frequency of mutants was evaluated by ultra-deep sequencing and clonal analysis. RESULTS: Five mutations (rtS106C [C], rtH126Y [Y], rtD134E [E], rtM204I/V, and rtL269I [I]) were commonly found in viral isolates from 2 patients. The novel mutations C, Y, and E were associated with drug resistance. In assays for drug susceptibility, the IC50 value for wild-type HBV was 3.8 ±â€¯0.6 µM, whereas the IC50 values for CYE and CYEI mutants were 14.1 ±â€¯1.8 and 58.1 ±â€¯0.9 µM, respectively. The IC90 value for wild-type HBV was 30 ±â€¯0.5 µM, whereas the IC90 values for CYE and CYEI mutants were 185 ±â€¯0.5 and 790 ±â€¯0.2 µM, respectively. Both tenofovir-resistant mutants and wild-type HBV had similar susceptibility to the capsid assembly modulator NVR 3-778 (IC50 <0.4 µM vs. IC50 = 0.4 µM, respectively). CONCLUSIONS: Our study reveals that the quadruple (CYEI) mutation increases the amount of tenofovir required to inhibit HBV by 15.3-fold in IC50 and 26.3-fold in IC90. These results demonstrate that tenofovir-resistant HBV mutants can emerge, although the genetic barrier is high. LAY SUMMARY: Tenofovir is the most potent nucleotide analogue for the treatment of chronic hepatitis B virus infection and there has been no hepatitis B virus mutation that confers >10-fold resistance to tenofovir up to 8 years. Herein, we identified, for the first time, a quadruple mutation that conferred 15.3-fold (IC50) and 26.3-fold (IC90) resistance to tenofovir in 2 patients who experienced viral breakthrough during tenofovir treatment.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis B virus/genetics , Hepatitis B, Chronic/drug therapy , Mutation , RNA-Directed DNA Polymerase/genetics , Reverse Transcriptase Inhibitors/therapeutic use , Tenofovir/therapeutic use , Aged , Cell Line, Tumor , Drug Resistance, Viral/genetics , Humans , Male
8.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29875248

ABSTRACT

Hepatitis B virus (HBV) infection is a leading cause of liver diseases; however, the host factors which facilitate the replication and persistence of HBV are largely unidentified. Cellular FLICE inhibitory protein (c-FLIP) is a typical antiapoptotic protein. In many cases of liver diseases, the expression level of c-FLIP is altered, which affects the fate of hepatocytes. We previously found that c-FLIP and its cleaved form interact with HBV X protein (HBx), which is essential for HBV replication, and regulate diverse cellular signals. In this study, we investigated the role of endogenous c-FLIP in HBV replication and its underlying mechanisms. The knockdown of endogenous c-FLIP revealed that this protein regulates HBV replication through two different mechanisms. (i) c-FLIP interacts with HBx and protects it from ubiquitin-dependent degradation. The N-terminal DED1 domain of c-FLIP is required for HBx stabilization. (ii) c-FLIP regulates the expression or stability of hepatocyte nuclear factors (HNFs), which have critical roles in HBV transcription and maintenance of hepatocytes. c-FLIP regulates the stability of HNFs through physical interactions. We verified our findings in three HBV infection systems: HepG2-NTCP cells, differentiated HepaRG cells, and primary human hepatocytes. In conclusion, our results identify c-FLIP as an essential factor in HBV replication. c-FLIP regulates viral replication through its multiple effects on viral and host proteins that have critical roles in HBV replication.IMPORTANCE Although the chronic hepatitis B virus (HBV) infection still poses a major health concern, the host factors which are required for the replication of HBV are largely uncharacterized. Our studies identify cellular FLICE inhibitory protein (c-FLIP) as an essential factor in HBV replication. We found the dual roles of c-FLIP in regulation of HBV replication: c-FLIP interacts with HBx and enhances its stability and regulates the expression or stability of hepatocyte nuclear factors which are essential for transcription of HBV genome. Our findings may provide a new target for intervention in persistent HBV infection.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Hepatitis B virus/physiology , Host-Pathogen Interactions , Trans-Activators/metabolism , Virus Replication , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Gene Knockdown Techniques , Hepatocytes/virology , Humans , Viral Regulatory and Accessory Proteins
9.
Autophagy ; 12(12): 2451-2466, 2016 12.
Article in English | MEDLINE | ID: mdl-27740879

ABSTRACT

Death receptors of TNFSF10/TRAIL (tumor necrosis factor superfamily member 10) contribute to immune surveillance against virus-infected or transformed cells by promoting apoptosis. Many viruses evade antiviral immunity by modulating TNFSF10 receptor signaling, leading to persistent infection. Here, we report that hepatitis B virus (HBV) X protein (HBx) restricts TNFSF10 receptor signaling via macroautophagy/autophagy-mediated degradation of TNFRSF10B/DR5, a TNFSF10 death receptor, and thus permits survival of virus-infected cells. We demonstrate that the expression of the TNFRSF10B protein is dramatically reduced both in liver tissues of chronic hepatitis B patients and in cell lines transfected with HBV or HBx. HBx-mediated downregulation of TNFRSF10B is caused by the lysosomal, but not proteasomal, degradation pathway. Immunoblotting analysis of LC3B and SQSTM1, and microscopy analysis of tandem-fluorescence-tagged LC3B revealed that HBx promotes complete autophagy. Inhibition of autophagy with a pharmacological inhibitor and LC3B knockdown revealed that HBx-induced autophagy is crucial for TNFRSF10B degradation. Immunoprecipitation and GST affinity isolation assays showed that HBx directly interacts with TNFRSF10B and recruits it to phagophores, the precursors to autophagosomes. We confirmed that autophagy activation is related to the downregulation of the TNFRSF10B protein in liver tissues of chronic hepatitis B patients. Inhibition of autophagy enhanced the susceptibility of HBx-infected hepatocytes to TNFSF10. These results identify the dual function of HBx in TNFRSF10B degradation: HBx plays a role as an autophagy receptor-like molecule, which promotes the association of TNFRSF10B with LC3B; HBx is also an autophagy inducer. Our data suggest a molecular mechanism for HBV evasion from TNFSF10-mediated antiviral immunity, which may contribute to chronic HBV infection.


Subject(s)
Autophagy , Hepatitis B virus/metabolism , Proteolysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Gene Expression Regulation , Hep G2 Cells , Hepatitis, Chronic/genetics , Hepatitis, Chronic/pathology , Humans , Liver/metabolism , Liver/pathology , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
10.
PLoS One ; 10(8): e0136728, 2015.
Article in English | MEDLINE | ID: mdl-26322642

ABSTRACT

The emergence of compensatory mutations in the polymerase gene of drug resistant hepatitis B virus (HBV) is associated with treatment failure. We previously identified a multi-drug resistant HBV mutant, which displayed resistance towards lamivudine (LMV), clevudine (CLV), and entecavir (ETV), along with a strong replication capacity. The aim of this study was to identify the previously unknown compensatory mutations, and to determine the clinical relevance of this mutation during antiviral therapy. In vitro mutagenesis, drug susceptibility assay, and molecular modeling studies were performed. The rtL269I substitution conferred 2- to 7-fold higher replication capacity in the wild-type (WT) or YMDD mutation backbone, regardless of drug treatment. The rtL269I substitution alone did not confer resistance to LMV, ETV, adefovir (ADV), or tenofovir (TDF). However, upon combination with YMDD mutation, the replication capacity under LMV or ETV treatment was enhanced by several folds. Molecular modeling studies suggested that the rtL269I substitution affects template binding, which may eventually lead to the enhanced activity of rtI269-HBV polymerase in both WT virus and YMDD mutant. The clinical relevance of the rtL269I substitution was validated by its emergence in association with YMDD mutation in chronic hepatitis B (CHB) patients with sub-optimal response or treatment failure to LMV or CLV. Our study suggests that substitution at rt269 in HBV polymerase is associated with multi-drug resistance, which may serve as a novel compensatory mutation for replication-defective multi-drug resistant HBV.


Subject(s)
Antiviral Agents/therapeutic use , Drug Resistance, Multiple, Viral/genetics , Gene Products, pol/genetics , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Adenine/analogs & derivatives , Adenine/therapeutic use , Amino Acid Substitution/genetics , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/therapeutic use , Cell Line, Tumor , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Humans , Lamivudine/therapeutic use , Microbial Sensitivity Tests , Models, Molecular , Organophosphonates/therapeutic use , Tenofovir/therapeutic use , Virus Replication/drug effects
11.
Biochim Biophys Acta ; 1842(9): 1648-57, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24769044

ABSTRACT

Hepatocystin/80K-H is known as a causative gene for autosomal dominant polycystic liver disease. However, the role of hepatocystin in hepatitis B virus-related liver disease remains unknown. Here, we investigated the role of hepatocystin on the cytokine-mediated antiviral response against hepatitis B virus infection. We investigated the antiviral effect and mechanism of hepatocystin by ectopic expression and RNAi knockdown in cell culture and mouse livers. Hepatocystin suppressed the replication of hepatitis B virus both in vitro and in vivo. This inhibitory effect was HBx-independent and mediated by the transcriptional regulation of viral genome via the activation of exogenous signal-regulated kinase 1/2 and the reduced expression of hepatocyte nuclear factor 4α, a transcription factor essential for hepatitis B virus replication. The amino-terminal region of hepatocystin was essential for regulation of this antiviral signaling pathway. We also found that hepatocystin acts as a critical component in interferon-mediated mitogen-activated protein kinase signaling pathway, and the interferon-induced antiviral activity against hepatitis B virus is associated with the expression levels of hepatocystin. We demonstrated that hepatocystin plays a critical role in modulating the susceptibility of hepatitis B virus to interferon, suggesting that the modulation of hepatocystin expression is important for cytokine-mediated viral clearance during hepatitis B virus infection.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/prevention & control , Gene Expression Regulation , Glucosidases/metabolism , Hepatitis B/prevention & control , Hepatocyte Nuclear Factor 4/metabolism , Interferon-gamma/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Calcium-Binding Proteins , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Cells, Cultured , Drug Synergism , Glucosidases/genetics , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B virus/pathogenicity , Humans , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Liver Neoplasms/virology , Male , Mice , Mice, Inbred BALB C , Signal Transduction , Virus Replication
12.
Biochim Biophys Acta ; 1832(10): 1569-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23644164

ABSTRACT

Hepatitis B virus (HBV) X protein (HBx) is a key player in HBV replication as well as HBV-induced hepatocellular carcinoma (HCC). However, the pathogenesis of HBV infection and the mechanisms of host-virus interactions are still elusive. In this study, a combination of affinity purification and mass spectrometry was applied to identify the host factors interacting with HBx in hepatoma cells. Thirteen proteins were identified as HBx binding partners. Among them, we first focused on determining the functional significance of the interaction between HBx and hepatocystin. A physical interaction between HBx and hepatocystin was confirmed by co-immunoprecipitation and Western blotting. Immunocytochemistry demonstrated that HBx and hepatocystin colocalized in the hepatoma cells. Domain mapping of both proteins revealed that the HBx C-terminus (amino acids 110-154) was responsible for binding to the mannose 6-phosphate receptor homology domain (amino acids, 419-525) of hepatocystin. Using translation and proteasome inhibitors, we found that hepatocystin overexpression accelerated HBx degradation via a ubiquitin-independent proteasome pathway. We demonstrated that this effect was mediated by an interaction between both proteins using a HBx deletion mutant. Hepatocystin overexpression significantly inhibited HBV DNA replication and expression of HBs antigen concomitant with HBx degradation. Using the hepatocystin mutant constructs that bind HBx, we also confirmed that hepatocystin inhibited HBx-dependent HBV replication. In conclusion, we demonstrated for the first time that hepatocystin functions as a chaperon-like molecule by accelerating HBx degradation, and thereby inhibits HBV replication. Our results suggest that inducing hepatocystin may provide a novel therapeutic approach to control HBV infection.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glucosidases/physiology , Hepatitis B virus/physiology , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/metabolism , Trans-Activators/metabolism , Virus Replication/physiology , Amino Acid Sequence , Calcium-Binding Proteins , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Glucosidases/chemistry , Glucosidases/metabolism , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Mass Spectrometry , Molecular Sequence Data , Protein Binding , Viral Regulatory and Accessory Proteins
13.
PLoS One ; 8(3): e57331, 2013.
Article in English | MEDLINE | ID: mdl-23483900

ABSTRACT

Sustained activation of NF-κB is one of the causative factors for various liver diseases, including liver inflammation and hepatocellular carcinoma (HCC). It has been known that activating the NF-κB signal by hepatitis B virus X protein (HBx) is implicated in the development of HCC. However, despite numerous studies on HBx-induced NF-κB activation, the detailed mechanisms still remain unsolved. Recently, p22-FLIP, a cleavage product of c-FLIPL, has been reported to induce NF-κB activation through interaction with the IκB kinase (IKK) complex in primary immune cells. Since our previous report on the interaction of HBx with c-FLIPL, we explored whether p22-FLIP is involved in the modulation of HBx function. First, we identified the expression of endogenous p22-FLIP in liver cells. NF-κB reporter assay and electrophoretic mobility shift assay (EMSA) revealed that the expression of p22-FLIP synergistically enhances HBx-induced NF-κB activation. Moreover, we found that HBx physically interacts with p22-FLIP and NEMO and potentially forms a ternary complex. Knock-down of c-FLIP leading to the downregulation of p22-FLIP showed that endogenous p22-FLIP is involved in HBx-induced NF-κB activation, and the formation of a ternary complex is necessary to activate NF-κB signaling. In conclusion, we showed a novel mechanism of HBx-induced NF-κB activation in which ternary complex formation is involved among HBx, p22-FLIP and NEMO. Our findings will extend the understanding of HBx-induced NF-κB activation and provide a new target for intervention in HBV-associated liver diseases and in the development of HCC.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Hepatocytes/metabolism , I-kappa B Kinase/metabolism , Liver/metabolism , NF-kappa B/metabolism , Signal Transduction , Trans-Activators/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Hepatitis B virus/physiology , Hepatocytes/pathology , Humans , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Binding , RNA, Small Interfering/metabolism , Viral Regulatory and Accessory Proteins , Virus Replication
14.
Anal Chem ; 83(1): 157-63, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21142121

ABSTRACT

Protein tyrosine nitration (PTN) is a post-translational modification that is related to several acute or chronic diseases. PTN introduces a nitro group in the ortho position of the phenolic hydroxyl group of tyrosine residues. PTN has been shown to be involved in the pathogenesis of inflammatory responses, cancers, and neurodegenerative and age-related disorders. Furthermore, it has been proposed that PTN regulates signal cascades related to nitric oxide (NO·) production and NO-mediated processes. Although nitrated proteins as markers of oxidative stress are confirmed by immunological assays in various affected cells or tissues, it is not known how many different types of proteins in living cells are nitrated. Since protein nitration is a low-abundance post-translational modification, development of an effective enrichment method for nitrated proteins is needed to detect nitrated peptides or proteins from the limited amount of pathophysiological samples. In the present study, we developed an enrichment method using specific chemical tagging. Nitroproteome profiling using chemical tagging and mass spectrometry was validated by model proteins. Furthermore, we successfully identified numerous nitrated proteins from the Huh7 human hepatoma cell line.


Subject(s)
Carbon/chemistry , Halogenation , Mass Spectrometry/methods , Nitro Compounds/chemistry , Peptides/analysis , Peptides/chemistry , Amino Acid Sequence , Animals , Cattle , Cell Extracts , Cell Line, Tumor , Computational Biology , Feasibility Studies , Fluorine/chemistry , Humans , Molecular Sequence Data , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptides/isolation & purification , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Solid Phase Extraction , Trypsin/metabolism , Tyrosine/chemistry
15.
J Virol ; 84(9): 4494-503, 2010 May.
Article in English | MEDLINE | ID: mdl-20164224

ABSTRACT

Clevudine (CLV) is a nucleoside analog with potent antiviral activity against chronic hepatitis B virus (HBV) infection. Viral resistance to CLV in patients receiving CLV therapy has not been reported. The aim of this study was to characterize CLV-resistant HBV in patients with viral breakthrough (BT) during long-term CLV therapy. The gene encoding HBV reverse transcriptase (RT) was analyzed from chronic hepatitis B patients with viral BT during CLV therapy. Sera collected from the patients at baseline and at the time of viral BT were studied. To characterize the mutations of HBV isolated from the patients, we subjected the HBV mutants to in vitro drug susceptibility assays. Several conserved mutations were identified in the RT domain during viral BT, with M204I being the most common. In vitro phenotypic analysis showed that the mutation M204I was predominantly associated with CLV resistance, whereas L229V was a compensatory mutation for the impaired replication of the M204I mutant. A quadruple mutant (L129M, V173L, M204I, and H337N) was identified that conferred greater replicative ability and strong resistance to both CLV and lamivudine. All of the CLV-resistant clones were lamivudine resistant. They were susceptible to adefovir, entecavir, and tenofovir, except for one mutant clone. In conclusion, the mutation M204I in HBV RT plays a major role in CLV resistance and leads to viral BT during long-term CLV treatment. Several conserved mutations may have a compensatory role in replication. Drug susceptibility assays reveal that adefovir and tenofovir are the most effective compounds against CLV-resistant mutants. These data may provide additional therapeutic options for CLV-resistant patients.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Arabinofuranosyluracil/analogs & derivatives , Drug Resistance, Viral , Hepatitis B virus/drug effects , Hepatitis B, Chronic/virology , Adult , Amino Acid Substitution/genetics , Arabinofuranosyluracil/pharmacology , Arabinofuranosyluracil/therapeutic use , DNA Mutational Analysis , Female , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/drug therapy , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Mutation, Missense , RNA-Directed DNA Polymerase/genetics , Sequence Analysis, DNA , Serum/virology , Treatment Failure , Viral Proteins/genetics
16.
Chem Biol Interact ; 182(1): 29-36, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19647729

ABSTRACT

Fibroblast-like synovial cells play a crucial role in the pathophysiology of rheumatoid arthritis (RA), as these cells are involved in inflammation and joint destruction. Apigenin, a dietary plant-flavonoid, is known to have many functions in animal cells including anti-proliferative and anticancer activities, but its role in human rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) has not been reported. In this study, we investigated the roles of apigenin in RA-FLSs. The survival rate decreased, and apoptotic cell death was induced by apigenin treatment in RA-FLSs. Apigenin treatment resulted in activation of the mitogen-activated protein kinase (MAPK) ERK1/2, and pretreatment with an ERK inhibitor PD98059 dramatically reduced apigenin-induced apoptosis. We found that apigenin-mediated production of a large amount of intracellular reactive oxygen species (ROS) caused activation of ERK1/2 and apoptosis; treatment with the antioxidant Tiron strongly inhibited the apigenin-induced generation of ROS, phosphorylation of ERK1/2, and apoptotic cell death. Apigenin-induced apoptotic cell death was mediated through activation of the effectors caspase-3 and caspase-7, and was blocked by pretreatment with Z-VAD-FMK (a pan-caspase inhibitor). These results showed that apigenin-induced ROS and oxidative stress-activated ERK1/2 caused apoptotic cell death in apigenin-treated RA-FLSs.


Subject(s)
Apigenin/pharmacology , Apoptosis/drug effects , Arthritis, Rheumatoid/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Reactive Oxygen Species/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/metabolism , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Flavonoids/pharmacology , Flow Cytometry , Humans , Immunoblotting , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology
17.
Virus Res ; 127(1): 71-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17499376

ABSTRACT

The nucleocapsid (N) protein of severe acute respiratory syndrome-coronavirus (SARS-CoV) is involved in the pathological reaction to SARS and is a key antigen for the development of a sensitive diagnostic assay. However, the antigenic properties of this N protein are largely unknown. To facilitate the studies on the function and antigenicity of the SARS-CoV N protein, 6x histidine-tagged recombinant SARS-CoV N (rSARS-N) with a molecular mass of 46 and 48kDa was successfully produced using the recombinant baculovirus system in insect cells. The rSARS-N expressed in insect cells (BrSARS-N) showed remarkably higher specificity and immunoreactivity than rSARS-N expressed in E. coli (ErSARS-N). Most of all, BrSARS-N proteins were expressed as a highly phosphorylated form with a molecular mass of 48kDa, but ErSARS-N was a nonphosphorylated protein. In further analysis to determine the correlation between the phosphorylation and the antigenicity of SARS-N protein, dephosphorylated SARS-N protein treated with protein phosphatase 1 (PP1) remarkably enhanced the cross-reactivity against SARS negative serum and considerably reduced immunoreactivity with SARS-N mAb. These results suggest that the phosphorylation plays an important role in the immunoreactivity and specificity of SARS-N protein. Therefore, the BrSARS-N protein may be useful for the development of highly sensitive and specific assays to determine SARS infection and for further research of SARS-N pathology.


Subject(s)
Antibodies, Viral/immunology , Antibody Specificity/physiology , Antigens, Viral/immunology , Nucleocapsid Proteins/immunology , Phosphorylation , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , Antibodies, Viral/blood , Antibody Affinity , Coronavirus Nucleocapsid Proteins , Enzyme-Linked Immunosorbent Assay , Insecta/cytology , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics
18.
Virus Res ; 122(1-2): 109-18, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16942813

ABSTRACT

Severe acute respiratory syndrome-coronavirus nucleocapsid (SARS-CoV N) protein has been found to be important to the processes related to viral pathogenesis, such as virus replication, interference of the cell process and modulation of host immune response; detection of the antigen has been used for the early diagnosis of infection. We have used recombinant N protein expressed in insect cells to generate 17 mAbs directed against this protein. We selected five mAbs that could be used in various diagnostic assays, and all of these mAbs recognized linear epitopes. Three IgG(2b) mAbs were recognized within the N-terminus of N protein, whereas the epitope of two IgG(1) mAbs localized within the C-terminus. These mAbs were found to have significant reactivity with both non-phosphorylated and phosphorylated N proteins, which resulted in high reactivity with native N protein in virus-infected cells; however, they did not show cross-reactivity with human coronavirus. Therefore, these results suggested that these mAbs would be useful in the development of various diagnostic kits and in future studies of SARS-CoV pathology.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Nucleocapsid Proteins/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Antibody Affinity , Cell Line , Coronavirus Nucleocapsid Proteins , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/immunology , Humans , Immunoblotting , Mice
19.
J Med Virol ; 73(1): 85-90, 2004 May.
Article in English | MEDLINE | ID: mdl-15042653

ABSTRACT

Serological and virological studies were carried out of a mumps outbreak which occurred in one region, Yoeju County, Southeast of Seoul in Korea from September to December, 1999. Sera from 736 children at 8-13 years of age of patients with mumps and healthy children were tested for mumps-specific antibodies by enzyme immunoassay. The overall IgM positive rate was 7.6% (56/736), compared with 69.8% (514/736) for IgG. Of the 49 children with both IgG and IgM, 32 were also confirmed by both clinical and serological diagnosis. IgM antibodies were detected even in the samples collected up to 3 months after the onset of symptoms. Although 436 children had been vaccinated before the outbreak, 27 (6.2%) were found to be IgM positive, particularly 6 (4.4%) of 136 were positive serologically despite a second-dose vaccinees. Sequence analysis of the small hydrophobic (SH) gene of 4 mumps viruses isolated from 42 saliva specimens revealed that these were related to the genotype H, but distinguishable from European strains. This is the first study on the outbreak due to mumps virus genotype H and provides information to assess the understanding of recent outbreaks of mumps in Korea.


Subject(s)
Disease Outbreaks , Mumps virus/genetics , Mumps/epidemiology , Mumps/virology , Adolescent , Amino Acid Sequence , Antibodies, Viral/blood , Antibody Specificity , Child , Female , Genes, Viral , Genotype , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Korea/epidemiology , Male , Molecular Sequence Data , Mumps/immunology , Mumps virus/immunology , Mumps virus/isolation & purification , Phylogeny , Sequence Homology, Amino Acid , Viral Proteins/genetics
20.
Virus Res ; 97(2): 111-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14602202

ABSTRACT

Sequence analyses of the entire small hydrophobic (SH) and hemagglutinin-neuraminidase (HN) genes of mumps viruses circulated in Korea from 1998 to 2001 showed that these isolates were grouped into two genotypes, H and I. While genotype I was predominant throughout the country during this period, genotype H was found in the restricted region, 1999. The nucleotide and deduced amino acid sequences of Korean isolates showed the type-specific changes including the signature motif at positions 28-30 in the SH gene and the neutralizing epitopes in the HN gene. Particularly, Asian strains including Korean isolates and European strains differed from 2.3 to 3.8% at the nucleotide sequence level in the SH gene although they belonged to the same genotype H. Furthermore, none of Korean isolates were genetically related to the vaccine strains used in Korea. The results provide important information to understand the epidemiology of mumps infection and to facilitate the development of more efficient vaccine program in Korea.


Subject(s)
HN Protein/genetics , Mumps virus/classification , Mumps virus/genetics , Mumps/virology , Viral Proteins/genetics , Amino Acid Motifs , Epitopes/genetics , Genotype , Humans , Korea/epidemiology , Molecular Sequence Data , Mumps/epidemiology , Mumps virus/isolation & purification , Phylogeny , Polymorphism, Genetic , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL