Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(45): 78948-78964, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-29108278

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a serious health problem in China and Southeast Asia. Relapse is the major cause of mortality, but mechanisms of relapse are mysterious. Epstein-Barr virus (EBV) reactivation and host genomic instability (GI) have correlated with NPC development. Previously, we reported that lytic early genes DNase and BALF3 induce genetic alterations and progressive malignancy in NPC cells, implying lytic proteins may be required for NPC relapse. In this study, we show that immediate early gene BRLF1 induces chromosome mis-segregation and genomic instability in the NPC cells. Similar phenomenon was also demonstrated in 293 and zebrafish embryonic cells. BRLF1 nuclear localization signal (NLS) mutant still induced genomic instability and inhibitor experiments revealed that BRLF1 interferes with chromosome segregation and induces genomic instability by activating Erk signaling. Furthermore, the chromosome aberrations and tumorigenic features of NPC cells were significantly increased with the rounds of BRLF1 expression, and these cells developed into larger tumor nodules in mice. Therefore, BRLF1 may be the important factor contributing to NPC relapse and targeting BRLF1 may benefit patients.

2.
J Biomed Sci ; 24(1): 2, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28056971

ABSTRACT

BACKGROUND: Lytic reactivation of EBV has been reported to play an important role in human diseases, including NPC carcinogenesis. Inhibition of EBV reactivation is considered to be of great benefit in the treatment of virus-associated diseases. For this purpose, we screened for inhibitory compounds and found that apigenin, a flavonoid, seemed to have the ability to inhibit EBV reactivation. METHODS: We performed western blotting, immunofluorescence and luciferase analyses to determine whether apigenin has anti-EBV activity. RESULTS: Apigenin inhibited expression of the EBV lytic proteins, Zta, Rta, EAD and DNase in epithelial and B cells. It also reduced the number of EBV-reactivating cells detectable by immunofluorescence analysis. In addition, apigenin has been found to reduce dramatically the production of EBV virions. Luciferase reporter analysis was performed to determine the mechanism by which apigenin inhibits EBV reactivation: apigenin suppressed the activity of the immediate-early (IE) gene Zta and Rta promoters, suggesting it can block initiation of the EBV lytic cycle. CONCLUSION: Taken together, apigenin inhibits EBV reactivation by suppressing the promoter activities of two viral IE genes, suggesting apigenin is a potential dietary compound for prevention of EBV reactivation.


Subject(s)
Apigenin/pharmacology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Viral Proteins/metabolism , Virus Activation/drug effects , Cell Line , Epstein-Barr Virus Infections/drug therapy , Humans , Viral Proteins/antagonists & inhibitors
3.
Antiviral Res ; 132: 99-110, 2016 08.
Article in English | MEDLINE | ID: mdl-27185626

ABSTRACT

The lytic reactivation of Epstein-Barr virus (EBV) has been reported to be strongly associated with several human diseases, including nasopharyngeal carcinoma (NPC). Inhibition of the EBV lytic cycle has been shown to be of great benefit in the treatment of EBV-associated diseases. The administration of dietary compounds is safer and more convenient than other approaches to preventing EBV reactivation. We screened several dietary compounds for their ability to inhibit EBV reactivation in NPC cells. Among them, the flavonoid luteolin showed significant inhibition of EBV reactivation. Luteolin inhibited protein expression from EBV lytic genes in EBV-positive epithelial and B cell lines. It also reduced the numbers of EBV-reactivating cells detected by immunofluorescence analysis and reduced the production of virion. Furthermore, luteolin reduced the activities of the promoters of the immediate-early genes Zta (Zp) and Rta (Rp) and also inhibited Sp1-luc activity, suggesting that disruption of Sp1 binding is involved in the inhibitory mechanism. CHIP analysis revealed that luteolin suppressed the activities of Zp and Rp by deregulating Sp1 binding. Taken together, luteolin inhibits EBV reactivation by repressing the promoter activities of Zp and Rp, suggesting luteolin is a potential dietary compound for prevention of virus infection.


Subject(s)
Genes, Immediate-Early , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Luteolin/pharmacology , Promoter Regions, Genetic , Transcriptional Activation/drug effects , Virus Activation/drug effects , Cell Line, Tumor , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/drug effects , Humans , Protein Binding , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Virus Replication/drug effects
4.
Oncotarget ; 7(14): 18999-9017, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26967558

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a malignancy derived from the epithelial cells of the nasopharynx. Although a combination of radiotherapy with chemotherapy is effective for therapy, relapse and metastasis after remission remain major causes of mortality. Epstein-Barr virus (EBV) is believed to be one of causes of NPC development. We demonstrated previously that EBV reactivation is important for the carcinogenesis of NPC. We sought, therefore, to determine whether EBV reactivation can be a target for retardation of relapse of NPC. After screening, we found luteolin is able to inhibit EBV reactivation. It inhibited EBV lytic protein expression and repressed the promoter activities of two major immediate-early genes, Zta and Rta. Furthermore, luteolin was shown to reduce genomic instability induced by recurrent EBV reactivation in NPC cells. EBV reactivation-induced NPC cell proliferation and migration, as well as matrigel invasiveness, were also repressed by luteolin treatment. Tumorigenicity in mice, induced by EBV reactivation, was decreased profoundly following luteolin administration. Together, these results suggest that inhibition of EBV reactivation is a novel approach to prevent the relapse of NPC.


Subject(s)
Herpesvirus 4, Human/physiology , Luteolin/pharmacology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/virology , Virus Activation/drug effects , Animals , Carcinogenesis , Carcinoma , Cell Line, Tumor , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Humans , Immediate-Early Proteins/genetics , Mice , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Trans-Activators/genetics
5.
PLoS One ; 7(9): e44810, 2012.
Article in English | MEDLINE | ID: mdl-23024765

ABSTRACT

Seroepidemiological studies imply a correlation between Epstein-Barr virus (EBV) reactivation and the development of nasopharyngeal carcinoma (NPC). N-nitroso compounds, phorbols, and butyrates are chemicals found in food and herb samples collected from NPC high-risk areas. These chemicals have been reported to be risk factors contributing to the development of NPC, however, the underlying mechanism is not fully understood. We have demonstrated previously that low dose N-methyl-N'-nitro-N-nitrosoguanidine (MNNG, 0.1 µg/ml) had a synergistic effect with 12-O-tetradecanoylphorbol-13-acetate (TPA) and sodium butyrate (SB) in enhancing EBV reactivation and genome instability in NPC cells harboring EBV. Considering that residents in NPC high-risk areas may contact regularly with these chemical carcinogens, it is vital to elucidate the relation between chemicals and EBV and their contributions to the carcinogenesis of NPC. In this study, we constructed a cell culture model to show that genome instability, alterations of cancer hallmark gene expression, and tumorigenicity were increased after recurrent EBV reactivation in NPC cells following combined treatment of TPA/SB and MNNG. NPC cells latently infected with EBV, NA, and the corresponding EBV-negative cell, NPC-TW01, were periodically treated with MNNG, TPA/SB, or TPA/SB combined with MNNG. With chemically-induced recurrent reactivation of EBV, the degree of genome instability was significantly enhanced in NA cells treated with a combination of TPA/SB and MNNG than those treated individually. The Matrigel invasiveness, as well as the tumorigenicity in mouse, was also enhanced in NA cells after recurrent EBV reactivation. Expression profile analysis by microarray indicates that many carcinogenesis-related genes were altered after recurrent EBV reactivation, and several aberrations observed in cell lines correspond to alterations in NPC lesions. These results indicate that cooperation between chemical carcinogens can enhance the reactivation of EBV and, over recurrent reactivations, lead to alteration of cancer hallmark gene expression with resultant enhancement of tumorigenesis in NPC.


Subject(s)
Carcinogens/pharmacology , Herpesvirus 4, Human/drug effects , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Virus Activation/drug effects , Animals , Butyrates/pharmacology , Carcinoma , Cell Line, Tumor , Cell Transformation, Viral , Disease Progression , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Genomic Instability , Herpesvirus 4, Human/genetics , Humans , Methylnitronitrosoguanidine/pharmacology , Mice , Mice, SCID , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Neoplasm Invasiveness , Tetradecanoylphorbol Acetate/pharmacology , Virus Activation/genetics
6.
PLoS One ; 6(3): e17809, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21423768

ABSTRACT

Epstein-Barr virus (EBV) Rta belongs to a lytic switch gene family that is evolutionarily conserved in all gamma-herpesviruses. Emerging evidence indicates that cell cycle arrest is a common means by which herpesviral immediate-early protein hijacks the host cell to advance the virus's lytic cycle progression. To examine the role of Rta in cell cycle regulation, we recently established a doxycycline (Dox)-inducible Rta system in 293 cells. In this cell background, inducible Rta modulated the levels of signature G1 arrest proteins, followed by induction of the cellular senescence marker, SA-ß-Gal. To delineate the relationship between Rta-induced cell growth arrest and EBV reactivation, recombinant viral genomes were transferred into Rta-inducible 293 cells. Somewhat unexpectedly, we found that Dox-inducible Rta reactivated both EBV and Kaposi's sarcoma-associated herpesvirus (KSHV), to similar efficacy. As a consequence, the Rta-mediated EBV and KSHV lytic replication systems, designated as EREV8 and ERKV, respectively, were homogenous, robust, and concurrent with cell death likely due to permissive lytic replication. In addition, the expression kinetics of EBV lytic genes in Dox-treated EREV8 cells was similar to that of their KSHV counterparts in Dox-induced ERKV cells, suggesting that a common pathway is used to disrupt viral latency in both cell systems. When the time course was compared, cell cycle arrest was achieved between 6 and 48 h, EBV or KSHV reactivation was initiated abruptly at 48 h, and the cellular senescence marker was not detected until 120 h after Dox treatment. These results lead us to hypothesize that in 293 cells, Rta-induced G1 cell cycle arrest could provide (1) an ideal environment for virus reactivation if EBV or KSHV coexists and (2) a preparatory milieu for cell senescence if no viral genome is available. The latter is hypothetical in a transient-lytic situation.


Subject(s)
Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Viral Proteins/metabolism , Virus Activation/physiology , Carcinoma , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Doxycycline/pharmacology , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Time Factors , Virus Activation/drug effects , Virus Replication/drug effects
7.
Am J Epidemiol ; 173(3): 292-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21148719

ABSTRACT

In the present study, the authors compared the long-term risk of nasopharyngeal carcinoma (NPC) of male participants in an NPC multiplex family cohort with that of controls in a community cohort in Taiwan after adjustment for anti-Epstein-Barr virus (EBV) seromarkers and cigarette smoking. A total of 43 incident NPC cases were identified from the 1,019 males in the NPC multiplex family cohort and the 9,622 males in the community cohort, for a total of 8,061 person-years and 185,587 person-years, respectively. The adjusted hazard ratio was 6.8 (95% confidence interval (CI): 2.3, 20.1) for the multiplex family cohort compared with the community cohort. In the evaluation of anti-EBV viral capsid antigen immunoglobulin A and anti-EBV deoxyribonuclease, the adjusted hazard ratios were 2.8 (95% CI: 1.3, 6.0) and 15.1 (95% CI: 4.2, 54.1) for those positive for 1 EBV seromarker and positive for both seromarkers, respectively, compared with those negative for both EBV seromarkers. The adjusted hazard ratio was 31.0 (95% CI: 9.7, 98.7) for participants who reported a family history of NPC and who were anti-EBV-seropositive compared with individuals without such a history who were anti-EBV-seronegative. The findings suggest that both family history of NPC and anti-EBV seropositivity are important determinants of subsequent NPC risk and that the effect of family history on NPC risk cannot be fully explained by mediation through EBV serologic responses.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Herpesvirus 4, Human/immunology , Nasopharyngeal Neoplasms/epidemiology , Nasopharyngeal Neoplasms/etiology , Adult , Antigens, Viral/blood , Biomarkers, Tumor/blood , Cohort Studies , Deoxyribonucleases/blood , Family , Female , Humans , Immunoglobulin A/blood , Incidence , Male , Middle Aged , Nasopharyngeal Neoplasms/blood , Proportional Hazards Models , Prospective Studies , Registries , Risk Factors , Smoking/adverse effects , Smoking/epidemiology , Surveys and Questionnaires , Taiwan/epidemiology , Viral Proteins/blood
8.
Nucleic Acids Res ; 38(6): 1932-49, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20034954

ABSTRACT

Epstein-Barr Virus (EBV) DNase (BGLF5) is an alkaline nuclease and has been suggested to be important in the viral life cycle. However, its effect on host cells remains unknown. Serological and histopathological studies implied that EBV DNase seems to be correlated with carcinogenesis. Therefore, we investigate the effect of EBV DNase on epithelial cells. Here, we report that expression of EBV DNase induces increased formation of micronucleus, an indicator of genomic instability, in human epithelial cells. We also demonstrate, using gammaH2AX formation and comet assay, that EBV DNase induces DNA damage. Furthermore, using host cell reactivation assay, we find that EBV DNase expression repressed damaged DNA repair in various epithelial cells. Western blot and quantitative PCR analyses reveal that expression of repair-related genes is reduced significantly in cells expressing EBV DNase. Host shut-off mutants eliminate shut-off expression of repair genes and repress damaged DNA repair, suggesting that shut-off function of BGLF5 contributes to repression of DNA repair. In addition, EBV DNase caused chromosomal aberrations and increased the microsatellite instability (MSI) and frequency of genetic mutation in human epithelial cells. Together, we propose that EBV DNase induces genomic instability in epithelial cells, which may be through induction of DNA damage and also repression of DNA repair, subsequently increases MSI and genetic mutations, and may contribute consequently to the carcinogenesis of human epithelial cells.


Subject(s)
Deoxyribonucleases/metabolism , Genomic Instability , Viral Proteins/metabolism , Cell Line , Chromosome Aberrations , DNA Breaks , DNA Damage , DNA Repair/genetics , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Humans , Micronuclei, Chromosome-Defective , Microsatellite Instability , Mutation , Protein Biosynthesis , Transcription, Genetic , Ultraviolet Rays
9.
Int J Cancer ; 124(9): 2016-25, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19132751

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an endemic malignancy prevalent in South East Asia. Epidemiological studies have associated this disease closely with Epstein-Barr virus (EBV) infection. Previous studies also showed that EBV reactivation is implicated in the progression of NPC. Thus, we proposed that recurrent reactivations of EBV may be important for its pathogenic role. In this study, NPC cell lines latently infected with EBV, NA and HA, and the corresponding EBV-negative NPC cell lines, NPC-TW01 (TW01) and HONE-1, were treated with 12-O-tetradecanoylphorbol-13-acetate (TPA) and sodium n-butyrate (SB) for lytic cycle induction. A single treatment with TPA/SB revealed that DNA double-strand breaks and formation of micronuclei (a marker for genome instability) were associated with EBV reactivation in NA and HA cells. Examination of EBV early genes had identified several lytic proteins, particularly EBV DNase, as potent activators that induced DNA double-strand breaks and contribute to genome instability. Recurrent reactivations of EBV in NA and HA cells resulted in a marked increase of genome instability. In addition, the degree of chromosomal aberrations, as shown by chromosome structural variants and DNA copy-number alterations, is proportional to the frequency of TPA/SB-induced EBV reactivation. Whereas these DNA abnormalities were limited in EBV-negative TW01 cells with mock or TPA/SB treatment, and were few in mock-treated NA cells. The invasiveness and tumorigenesis assays also revealed a profound increase in both characteristics of the repeatedly reactivated NA cells. These results suggest that recurrent EBV reactivations may result in accumulation of genome instability and promote the tumor progression of NPC.


Subject(s)
Epstein-Barr Virus Infections/virology , Genomic Instability , Herpesvirus 4, Human/physiology , Micronuclei, Chromosome-Defective/drug effects , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Virus Activation/drug effects , Animals , Butyrates/pharmacology , Carcinogens/pharmacology , Comparative Genomic Hybridization , DNA Damage/drug effects , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Fluorescent Antibody Technique , Gene Dosage , Gene Expression Regulation, Viral , Genome, Viral/drug effects , Genome, Viral/physiology , Humans , Mice , Mice, SCID , Nasopharyngeal Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Recurrence , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Virus Replication/drug effects
10.
Int J Cancer ; 111(1): 117-23, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15185352

ABSTRACT

Most adults have been infected with EBV. Many studies have indicated that antibodies against specific EBV antigens, particularly IgA antibodies, can be predictive or prognostic of EBV-associated malignancies, such as NPC. We hypothesized that healthy individuals from families with a history of multiple members affected with NPC (who therefore might be genetically susceptible to NPC themselves) might have an EBV antibody profile that is distinct from that seen in healthy individuals from the community at large. To explore this possibility and examine determinants of anti-EBV antibody levels in healthy, high-risk individuals, we evaluated data from 2 parallel studies of NPC in Taiwan, which included 1,229 healthy members of families in which 2 or more individuals were affected with NPC and 320 controls from the community at large. Blood collected from participants was tested for IgA antibodies against EBV VCA and EBNA-1 and for neutralizing antibodies against EBV DNase using standard assays. We observed evidence of familial aggregation of EBV seroreactivity among individuals from high-risk, multiplex NPC families. Anti-VCA IgA and anti-EBNA-1 IgA antibody seroprevalence in unaffected family members of NPC cases was 5-6 times higher than in members of the community (p < 0.01). This elevated seroprevalence among unaffected individuals from high-risk families was observed regardless of the relationship of the unaffected individual to the closest affected relative (siblings, parents, children or spouses). No sociodemographic or environmental factors examined were found to strongly and consistently correlate with elevated seroprevalence, but patterns emerged of increasing seroprevalence among older individuals and among females. Unaffected individuals from high-risk NPC families have elevated anti-EBV IgA antibody titers. The etiologic and clinical implications of this finding remain to be established.


Subject(s)
Antibodies, Viral/analysis , Carcinoma/virology , Epstein-Barr Virus Infections/immunology , Genetic Predisposition to Disease , Immunoglobulin A/analysis , Nasopharyngeal Neoplasms/virology , Adolescent , Adult , Aged , Carcinoma/genetics , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Nasopharyngeal Neoplasms/genetics , Pedigree , Risk Factors , Seroepidemiologic Studies , Taiwan
11.
Hybrid Hybridomics ; 23(1): 29-37, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15000846

ABSTRACT

A monoclonal antibody (MAb), designated 3E8, was produced against the Epstein-Barr virus BHRF1 which is a viral homologue of the anti-apoptotic protein Bcl-2. The MAb recognized the BHRF1 protein in extracts from EBV-containing cell lines after activation and EBV-negative cell lines transfected by the BHRF1 gene. Epitope mapping by Western blot analysis revealed that the antibody bound region encompassing amino acid residues 28-33 of the BHRF1. In addition to immunoblotting, the MAb could be applied widely in detection of the BHRF1 in many assays, including immunofluorescence assay, immunohistochemistry, enzyme-linked immunosorbent assay and immunoprecipitation. Most of all, when used in immunoprecipitation experiments, the MAb 3E8 showed a better effect than the existing anti-BHRF1 MAbs since radioactive isotopes were not required to intensify signals of its target antigen. Based on its great use in a variety of immunological reactions, it is a powerful tool to elucidate the biological functions of BHRF1.


Subject(s)
Antibodies, Monoclonal/immunology , Viral Proteins/immunology , Animals , Blotting, Western , Callithrix , Cell Line , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Plasmids , Proto-Oncogene Proteins c-bcl-2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...