Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Virus Res ; 345: 199387, 2024 Jul.
Article En | MEDLINE | ID: mdl-38719025

Influenza A virus can infect respiratory tracts and may cause severe illness in humans. Proteins encoded by influenza A virus can interact with cellular factors and dysregulate host biological processes to support viral replication and cause pathogenicity. The influenza viral PA protein is not only a subunit of influenza viral polymerase but also a virulence factor involved in pathogenicity during infection. To explore the role of the influenza virus PA protein in regulating host biological processes, we performed immunoprecipitation and LC‒MS/MS to globally identify cellular factors that interact with the PA proteins of the influenza A H1N1, 2009 pandemic H1N1, and H3N2 viruses. The results demonstrated that proteins located in the mitochondrion, proteasome, and nucleus are associated with the PA protein. We further discovered that the PA protein is partly located in mitochondria by immunofluorescence and mitochondrial fractionation and that overexpression of the PA protein reduces mitochondrial respiration. In addition, our results revealed the interaction between PA and the mitochondrial matrix protein PYCR2 and the antiviral role of PYCR2 during influenza A virus replication. Moreover, we found that the PA protein could also trigger autophagy and disrupt mitochondrial homeostasis. Overall, our research revealed the impacts of the influenza A virus PA protein on mitochondrial function and autophagy.


Mitochondria , Viral Proteins , Virus Replication , Humans , Mitochondria/metabolism , Mitochondria/virology , Viral Proteins/metabolism , Viral Proteins/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Influenza A virus/physiology , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza A virus/metabolism , Host-Pathogen Interactions , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H3N2 Subtype/metabolism , Autophagy , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N1 Subtype/pathogenicity , HEK293 Cells , Influenza, Human/virology , Influenza, Human/metabolism , A549 Cells , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Tandem Mass Spectrometry
2.
Pathogens ; 13(3)2024 Mar 20.
Article En | MEDLINE | ID: mdl-38535609

Autophagy plays a fundamental role in maintaining cellular homeostasis by eliminating intracellular components via lysosomes. Successful degradation through autophagy relies on the fusion of autophagosomes to lysosomes, which leads to the formation of autolysosomes containing acidic proteases that degrade the sequestered materials. Viral infections can exploit autophagy in infected cells to balance virus-host cell interactions by degrading the invading virus or promoting viral growth. In recent years, cumulative studies have indicated that viral infections may interfere with the fusion of autophagosomes and lysosomes, thus benefiting viral replication and associated pathogenesis. In this review, I provide an overview of the current understanding of the molecular mechanism by which human viral infections deregulate autophagosome-lysosome fusion and summarize the physiological significance in the virus life cycle and host cell damage.

3.
Cells ; 13(6)2024 Mar 13.
Article En | MEDLINE | ID: mdl-38534345

In eukaryotes, targeting intracellular components for lysosomal degradation by autophagy represents a catabolic process that evolutionarily regulates cellular homeostasis. The successful completion of autophagy initiates the engulfment of cytoplasmic materials within double-membrane autophagosomes and subsequent delivery to autolysosomes for degradation by acidic proteases. The formation of autolysosomes relies on the precise fusion of autophagosomes with lysosomes. In recent decades, numerous studies have provided insights into the molecular regulation of autophagosome-lysosome fusion. In this review, an overview of the molecules that function in the fusion of autophagosomes with lysosomes is provided. Moreover, the molecular mechanism underlying how these functional molecules regulate autophagosome-lysosome fusion is summarized.


Autophagosomes , Autophagy , Animals , Autophagosomes/metabolism , Autophagy/physiology , Macroautophagy , Homeostasis , Lysosomes/metabolism , Mammals
4.
Cells ; 12(6)2023 03 21.
Article En | MEDLINE | ID: mdl-36980296

Autophagy plays a homeostatic role in regulating cellular metabolism by degrading unwanted intracellular materials and acts as a host defense mechanism by eliminating infecting pathogens, such as viruses. Upon viral infection, host cells often activate retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) signaling to induce the transcription of type I interferons, thus establishing the first line of the innate antiviral response. In recent years, numerous studies have shown that virus-mediated autophagy activation may benefit viral replication through different actions on host cellular processes, including the modulation of RLR-mediated innate immunity. Here, an overview of the functional molecules and regulatory mechanism of the RLR antiviral immune response as well as autophagy is presented. Moreover, a summary of the current knowledge on the biological role of autophagy in regulating RLR antiviral signaling is provided. The molecular mechanisms underlying the crosstalk between autophagy and RLR innate immunity are also discussed.


Signal Transduction , Viruses , Immunity, Innate , Antiviral Agents , Autophagy
5.
Lupus ; 31(8): 927-938, 2022 Jul.
Article En | MEDLINE | ID: mdl-35531921

OBJECTIVE: To study the association of α2,6-sialic acid (SIA) content in serum IgG anti-dsDNA with human systemic lupus erythematosus disease activity index (SLEDAI) and the effect of sialylated and desialylated (deSIA) IgG anti-dsDNA on lupus B cells. METHODS: Blood from lupus patients was collected to determine the ratio of SIA in isolated IgG anti-dsDNA over serum IgG anti-dsDNA (SIA/IgG anti-dsDNA) ratios, which were plotted against SLEDAI using a receiver-operating-characteristics curve. Lupus B cells were cultured in vitro with chimeric sialylated IgG anti-dsDNA and its deSIA form. Culture supernatants were assayed for anti-inflammatory IL-10 and SIA/IgG anti-dsDNA ratios, which were compared among different pre-treatment groups using t-tests. RESULTS: The area-under-the-curve (AUC) for anti-dsDNA levels against SLEDAI was 0.791 positively (95% confidence interval [C.I.]: 0.699-0.884) and SIA/IgG anti-dsDNA ratios against SLEDAI yielded an AUC of 0.705 inversely (95% C.I: 0.601-0.809): not significantly different. SIA/IgG anti-dsDNA ratios discriminated significantly between patients without and patients with proteinuria (p = .046). SIA/IgG anti-dsDNA ratios correlated significantly and positively with serum C3c and C4 levels. Pre-treatment with IgG anti-dsDNA and its immune complexes (dsDNA/IgG anti-dsDNA IC) induced higher IL-10 from lupus B cells than medium pre-treatment (most p < .01 from day 2 to day 5 culture). DeSIA IgG anti-dsDNA IC induced lower IL-10 (p < .05) and lower SIA/IgG anti-dsDNA ratios (p < .001) from lupus B cells than medium and dsDNA pre-treatment. CONCLUSION: α2,6-SIA/IgG anti-dsDNA ratios inversely forecasted SLEDAI scores. Possible mechanisms may be due to the different effects of sialylated and deSIA IgG anti-dsDNA on lupus B cells in terms of IL-10 secretion and SIA/IgG anti-dsDNA ratios.


Lupus Erythematosus, Systemic , N-Acetylneuraminic Acid , Antibodies, Antinuclear , DNA , Humans , Immunoglobulin G , Interleukin-10 , Pilot Projects
6.
Cells ; 11(7)2022 03 28.
Article En | MEDLINE | ID: mdl-35406696

The elimination of intracellular components by autophagy maintains metabolic homeostasis and is a quality-control pathway that enables organelle regeneration. Mitophagy is a type of selective autophagy that regulates mitochondrial turnover, and the dysregulation of mitophagy has been implicated in the pathogenesis of liver diseases. However, the detailed molecular mechanism underlying mitophagy regulation in liver cells remains unclear, and the small molecules that may potentially modulate hepatic mitophagy are still unavailable. Here, we report that baicalein, a flavonoid extracted from Scutellaria baicalensis, induces the entire autophagy that proceeds through the autolysosome maturation stage in human hepatoma cells. In addition, baicalein-induced autophagy is demonstrated to target mitochondria for degradation. Further studies show that baicalein triggers the translocation of Parkin and TBK1 to mitochondria to induce mitophagy. Moreover, the phosphorylation of TBK1 at Ser172 and ubiquitin at Ser65 is shown to trigger mitophagy in baicalein-treated cells. Furthermore, two specific autophagy cargo receptors, NDP52 and OPTN, that function in baicalein-activated mitophagy are identified. Taken together, these findings not only delineate the molecular process of Parkin-dependent mitophagy in liver cells, but also reveal baicalein as a novel inducer of hepatic mitophagy.


Cell Cycle Proteins , Flavanones , Membrane Transport Proteins , Mitophagy , Nuclear Proteins , Ubiquitin-Protein Ligases , Cell Cycle Proteins/metabolism , Flavanones/pharmacology , Humans , Membrane Transport Proteins/metabolism , Mitophagy/physiology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
IUBMB Life ; 74(4): 317-338, 2022 04.
Article En | MEDLINE | ID: mdl-34859938

Targeting intracellular components for lysosomal degradation by autophagy not only maintains cellular homeostasis but also counteracts the effects of external stimuli, including invading pathogens. Among various kinds of pathogens, viruses have been extensively shown to induce autophagy to benefit viral growth in infected cells and to modulate host defense responses, such as innate antiviral immunity. Recently, numerous lines of evidence have implied that virus-induced autophagy triggers multilayer mechanisms to regulate the innate antiviral response of host cells, thus promoting a balance in virus-host cell interactions. In this review, the detailed mechanisms underlying autophagy and the innate antiviral immune response are first described. Then, I summarize the current information regarding the diverse functional role(s) of autophagy in the control of antiviral defenses against different types of viral infections. Moreover, the physiological significance of autophagy-regulated antiviral responses on the viral life cycle and the potential autophagy alterations induced by virus-associated antiviral signaling is further discussed.


Virus Diseases , Viruses , Antiviral Agents/pharmacology , Autophagy , Host-Pathogen Interactions , Humans , Immunity, Innate
8.
Aging (Albany NY) ; 13(10): 13474-13495, 2021 05 20.
Article En | MEDLINE | ID: mdl-34091442

The medicinal fungus Ganoderma lucidum is used as a dietary supplement and health tonic, but whether it affects longevity remains unclear. We show here that a water extract of G. lucidum mycelium extends lifespan of the nematode Caenorhabditis elegans. The G. lucidum extract reduces the level of fibrillarin (FIB-1), a nucleolar protein that correlates inversely with longevity in various organisms. Furthermore, G. lucidum treatment increases expression of the autophagosomal protein marker LGG-1, and lifespan extension is abrogated in mutant C. elegans strains that lack atg-18, daf-16, or sir-2.1, indicating that autophagy and stress resistance pathways are required to extend lifespan. In cultured human cells, G. lucidum increases concentrations of the LGG-1 ortholog LC3 and reduces levels of phosphorylated mTOR, a known inhibitor of autophagy. Notably, low molecular weight compounds (<10 kDa) isolated from the G. lucidum water extract prolong lifespan of C. elegans and the same compounds induce autophagy in human cells. These results suggest that G. lucidum can increase longevity by inducing autophagy and stress resistance.


Autophagy , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Longevity/physiology , Reishi/chemistry , Animals , Caenorhabditis elegans Proteins/metabolism , Cell Line, Tumor , Humans , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
Hepatol Int ; 14(6): 973-984, 2020 Dec.
Article En | MEDLINE | ID: mdl-32770306

BACKGROUND: Hepatitis B virus (HBV) X gene (HBx) mutants can develop during the natural course of chronic HBV infection. However, little is known about whether the emergence of HBx mutants during long-term antiviral therapy is an adaptation of HBV to antiviral stress. This study was to identify HBx mutants that emerged in patients experiencing Lamivudine resistance or suboptimal treatment. METHODS: Forty-six Lamivudine-resistant patients and 46 patients with suboptimal treatment responses to Entecavir were enrolled in this study. HBx mutants were identified by sequence analysis and their roles in the HBV replication cycle were characterized. RESULTS: We show that deletion/truncation/insertion mutations were only detected in the Lamivudine resistance group, while synonymous mutations were found in both groups. Follow-up analyses revealed that five patients in the Lamivudine group developed hepatocellular carcinoma, while patients in the Entecavir group did not. These mutants were characterized by a significant decrease in transactivation of the pre-S1 promoter, and varying effects on transactivation of the X promoter. Co-transfection of HBx-mutant plasmid and HBV replication-competent clone into HepG2 cells resulted in increased nuclear-to-cytoplamic HBV core antigen, HBV-DNA ratios, and nuclear covalently closed circular DNA (cccDNA). Antiviral drug sensitivity assays revealed that these mutants exhibited a compensatory effect to counteract antiviral drug suppression, resulting in elevated secretory HBV-DNA levels. CONCLUSIONS: Our study demonstrates that HBx mutants can emerge during Lamivudine or Entecavir therapy. These mutants exhibit altered transactivation of the HBV pre-S1 and X promoters, leading to increased cccDNA levels to compensate for replication suppression.


Hepatitis B virus , Hepatitis B, Chronic , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA, Circular/genetics , DNA, Viral/genetics , Drug Resistance, Viral , Hepatitis B virus/genetics , Hepatitis B, Chronic/drug therapy , Humans , Lamivudine/pharmacology , Lamivudine/therapeutic use , Liver Neoplasms/drug therapy , Mutation , Trans-Activators , Viral Regulatory and Accessory Proteins , Virus Replication
10.
Cells ; 9(4)2020 03 30.
Article En | MEDLINE | ID: mdl-32235615

Autophagy is a catabolic process involving vacuolar sequestration of intracellular components and their targeting to lysosomes for degradation, thus supporting nutrient recycling and energy regeneration. Accumulating evidence indicates that in addition to being a bulk, nonselective degradation mechanism, autophagy may selectively eliminate damaged mitochondria to promote mitochondrial turnover, a process termed "mitophagy". Mitophagy sequesters dysfunctional mitochondria via ubiquitination and cargo receptor recognition and has emerged as an important event in the regulation of liver physiology. Recent studies have shown that mitophagy may participate in the pathogenesis of various liver diseases, such as liver injury, liver steatosis/fatty liver disease, hepatocellular carcinoma, viral hepatitis, and hepatic fibrosis. This review summarizes the current knowledge on the molecular regulations and functions of mitophagy in liver physiology and the roles of mitophagy in the development of liver-related diseases. Furthermore, the therapeutic implications of targeting hepatic mitophagy to design a new strategy to cure liver diseases are discussed.


Liver Diseases/pathology , Mitophagy , Autophagosomes/metabolism , Autophagy/genetics , Humans , Liver/pathology , Liver/physiopathology , Liver Diseases/genetics , Liver Diseases/physiopathology , Translational Research, Biomedical
11.
Trends Endocrinol Metab ; 30(6): 335-346, 2019 06.
Article En | MEDLINE | ID: mdl-31060881

Caloric restriction, intermittent fasting, and exercise activate defensive cellular responses such as autophagy, DNA repair, and the induction of antioxidant enzymes. These processes improve health and longevity by protecting cells and organs against damage, mutations, and reactive oxygen species. Consuming a diet rich in vegetables, fruits, and mushrooms can also improve health and longevity. Phytochemicals such as alkaloids, polyphenols, and terpenoids found in plants and fungi activate the same cellular processes as caloric restriction, fasting, and exercise. Many of the beneficial effects of fruits and vegetables may thus be due to activation of stress resistance pathways by phytochemicals. A better understanding of the mechanisms of action of phytochemicals may provide important insights to delay aging and prevent chronic diseases.


Phytochemicals , Aging/physiology , Caloric Restriction , Dietary Supplements , Hormesis/physiology , Humans , Longevity/physiology
12.
BMC Cancer ; 19(1): 250, 2019 Mar 20.
Article En | MEDLINE | ID: mdl-30894157

BACKGROUND: Although men carry a higher risk of hepatocellular carcinoma (HCC) than women, it is still controversial whether men also have a poorer postoperative prognosis. A retrospective study was conducted to evaluate the postoperative prognostic predictors of HCC focusing on sex differences. METHODS: We enrolled 516 consecutive adult patients with HCC (118 women, 398 men), who received surgical resection between January 2000 and December 2007, and were followed-up for >10 years. Clinical and laboratory data together with postoperative outcomes were reviewed. RESULTS: At baseline, female patients had a higher anti-hepatitis C virus antibody prevalence (P = 0.002); lower hepatitis B virus surface antigen prevalence (P = 0.006); less microvascular invasion (P = 0.019); and lower alpha-fetoprotein (P = 0.023), bilirubin (P = 0.002), and alanine transaminase (P = 0.001) levels. Overall, there were no significant sex differences in terms of intrahepatic recurrence-free survival (RFS), distant metastasis-free survival (MFS), and overall survival (OS). However, subgroup analysis showed that women had favorable RFS (P = 0.019) and MFS (P = 0.034) in patients with alpha-fetoprotein ≤ 35 ng/mL, independent of other clinical variables (adjusted P = 0.008 and 0.043, respectively). Additionally, men had favorable OS in patients with prothrombin time (international normalized ratio [INR]) <1.1 (P = 0.033), independent of other clinical variables (adjusted P = 0.042). CONCLUSIONS: Female sex is independently associated with favorable postoperative RFS and MFS in patients with alpha-fetoprotein ≤35 ng/mL, while male sex is independently associated with favorable OS in patients with prothrombin time INR <1.1.


Carcinoma, Hepatocellular/mortality , Health Status Disparities , Hepatectomy , Liver Neoplasms/mortality , Neoplasm Recurrence, Local/epidemiology , Adult , Aged , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Disease-Free Survival , Female , Follow-Up Studies , Humans , International Normalized Ratio , Liver Neoplasms/blood , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Postoperative Period , Prognosis , Prothrombin Time , Retrospective Studies , Sex Factors , alpha-Fetoproteins/analysis
13.
Int J Mol Sci ; 20(2)2019 Jan 13.
Article En | MEDLINE | ID: mdl-30642133

Autophagy is a catabolic process by which eukaryotic cells eliminate cytosolic materials through vacuole-mediated sequestration and subsequent delivery to lysosomes for degradation, thus maintaining cellular homeostasis and the integrity of organelles. Autophagy has emerged as playing a critical role in the regulation of liver physiology and the balancing of liver metabolism. Conversely, numerous recent studies have indicated that autophagy may disease-dependently participate in the pathogenesis of liver diseases, such as liver hepatitis, steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma. This review summarizes the current knowledge on the functions of autophagy in hepatic metabolism and the contribution of autophagy to the pathophysiology of liver-related diseases. Moreover, the impacts of autophagy modulation on the amelioration of the development and progression of liver diseases are also discussed.


Autophagy , Liver Diseases/pathology , Liver/physiology , Animals , Disease Progression , Humans , Liver/metabolism , Liver Diseases/metabolism , Signal Transduction , Vacuoles/metabolism
14.
Int J Mol Sci ; 19(12)2018 Dec 07.
Article En | MEDLINE | ID: mdl-30544615

Autophagy is an evolutionarily conserved cellular process in which intracellular components are eliminated via lysosomal degradation to supply nutrients for organelle biogenesis and metabolic homeostasis. Flavivirus infections underlie multiple human diseases and thus exert an immense burden on public health worldwide. Mounting evidence indicates that host autophagy is subverted to modulate the life cycles of flaviviruses, such as hepatitis C virus, dengue virus, Japanese encephalitis virus, West Nile virus and Zika virus. The diverse interplay between autophagy and flavivirus infection not only regulates viral growth in host cells but also counteracts host stress responses induced by viral infection. In this review, we summarize the current knowledge on the role of autophagy in the flavivirus life cycle. We also discuss the impacts of virus-induced autophagy on the pathogeneses of flavivirus-associated diseases and the potential use of autophagy as a therapeutic target for curing flavivirus infections and related human diseases.


Autophagy/physiology , Flavivirus/pathogenicity , Animals , Dengue Virus/pathogenicity , Encephalitis Virus, Japanese/pathogenicity , Hepacivirus/pathogenicity , Host-Pathogen Interactions , Humans , Zika Virus/pathogenicity
15.
EMBO Rep ; 19(8)2018 08.
Article En | MEDLINE | ID: mdl-29880709

Toll-like receptors (TLRs) are important sensors that recognize pathogen-associated molecular patterns. Generally, TLR9 is known to recognize bacterial or viral DNA but not viral RNA and initiate an immune response. Herein, we demonstrate that infection with dengue virus (DENV), an RNA virus, activates TLR9 in human dendritic cells (DCs). DENV infection induces release of mitochondrial DNA (mtDNA) into the cytosol and activates TLR9 signaling pathways, leading to production of interferons (IFNs). The DENV-induced mtDNA release involves reactive oxygen species generation and inflammasome activation. DENV infection disrupts the association between transcription factor A mitochondria (TFAM) and mtDNA and activates the mitochondrial permeability transition pores. The side-by-side comparison of TLR9 and cyclic GMP-AMP synthase (cGAS) knockdown reveals that both cGAS and TLR9 comparably contribute to DENV-induced immune activation. The significance of TLR9 in DENV-induced immune response is also confirmed in examination with the bone marrow-derived DCs prepared from Tlr9-knockout mice. Our study unravels a previously unrecognized phenomenon in which infection with an RNA virus, DENV, activates TLR9 signaling by inducing mtDNA release in human DCs.


Dendritic Cells/metabolism , Dendritic Cells/virology , Dengue Virus/physiology , Dengue/metabolism , Dengue/virology , RNA, Viral/metabolism , Signal Transduction , Toll-Like Receptor 9/metabolism , Animals , Cytosol/metabolism , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/pathology , Dengue/pathology , Endodeoxyribonucleases/metabolism , Extracellular Space/metabolism , Gene Knockdown Techniques , Humans , Inflammasomes/metabolism , Interferons/genetics , Interferons/metabolism , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
16.
Sci Rep ; 8(1): 8608, 2018 06 05.
Article En | MEDLINE | ID: mdl-29872158

Liver cirrhotic patients suffer from a seemingly unpredictable risk of hepatocellular carcinoma (HCC). Here, an HCC risk score R (0 ≦ R ≦ 1) was derived from commonly tested haematological and biochemical parameters. In the score-derivation Taiwanese cohort (144 cirrhosis versus 48 HCC-remission patients), the score had an area-under-the-curve (AUC) of 0.70 (95% confidence interval [CI], 0.61-0.78, P < 0.001). When validated in a Korean cohort (78 cirrhosis versus 23 HCC-remission patients), the AUC was 0.68 (CI, 0.56-0.80, P = 0.009). In a multicentre prospective cohort (478 cirrhotic patients prospectively followed for HCC occurrence), the hazard ratio with respect to R was 2.344 (CI = 1.183-4.646, P = 0.015). The cumulative incidences of HCC at two years after patient enrolment were 9.6% and 1.7% for the high-risk (R ≧ 0.5) and low-risk (R < 0.5) groups, respectively (P < 0.001). At the end of the study, the incidences were 10.9% and 5.0%, respectively (P = 0.012). The majority of HCCs (23/26) in the high-risk group emerged within the first two years of follow-up. In conclusion, an HCC risk score was developed for cirrhotic patients that effectively predicted HCC in a prospective cohort study.


Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/epidemiology , Decision Support Techniques , Liver Cirrhosis/complications , Aged , Biomarkers/blood , Blood Chemical Analysis , Female , Humans , Incidence , Korea/epidemiology , Male , Middle Aged , Prospective Studies , Risk Assessment , Taiwan/epidemiology
17.
Anticancer Res ; 37(6): 2885-2891, 2017 06.
Article En | MEDLINE | ID: mdl-28551625

Background /Aim: Reticulon 3 (RTN3), resides predominantly in the endoplasmic reticulum and has opposite regulatory effects on Hepatitis C virus (HCV) replication through interacting with NS5A and NS4B proteins. This study aimed to unravel the actual effect of RTN3 on HCV replication. MATERIALS AND METHODS: A total of 115 HCV-related hepatocellular carcinoma patients receiving hepatectomy was enrolled in this study. The hepatic HCV RNA and RTN3 protein levels in the non-cancerous liver tissues were examined for clinical analysis. RESULTS: Of the 115 patients, 16 (11.5%) were occult HBV infection (positive for tissue HBV DNA. Univariate followed by multivariate analysis revealed that intrahepatic RTN3 levels were independently associated with higher HCV viral load (p=0.018) and HCV genotype 1 (p=0.017). Multivariate analysis revealed that HCV genotype 1, tumor size, albumin and aspartate transaminase associated with a shorter recurrence-free survival (p<0.05). CONCLUSION: Higher intrahepatic RTN3 levels independently correlated with higher intrahepatic HCV RNA levels and genotype 1 HCV.


Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Hepacivirus/genetics , Hepatitis C, Chronic/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Aged , Antibodies, Viral/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Female , Genotype , Hepacivirus/immunology , Hepacivirus/physiology , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/virology , Humans , Liver/pathology , Liver/virology , Liver Neoplasms/blood , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Middle Aged , RNA, Viral/analysis , Tumor Burden , Virus Replication
18.
Sci Rep ; 7(1): 523, 2017 03 31.
Article En | MEDLINE | ID: mdl-28364124

With the availability of potent antiviral therapies, complete suppression of hepatitis B virus (HBV) replication and total eradication of hepatitis C virus (HCV) can now be achieved. Despite these advances, hepatocellular carcinoma (HCC) still develops in a substantial proportion of cirrhotic patients, suggesting that host factors remain critical. Dysregulation of miRNAs is noted in many cancers, and circulating miRNAs can be readily assayed. In this study, we aimed to develop a circulating miRNA signature to assess the risk of HCC in cirrhotic patients. We first discovered that HBV- and HCV-related cirrhotic patients had distinguishable circulating miRNA profiles. A cohort of 330 cirrhotic patients was then compared against a cohort of 42 early HCC patients with complete remission. A score comprising 5 miRNAs and a binary etiology variable was established that was capable of differentiating between these two groups (AUC = 72.5%, P < 0.001). The 330 cirrhotic patients were further stratified into high- and low-risk groups, and all patients were longitudinally followed for 752 (11-891) days. Of them, 19 patients developed HCC. The high-risk group had significantly higher cumulative HCC incidence (P = 0.038). In summary, a circulating miRNA-based score was developed that is capable of assessing HCC risks in cirrhotic patients.


Carcinoma, Hepatocellular/etiology , Circulating MicroRNA , Liver Cirrhosis/complications , Liver Neoplasms/etiology , Aged , Biomarkers , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/epidemiology , Cross-Sectional Studies , Female , Humans , Liver Cirrhosis/blood , Liver Neoplasms/blood , Liver Neoplasms/diagnosis , Liver Neoplasms/epidemiology , Male , Middle Aged , Prognosis , ROC Curve , Reproducibility of Results , Risk Assessment , Risk Factors
19.
Biomed J ; 40(1): 5-8, 2017 Feb.
Article En | MEDLINE | ID: mdl-28411883

Autophagy is an evolutionarily conserved process by which eukaryotic cells eliminate intracellular components via the lysosomal degradation process. This cell self-digestion process was first discovered and morphologically characterized in the late 1950s and early 1960s. The genetic screen studies in baker's yeast in the 1990s further identified the essential genes functioning in the autophagic process. In the past two decades, the detailed molecular process involved in the completion of autophagy was delineated. Additionally, autophagy has been implied to function in many aspects of biological processes, including maintenance of organelle integrity, protein quality control, regulation of the stress response, and immunity. In addition to maintain cell homeostasis, autophagy has recently been shown to be modulated and to participate in the pathogenesis of human diseases, such as pathogen infections, neurodegenerative diseases, and tumor development. Overall, the breakthrough in autophagy research relies on the discovery of autophagy-related genes (ATGs) using a genetic screening approach in Saccharomyces cerevisiae, which was established by Yoshinori Ohsumi. This year the Nobel Committee has awarded Yoshinori Ohsumi the Nobel Prize in Physiology or Medicine for his remarkable contribution to autophagy research.


Autophagy/physiology , Homeostasis/physiology , Nobel Prize , Animals , Humans , Neoplasms/metabolism , Saccharomyces cerevisiae/genetics
20.
Biochem Biophys Res Commun ; 454(1): 19-24, 2014 Nov 07.
Article En | MEDLINE | ID: mdl-25305482

Hepatitis C virus (HCV) is a positive-strand RNA virus responsible for chronic liver disease and hepatocellular carcinoma (HCC). RacGTPase-activating protein 1 (RacGAP1) plays an important role during GTP hydrolysis to GDP in Rac1 and CDC42 protein and has been demonstrated to be upregulated in several cancers, including HCC. However, the molecular mechanism leading to the upregulation of RacGAP1 remains poorly understood. Here, we showed that RacGAP1 levels were enhanced in HCV cell-culture-derived (HCVcc) infection. More importantly, we illustrated that RacGAP1 interacts with the viral protein NS5B in mammalian cells. The small interfering RNA (siRNA)-mediated knockdown of RacGAP1 in human hepatoma cell lines inhibited replication of HCV RNA, protein, and production of infectious particles of HCV genotype 2a strain JFH1. Conversely, these were reversed by the expression of a siRNA-resistant RacGAP1 recombinant protein. In addition, viral protein NS5B polymerase activity was significantly reduced by silencing RacGAP1 and, vice versa, was increased by overexpression of RacGAP1 in a cell-based reporter assay. Our results suggest that RacGAP1 plays a crucial role in HCV replication by affecting viral protein NS5B polymerase activity and holds importance for antiviral drug development.


GTPase-Activating Proteins/metabolism , Hepacivirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Allosteric Regulation , Antiviral Agents/pharmacology , Cell Line, Tumor , Drug Discovery , Enzyme Inhibitors/pharmacology , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , Genotype , HEK293 Cells , Hepacivirus/drug effects , Hepacivirus/genetics , Host-Pathogen Interactions/physiology , Humans , RNA, Small Interfering/genetics , Replicon , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics
...