Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
PLoS One ; 19(4): e0299198, 2024.
Article in English | MEDLINE | ID: mdl-38635661

ABSTRACT

Herpesviruses have two distinct life cycle stages, latency and lytic replication. Epstein-Barr virus (EBV), a gamma-herpesvirus, establishes latency in vivo and in cultured cells. Cell lines harboring latent EBV can be induced into the lytic cycle by treatment with chemical inducing agents. In the Burkitt lymphoma cell line HH514-16 the viral lytic cycle is triggered by butyrate, a histone deacetylase (HDAC) inhibitor. Butyrate also alters expression of thousands of cellular genes. However, valproic acid (VPA), another HDAC inhibitor with global effects on cellular gene expression blocks EBV lytic gene expression in Burkitt lymphoma cell lines. Valpromide (VPM), an amide derivative of VPA, is not an HDAC inhibitor, but like VPA blocks induction of the EBV lytic cycle. VPA and VPM are the first examples of inhibitors of initial stages of lytic reactivation. We compared the effects of VPA and VPM, alone and in combination with butyrate, on host cellular gene expression using whole transcriptome analysis (RNA-seq). Gene expression was analyzed 6 h after addition of the compounds, a time before the first EBV lytic transcripts are detected. The results address two alternative, yet possibly complementary, mechanisms for regulation of EBV lytic reactivation. First, cellular genes that were up- or down-regulated by butyrate, but no longer altered in the presence of VPA or VPM, represent genes that correlated with EBV lytic reactivation. Second, genes regulated similarly by VPA and VPM in the absence and presence of butyrate are candidates for suppressors of EBV reactivation. Two genes upregulated by the lytic cycle inhibitors, CHAC1 and SLC7A11, are related to redox status and the iron-dependent cell death pathway ferroptosis. This study generates new hypotheses for control of the latency to lytic cycle switch of EBV and provides the first description of effects of the anti-convulsant drug VPM on global human cellular gene expression.


Subject(s)
Burkitt Lymphoma , Epstein-Barr Virus Infections , Valproic Acid/analogs & derivatives , Humans , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Herpesvirus 4, Human/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/metabolism , Epstein-Barr Virus Infections/drug therapy , Virus Activation , Gene Expression Profiling , Butyrates/pharmacology
2.
J Clin Immunol ; 42(1): 108-118, 2022 01.
Article in English | MEDLINE | ID: mdl-34655400

ABSTRACT

X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus (EBV) infection and N-linked glycosylation defect (XMEN) disease is an inborn error of immunity caused by loss-of-function mutations in the magnesium transporter 1 (MAGT1) gene. The original studies of XMEN patients focused on impaired magnesium regulation, leading to decreased EBV-cytotoxicity and the loss of surface expression of the activating receptor "natural killer group 2D" (NKG2D) on CD8+ T cells and NK cells. In vitro studies showed that supraphysiological supplementation of magnesium rescued these defects. Observational studies in 2 patients suggested oral magnesium supplementation could decrease EBV viremia. Hence, we performed a randomized, double-blind, placebo-controlled, crossover study in 2 parts. In part 1, patients received either oral magnesium L-threonate (MLT) or placebo for 12 weeks followed by 12 weeks of the other treatment. Part 2 began with 3 days of high-dose intravenous (IV) magnesium sulfate (MgSO4) followed by open-label MLT for 24 weeks. One EBV-infected and 3 EBV-naïve patients completed part 1. One EBV-naïve patient was removed from part 2 of the study due to asymptomatic elevation of liver enzymes during IV MgSO4. No change in EBV or NKG2D status was observed. In vitro magnesium supplementation experiments in cells from 14 XMEN patients failed to significantly rescue NKG2D expression and the clinical trial was stopped. Although small, this study indicates magnesium supplementation is unlikely to be an effective therapeutic option in XMEN disease.


Subject(s)
Cation Transport Proteins , Epstein-Barr Virus Infections , Neoplasms , X-Linked Combined Immunodeficiency Diseases , CD8-Positive T-Lymphocytes , Cation Transport Proteins/genetics , Cross-Over Studies , Dietary Supplements , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Humans , Magnesium/metabolism , Magnesium/therapeutic use , Neoplasms/genetics , X-Linked Combined Immunodeficiency Diseases/genetics
3.
Pharm Biol ; 59(1): 741-747, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34155950

ABSTRACT

CONTEXT: Simiao Qingwen Baidu decoction (SQBD), a traditional Chinese medicine prescription, can ameliorate Epstein-Barr virus (EBV) induced disease. However, its mechanism still remains unknown. OBJECTIVE: To detect the mechanism of SQBD in EBV-induced B lymphoproliferative disease in vitro. MATERIALS AND METHODS: Sprague-Dawley (SD) rats (n = 20) were given SQBD (10 mL/kg) by gavage once a day for 7 d. SQBD-containing serum was obtained from abdominal aortic blood of rats, and diluted with medium to obtain 5%, 10% or 20%-medicated serum. SD rats (n = 10) were given normal saline, and normal serum was collected as a control. EBV-transformed B cells (CGM1) were cultured in medium containing 5%, 10% or 20%-medicated serum. CGM1 cells were treated with normal serum as a control. Cell viability and apoptosis were examined. The expression and activity of proteins were assessed. RESULTS: We found that IC50 (83 ± 26.07%, 24 h; 69.88 ± 4.69%, 48 h) of 10% medicated serum was higher than that of 5% (25.47 ± 6.98%, 24 h; 21.62 ± 7.30%, 48 h) and 20%-medicated serum (51 ± 7.25%, 24 h; 56.03 ± 2.56%, 48 h). Moreover, SQBD promoted apoptosis of CGM1 cells by regulating EBV latency proteins expression. SQBD inhibited EBV-induced lytic viral replication. CONCLUSIONS: Our data confirmed that SQBD inhibits EBV-induced B lymphoproliferative disease and lytic viral replication. This work provides a theoretical basis for the mechanism of SQBD in EBV-induced B lymphoproliferative disease, and SQBD may be an effectively therapeutic drug for EBV-induced B lymphoproliferative disease.


Subject(s)
B-Lymphocytes/drug effects , Drugs, Chinese Herbal/therapeutic use , Herpesvirus 4, Human/drug effects , Lymphoproliferative Disorders/drug therapy , Virus Replication/drug effects , Animals , B-Lymphocytes/physiology , Drugs, Chinese Herbal/pharmacology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Male , Rats , Rats, Sprague-Dawley , Virus Replication/physiology
4.
Am J Transplant ; 21(10): 3465-3471, 2021 10.
Article in English | MEDLINE | ID: mdl-33942495

ABSTRACT

Primary central nervous system lymphoma (PCNSL) occurring following organ transplantation (post-transplantation lymphoproliferative disorder [PTLD]) is a highly aggressive non-Hodgkin lymphoma. It is typically treated with high-dose methotrexate-based regimens. Outcomes are dismal and clinical trials are lacking. It is almost always Epstein-Barr virus (EBV) associated. Two patients (CA1-2) presented with EBV-associated PCNSL after renal transplant. CA1 was on hemodialysis and had prior disseminated cryptococcus and pseudomonas bronchiectasis, precluding treatment with methotrexate. CA2 was refractory to methotrexate. Both were treated off-label with the first-generation Bruton's tyrosine kinase inhibitor ibrutinib for 12 months. Cerebrospinal fluid penetration at therapeutic levels was confirmed in CA1 despite hemodialysis. Both patients entered remission by 2 months. Sequencing confirmed absence of genetic aberrations in human leukocyte antigen (HLA) class I/II and antigen-presentation/processing genes, indicating retention of the ability to present EBV-antigens. Between Weeks 10 and 13, they received third-party EBV-specific T cells for consolidation with no adverse effects. They remain in remission ≥34 months since therapy began. The strength of these findings led to an ongoing phase I study (ACTRN12618001541291).


Subject(s)
Epstein-Barr Virus Infections , Lymphoma, Non-Hodgkin , Lymphoproliferative Disorders , Adenine/analogs & derivatives , Central Nervous System , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/etiology , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/etiology , Piperidines , T-Lymphocytes
5.
Phytomedicine ; 87: 153591, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34029937

ABSTRACT

BACKGROUND: The outbreak of coronavirus (SARS-CoV-2) disease caused more than 100,000,000 people get infected and over 2,200,000 people being killed worldwide. However, the current developed vaccines or drugs may be not effective in preventing the pandemic of COVID-19 due to the mutations of coronavirus and the severe side effects of the newly developed vaccines. Chinese herbal medicines and their active components play important antiviral activities. Corilagin exhibited antiviral effect on human immunodeficiency virus (HIV), hepatitis C virus (HCV) and Epstein-Barr virus (EBV). However, whether it blocks the interaction between SARS-CoV-2 RBD and hACE2 has not been elucidated. PURPOSE: To characterize an active compound, corilagin derived from Phyllanthus urinaria as potential SARS-CoV-2 entry inhibitors for its possible preventive application in daily anti-virus hygienic products. METHODS: Computational docking coupled with bio-layer interferometry, BLI were adopted to screen more than 1800 natural compounds for the identification of SARS-CoV-2 spike-RBD inhibitors. Corilagin was confirmed to have a strong binding affinity with SARS-CoV-2-RBD or human ACE2 (hACE2) protein by the BLI, ELISA and immunocytochemistry (ICC) assay. Furthermore, the inhibitory effect of viral infection of corilagin was assessed by in vitro pseudovirus system. Finally, the toxicity of corilagin was examined by using MTT assay and maximal tolerated dose (MTD) studies in C57BL/6 mice. RESULTS: Corilagin preferentially binds to a pocket that contains residues Cys 336 to Phe 374 of spike-RBD with a relatively low binding energy of -9.4 kcal/mol. BLI assay further confirmed that corilagin exhibits a relatively strong binding affinity to SARS-CoV-2-RBD and hACE2 protein. In addition, corilagin dose-dependently blocks SARS-CoV-2-RBD binding and abolishes the infectious property of RBD-pseudotyped lentivirus in hACE2 overexpressing HEK293 cells, which mimicked the entry of SARS-CoV-2 virus in human host cells. Finally, in vivo studies revealed that up to 300 mg/kg/day of corilagin was safe in C57BL/6 mice. Our findings suggest that corilagin could be a safe and potential antiviral agent against the COVID-19 acting through the blockade of the fusion of SARS-CoV-2 spike-RBD to hACE2 receptors. CONCLUSION: Corilagin could be considered as a safe and environmental friendly anti-SARS-CoV-2 agent for its potential preventive application in daily anti-virus hygienic products.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Glucosides/pharmacology , Host-Pathogen Interactions/drug effects , Hydrolyzable Tannins/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/toxicity , COVID-19 , Epstein-Barr Virus Infections/drug therapy , Glucosides/chemistry , Glucosides/toxicity , HEK293 Cells , Humans , Hydrolyzable Tannins/chemistry , Hydrolyzable Tannins/toxicity , Lentivirus Infections/drug therapy , Male , Maximum Tolerated Dose , Mice, Inbred C57BL , Molecular Docking Simulation , Spike Glycoprotein, Coronavirus/chemistry
6.
Br J Haematol ; 189(2): 257-268, 2020 04.
Article in English | MEDLINE | ID: mdl-31958882

ABSTRACT

Patients with EBV-positive diffuse large B cell lymphoma not otherwise specified (EBV+ DLBCL (NOS)) recurrently present with advanced age and reduced performance status. They are therefore insufficiently represented in clinical trials and treatment is likely to differ. Here we assess clinicopathological characteristics, therapeutic variability and clinical outcome in the largest consecutively diagnosed EBV+ DLBCL (NOS) cohort published to date (n = 80; median age 70 years; range 19-90). Centralized and systematic haematopathological panel review was performed. By immunohistochemistry 60/80 patients were CD30-positive. Further, we identified nine EBV+ DLBCL (NOS) patients with associated or composite peripheral T cell lymphoma at diagnosis or relapse (preceded by clonal T cell populations within the initial DLBCL biopsy in 4/5 cases). Most patients (80%) were treated with R-CHOP-type therapy and 16 patients received none or less intensiveprotocols. Upon univariate analysis both R-CHOP-type therapy (OS: P < 0.0001; PFS: P = 0.0617) and negativity for CD30 (OS: P = 0.0002; PFS: P = 0.0002) showed a protective 66 effect, maintained upon multivariate analysis. In a propensity-score matched analysis with a cohort of non-EBV+ DLBCL (NOS) patients, balanced for all revised-international prognostic index factors, we found an EBV-association to hold no significant impact on progression-free and overall survival whilst exhibiting a trend favouring EBV-negativity (OS: P = 0.116; PFS: P = 0.269). Our findings provide insight into the clinical course of EBV+ DLBCL (NOS), highlight the ramifications of CD30-expression and underline the superior therapeutic efficacy of R-CHOP immunochemotherapy. Alternative therapies, incorporating tumour biology (e.g. CD30 directed therapies) need to be explored in EBV+ DLBCL (NOS) patients. Moreover our data advert to the close relationship between EBV+ DLBCL (NOS) and peripheral T cell lymphomas.


Subject(s)
Epstein-Barr Virus Infections/complications , Lymphoma, Large B-Cell, Diffuse/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Epstein-Barr Virus Infections/drug therapy , Female , Humans , Male , Middle Aged , Survival Analysis , Young Adult
7.
Molecules ; 24(21)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31653035

ABSTRACT

Mycotherapy has been shown to improve the overall response rate during cancer treatment and reduce some chemotherapy-related adverse events. Ganoderma lucidum is a traditional mushroom used for pharmaceutical purposes. G. lucidum extracts (GLE) showed potential antitumor activities against several cancers. These tumor inhibitory effects of GLE were attributed to the suppression of the proliferation and metastasis of cancer cells. Epstein-Barr virus (EBV)-associated gastric carcinoma (EBVaGC) is defined as the monoclonal proliferation of carcinoma cells with latent EBV infection. The inhibitory effects of GLE against EBVaGC are questionable. The aim of this study was to investigate GLE as potential antitumor agents and a counterpart of quercetin (QCT) for the cotreatment in suppressing EBVaGC development. Therefore, this study conducted antitumor assays using a EBVaGC xenograft mice model and found that GLE could suppress tumor development. These inhibitory effects were significantly augmented by the low concentration of the quercetin (QCT) cotreatment in the xenograft mice. The addition of GLE in low concentrations synergistically reinforced QCT-induced apoptosis and EBV lytic reactivation. GLE contains various polysaccharides and triterpenes, such as ganoderic acid. Interestingly, the addition of ganoderic acid A (GAA) could produce similar bioactive effects like GLE in QCT-mediated antitumor activity. The GAA addition in low concentrations synergistically reinforced QCT-induced apoptosis and EBV lytic reactivation. GAA was sufficiently effective as much as GLE. Therefore, our results suggested that QCT-supplemented GLE could be a potential food adjunct for the prevention of EBVaGC development.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Epstein-Barr Virus Infections , Herpesvirus 4, Human/physiology , Plant Extracts/pharmacology , Quercetin/pharmacology , Reishi/chemistry , Stomach Neoplasms , Virus Activation/drug effects , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Female , Humans , Mice , Mice, Nude , Plant Extracts/chemistry , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Triterpenes/chemistry , Triterpenes/pharmacology
8.
J Clin Pathol ; 72(10): 651-658, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31315893

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous human virus which infects almost all humans during their lifetime and following the acute phase, persists for the remainder of the life of the individual. EBV infects B lymphocytes leading to their immortalisation, with persistence of the EBV genome as an episome. In the latent phase, EBV is prevented from reactivating through efficient cytotoxic cellular immunity. EBV reactivates (lytic phase) under conditions of psychological stress with consequent weakening of cellular immunity, and EBV reactivation has been shown to occur in a subset of individuals with each of a variety of cancers, autoimmune diseases, the autoimmune-like disease, chronic fatigue syndrome/myalgic encephalitis and under other circumstances such as being an inpatient in an intensive care unit. Chronic EBV reactivation is an important mechanism in the pathogenesis of many such diseases, yet is rarely tested for in immunocompetent individuals. This review summarises the pathogenesis of EBV infection, EBV reactivation and its role in disease, and methods which may be used to detect it. Known inhibitors of EBV reactivation and replication are discussed, including drugs licensed for treatment of other herpesviruses, licensed or experimental drugs for various other indications, compounds at an early stage of drug development and nutritional constituents such as vitamins and dietary supplements.


Subject(s)
Antiviral Agents/therapeutic use , Dietary Supplements , Epstein-Barr Virus Infections/virology , Genome, Viral/genetics , Herpesvirus 4, Human/physiology , Vitamins , B-Lymphocytes/virology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/psychology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Stress, Psychological , Virus Activation/drug effects , Virus Latency , Virus Replication/drug effects
9.
PLoS One ; 13(10): e0200043, 2018.
Article in English | MEDLINE | ID: mdl-30332435

ABSTRACT

Infection of immunocompromised individuals with normally benign opportunistic viruses is a major health burden globally. Infections with viruses such as Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), Kaposi's sarcoma virus (KSHV), adenoviruses (AdV), BK virus (BKPyV), John Cunningham virus (JCPyV), and human papillomavirus (HPV) are significant concerns for the immunocompromised, including when these viruses exist as a co-infection with human immunodeficiency virus (HIV). These viral infections are more complicated in patients with a weakened immune system, and often manifest as malignancies resulting in significant morbidity and mortality. Vaccination is not an attractive option for these immune compromised individuals due to defects in their adaptive immune response. Verdinexor is part of a novel class of small molecules known as SINE (Selective Inhibitor of Nuclear Export) compounds. These small molecules demonstrate specificity for the nuclear export protein XPO1, to which they bind and block function, resulting in sequestration of XPO1-dependent proteins in the nucleus of the cell. In antiviral screening, verdinexor demonstrated varying levels of efficacy against all of the aforementioned viruses including previously with HIV. Studies by other labs have discussed likely mechanisms of action for verdinexor (ie. XPO1-dependence) against each virus. GLP toxicology studies suggest that anti-viral activity can be achieved at a tolerable dose range, based on the safety profile of a previous phase 1 clinical trial of verdinexor in healthy human volunteers. Taken together, these results indicate verdinexor has the potential to be a broad spectrum antiviral for immunocompromised subjects for which vaccination is a poor option.


Subject(s)
Acrylamides/pharmacology , Hydrazines/pharmacology , Immunocompromised Host/drug effects , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Virus Diseases/drug therapy , Adenoviridae Infections/drug therapy , Animals , Cell Line, Tumor , Cytomegalovirus Infections/drug therapy , Drug Evaluation, Preclinical , Epstein-Barr Virus Infections/drug therapy , Fibroblasts/virology , Guinea Pigs , HEK293 Cells , HIV Infections/complications , HeLa Cells , Humans , Mice , Papillomavirus Infections/drug therapy , Polyomavirus Infections/drug therapy , Reproducibility of Results , Sarcoma, Kaposi/drug therapy , Tumor Virus Infections/drug therapy , Virus Diseases/complications , Exportin 1 Protein
10.
Mult Scler ; 23(3): 395-402, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27325604

ABSTRACT

BACKGROUND: Elevated antibody levels against Epstein-Barr virus (EBV) and a poor vitamin D status are environmental factors that may interact in relapsing-remitting multiple sclerosis (RRMS) aetiology. OBJECTIVES: To examine effects of high-dose oral vitamin D3 supplementation on antibody levels against EBV nuclear antigen 1 (EBNA1) in RRMS. METHODS: Serum 25-hydroxyvitamin D3 (25(OH)D) and immunoglobulin G antibody levels against EBNA1 (whole protein and amino acid 385-420 fragment), EBV viral capsid antigen (VCA), cytomegalovirus (CMV) and varicella zoster virus (VZV) were measured in 68 RRMS patients enrolled in a 96-week randomised double-blinded placebo-controlled clinical trial of oral vitamin D3 supplementation (20,000 IU/week) (NCT00785473). RESULTS: The mean 25(OH)D level more than doubled in the vitamin D group and was significantly higher than in the placebo group at study conclusion (123.2 versus 61.8 nmol/L, p < 0.001). Compared to the placebo group, both anti-EBNA1 protein and fragment antibody levels decreased in the vitamin D group from baseline to week 48 ( p = 0.038 and p = 0.004, respectively), but not from baseline to week 96. Vitamin D3 supplementation did not affect antibodies against VCA, CMV or VZV. CONCLUSION: The results indicate that high-dose oral vitamin D3 supplementation can affect humoral immune responses against the latent EBV antigen EBNA1 in RRMS.


Subject(s)
Cholecalciferol/therapeutic use , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human/drug effects , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adolescent , Adult , Antibodies, Viral/blood , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/blood , Female , Herpesvirus 4, Human/pathogenicity , Humans , Immunoglobulin G/blood , Male , Middle Aged , Young Adult
11.
Med Sci (Paris) ; 30(12): 1161-8, 2014 Dec.
Article in French | MEDLINE | ID: mdl-25537047

ABSTRACT

Since its discovery and description by Louis Pasteur, the budding yeast Saccharomyces cerevisiae, which was used for thousands of years for alcoholic fermentation and as a leavening agent, has become a popular model system in biology. One of the reasons for this popularity is the strong conservation from yeast to human of most of the pathways controlling cell growth and fate. In addition, at least 30 % of human genes involved in diseases have a functional homolog in yeast. Hence, yeast is now widely used for modelling and deciphering physiopathological mechanisms as well as for developing pharmacological approaches like phenotype-based drug screening. Three examples of such yeast-based chemobiological studies are presented.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Saccharomyces cerevisiae , Animals , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/immunology , Humans , Mice , Mitochondrial Diseases/drug therapy , Mitochondrial Myopathies/drug therapy , Phenotype , Prion Diseases/drug therapy , Retinitis Pigmentosa/drug therapy , Saccharomyces cerevisiae/genetics
12.
Med Sci Monit ; 20: 725-32, 2014 May 03.
Article in English | MEDLINE | ID: mdl-24793092

ABSTRACT

Background Many natural compounds were tested for the ability to suppress viral replication. The present manuscript details an analysis of high dose vitamin C therapy on patients with EBV infection. Material and Methods The data were obtained from the patient history database at the Riordan Clinic. Among people in our database who were treated with intravenous vitamin C (7.5 g to 50 g infusions) between 1997 and 2006, 178 patients showed elevated levels of EBV EA IgG (range 25 to 211 AU) and 40 showed elevated levels of EBV VCA IgM (range 25 to 140 AU). Most of these patients had a diagnosis of chronic fatigue syndrome, with the rest being diagnosed as having mononucleosis, fatigue, or EBV infection. Results Our data provide evidence that high dose intravenous vitamin C therapy has a positive effect on disease duration and reduction of viral antibody levels. Plasma levels of ascorbic acid and vitamin D were correlated with levels of antibodies to EBV. We found an inverse correlation between EBV VCA IgM and vitamin C in plasma in patients with mononucleosis and CFS meaning that patients with high levels of vitamin C tended to have lower levels of antigens in the acute state of disease. In addition, a relation was found between vitamin D levels and EBV EA IgG with lower levels of EBV early antigen IgG for higher levels of vitamin D. Conclusions The clinical study of ascorbic acid and EBV infection showed the reduction in EBV EA IgG and EBV VCA IgM antibody levels over time during IVC therapy that is consistent with observations from the literature that millimolar levels of ascorbate hinder viral infection and replication in vitro.


Subject(s)
Ascorbic Acid/administration & dosage , Ascorbic Acid/therapeutic use , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Antibodies, Viral/blood , Antigens, Viral/immunology , Ascorbic Acid/blood , Ascorbic Acid/pharmacology , Databases, Factual , Dose-Response Relationship, Drug , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/immunology , Humans , Lymphocytes/immunology , Retrospective Studies
13.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(2): 167-73, 2014 Feb.
Article in Chinese | MEDLINE | ID: mdl-24672940

ABSTRACT

OBJECTIVE: To provide evidence for Chinese medical treatment of children with EB virus infection by exploring its clinical efficacy from multiple angles. METHODS: Totally 81 children patients were randomly assigned to the treatment group (46 cases) and the control group (35 cases). Patients in the treatment group took Chinese medical decoction, while those in the control received intravenous dripping of Ganciclovir and oral administration of pidotimod. The treatment period for the two groups was 2 weeks. Patients were followed-up till the 12th week. Clinical symptoms such as fever, lymphadenopathy and hepatosplenomegaly, as well as lab indices such as abnormal lymphocyte percentage, EB virus antibody, virus DNA load, T cell subsets, immunoglobulin, and so on were observed before and after treatment, at week 4 and 12 of follow-ups. RESULTS: (1) The total effective rate at week 2 was 95.6% in the treatment group, higher than that of the control group (94.3%), but there was no statistical difference between the two groups. (2) The time for defervescence, duration of pharyngeal hyperemia, duration of swollen tonsils was shorter in the treatment group than in the control group (P<0.05). The subsidence of lymphadenopathy, hepatomegaly, and abnormal lymphocytes was better in the treatment group than in the control group (P < 0.05). (3) The positive cases of peripheral blood hetero-lymphocyte was significantly reduced after treatment, at week 4 and 12 of follow-ups both in the treatment group and the control group (P < 0.01). The expression of IgA and IgM decreased after treatment in the two groups when compared with before treatment in the same group (P < 0.05, P < 0.01). IgG in the treatment group also obviously decreased after treatment, at week 4 and 12 of follow-ups (P < 0.05, P < 0.01), while it decreased only after treatment in the control group (P < 0.05). Activities of AST and ALT in the treatment group and the AST activity in the control group were markedly improved when compared with those before treatment (P < 0.05). Compared with the control group, the abnormal lymphocyte positive case number obviously decreased in the treatment group after treatment, at week 4 and 12 of follow-ups (P < 0.05). (4) After treatment, at week 4 and 12 of follow-ups, CD3+ and CD8+ significantly decreased; CD4+, CD4/CD8, and B cells significantly increased in the two groups, when compared with before treatment (P < 0.05). NK cells significantly increased more in the treatment group after treatment, at week 4 and 12 of follow-ups, higher than before treatment as well as the control group (P < 0.05). (5) EB viral DNA and EB viral CA-IgM negative conversion case numbers significantly increased in the two groups after treatment, at week 4 and 12 of follow-ups (P < 0.05). Compared with the control group, EB viral DNA and EB viral CA-IgM negative conversion case numbers significantly increased in the treatment group after treatment and at week 4 of follow-ups (P < 0.05). CONCLUSIONS: Treatment of EB virus infection by Chinese medical treatment was effective. It could promote the recovery of EB viral infection, and reduce the risk of vicious disease after EB viral infection.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Epstein-Barr Virus Infections/drug therapy , Phytotherapy , Adolescent , Child , Child, Preschool , Epstein-Barr Virus Infections/immunology , Female , Herpesvirus 4, Human , Humans , Infant , Male , T-Lymphocyte Subsets/immunology
14.
Mol Carcinog ; 52(12): 946-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-22641235

ABSTRACT

Epstein-Barr virus (EBV) has been associated with several human malignancies including nasopharyngeal carcinoma (NPC). Reactivation of latent EBV has been considered to contribute to the carcinogenesis of NPC. Blocking the EBV lytic cycle has been shown effective in the treatment of EBV-associated diseases. We have searched for natural dietary compounds inhibiting EBV reactivation in NPC cells. Among them, sulforaphane (SFN) was found to be effective in the inhibition of EBV reactivation in latent EBV-positive NPC cells, NA and HA. SFN is a histone deacetylase (HDAC) inhibitor and has been recognized as an antioxidant and antitumor compound for chemoprevention. However, its antiviral effect is less well elucidated. In this study, after determination of the cytotoxicity of SFN on various epithelial cells, we showed that SFN treatment inhibits EBV reactivation, rather than induction, by detection of EBV lytic gene expression in EBV-positive NPC cells. We also determined that the number of cells supporting the EBV lytic cycle is decreased using immunofluorescence and flow cytometric analysis. Moreover, we have found that this inhibitory effect decreases virus production. To elucidate the inhibitory mechanism of SFN on the EBV lytic cycle, luciferase reporter assays were carried out on the Zta and Rta promoters. The results show that SFN inhibits transactivation activity of the EBV immediate-early gene Rta but not Zta. Together, our results suggest that SFN has the capability to inhibit EBV lytic cycle and the potential to be taken as a dietary compound for prevention of EBV reactivation.


Subject(s)
Herpesvirus 4, Human/drug effects , Isothiocyanates/pharmacology , Nasopharyngeal Neoplasms/virology , Antiviral Agents/pharmacology , Carcinoma , Dietary Supplements , Epithelial Cells/drug effects , Epithelial Cells/virology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/prevention & control , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Herpesvirus 4, Human/pathogenicity , Herpesvirus 4, Human/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Immediate-Early Proteins/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Promoter Regions, Genetic , Sulfoxides , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Cells, Cultured , Virus Activation/drug effects
15.
Integr Cancer Ther ; 12(3): 257-63, 2013 May.
Article in English | MEDLINE | ID: mdl-23089554

ABSTRACT

Epstein--Barr virus (EBV) is a human virus with oncogenic potentials that is implicated in various human diseases and malignancies. In this study, the modulator activity of the potent herbal extract drug thymoquinone on EBV was assessed in vitro. Thymoquinone was tested for cytotoxicity on human cells of lymphoblastoid cells, Raji Burkitt's lymphoma, DG-75 Burkitt's lymphoma, peripheral blood mononuclear cells, and periodontal ligament fibroblast. Apoptosis induction was analyzed via TUNEL assay and activity studies of caspase-3. The effect of thymoquinone on EBV gene expression was determined using real-time polymerase chain reaction. We report here, for the first time, a promising selective inhibitory affect of thymoquinone on EBV-infected B cell lines in vitro, compared with lower activity on EBV negative B cell line and very low toxicity on human peripheral blood mononuclear cells and periodontal ligament fibroblasts. Moreover, the drug was found to efficiently suppress the RNA expression of EBNA2, LMP1, and EBNA1 genes. Specifically, EBNA2 expression levels were the most affected indicating that this gene might have a major contribution to thymoquinone potency against EBV infected cells. Overall, our results suggest that thymoquinone has the potential to suppress the growth of EBV-infected B cells efficiently.


Subject(s)
B-Lymphocytes/drug effects , B-Lymphocytes/virology , Benzoquinones/pharmacology , Cell Survival/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/virology , Caspase 3/genetics , Cell Proliferation/drug effects , Cell Survival/genetics , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression/drug effects , Herbal Medicine/methods , Herpesvirus 4, Human/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Tumor Cells, Cultured , Viral Proteins/genetics
16.
Indian J Exp Biol ; 50(10): 708-13, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23214264

ABSTRACT

Pinus massoniana bark extract (PMBE) at a concentration of 60 microg/mL or more inhibits the expression of Epstein-Barr virus (EBV) lytic proteins, such as Rta, Zta, and EA-D. EBV lytic cycle was blocked by inhibiting the transcription of immediate-early genes. The results suggest that the PMBE has anti-EBV activity. Thus, the extract is potentially useful in preventing the lytic development of EBV in vitro.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Pinus/chemistry , Plant Bark/chemistry , Plant Extracts/pharmacology , Virus Replication/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Chromatography, High Pressure Liquid , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/metabolism , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/physiology , Humans , Immediate-Early Proteins/genetics , Transcription, Genetic , Tumor Cells, Cultured , Viral Proteins/metabolism
17.
J Gen Virol ; 92(Pt 8): 1760-1768, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21490247

ABSTRACT

Epstein-Barr virus (EBV) expresses two transcription factors, Rta and Zta, during the immediate-early stage of the lytic cycle to activate the transcription of early and late genes. This study finds that 0.31 mM protoapigenone from Thelypteris torresiana (Gaud.) inhibits the expression of EBV lytic proteins, including Rta, Zta, EA-D and VCA, in P3HR1 cells after lytic induction with 12-O-tetradecanoylphorbol-13-acetate and sodium butyrate. The lack of expression of EBV lytic proteins after protoapigenone treatment is attributed to the inhibition of the transactivation function of Zta because protoapigenone reduces the transactivation activity of Zta and Gal4-Zta, which contains the transactivation domain of Zta fused with Gal4. In contrast, protoapigenone does not affect the ability of Rta to activate a promoter that contains an Rta-response element, showing that the inhibition is unrelated to Rta. Furthermore, in a lactate dehydrogenase assay, protoapigenone is not toxic to P3HR1 cells at the concentrations that inhibit the function of Zta, showing that protoapigenone is valuable for studying the function of Zta and preventing EBV lytic proliferation.


Subject(s)
Cyclohexanones/pharmacology , Down-Regulation/drug effects , Ferns/chemistry , Flavones/pharmacology , Herpesvirus 4, Human/physiology , Plant Extracts/pharmacology , Cell Line , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/genetics , Humans , Viral Proteins/genetics , Viral Proteins/metabolism
18.
PLoS One ; 5(4): e10126, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20405039

ABSTRACT

BACKGROUND: Epstein-Barr Virus (EBV) latent infection is associated with several human malignancies and is a causal agent of lymphoproliferative diseases during immunosuppression. While inhibitors of herpesvirus DNA polymerases, like gancyclovir, reduce EBV lytic cycle infection, these treatments have limited efficacy for treating latent infection. EBNA1 is an EBV-encoded DNA-binding protein required for viral genome maintenance during latent infection. METHODOLOGY: Here, we report the identification of a new class of small molecules that inhibit EBNA1 DNA binding activity. These compounds were identified by virtual screening of 90,000 low molecular mass compounds using computational docking programs with the solved crystal structure of EBNA1. Four structurally related compounds were found to inhibit EBNA1-DNA binding in biochemical assays with purified EBNA1 protein. Compounds had a range of 20-100 microM inhibition of EBNA1 in fluorescence polarization assays and were further validated for inhibition using electrophoresis mobility shift assays. These compounds exhibited no significant inhibition of an unrelated DNA binding protein. Three of these compounds inhibited EBNA1 transcription activation function in cell-based assays and reduced EBV genome copy number when incubated with a Burkitt lymphoma cell line. CONCLUSIONS: These experiments provide a proof-of-principle that virtual screening can be used to identify specific inhibitors of EBNA1 that may have potential for treatment of EBV latent infection.


Subject(s)
Drug Discovery/methods , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Nuclear Antigens/drug effects , Transcription, Genetic/drug effects , Viral Proteins/antagonists & inhibitors , Burkitt Lymphoma , Cell Line, Tumor , Computer Simulation , DNA/metabolism , Drug Evaluation, Preclinical/methods , Herpesvirus 4, Human , Humans , Protein Binding/drug effects
19.
J Clin Invest ; 115(3): 591-3, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15765143

ABSTRACT

Numerous viruses cause latent infections in humans, and reactivation often results in pain and suffering. While vaccines for several of these viruses are available or currently being studied in clinical trials, and antiviral therapies have been successful in preventing or treating active infection, therapy to eradicate latent infection has lagged behind. A new study reported in this issue of the JCI shows that treatment of cells latently infected with Kaposi sarcoma-associated herpesvirus (KSHV) with glycyrrhizic acid, a component of licorice, reduces synthesis of a viral latency protein and induces apoptosis of infected cells. This finding suggests a novel way to interrupt latency.


Subject(s)
Anti-Infective Agents/therapeutic use , Glycyrrhiza/chemistry , Glycyrrhizic Acid/therapeutic use , Herpesviridae Infections/drug therapy , Herpesvirus 8, Human , Virus Latency , Animals , Anti-Infective Agents/pharmacology , Apoptosis/physiology , Epstein-Barr Virus Infections/drug therapy , Glycyrrhizic Acid/pharmacology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/physiology , Humans , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Virus Activation
20.
BMC Complement Altern Med ; 2: 11, 2002 Nov 20.
Article in English | MEDLINE | ID: mdl-12443533

ABSTRACT

BACKGROUND: We sought to assess whether GFS, a proprietary preparation of Tasmanian Undaria pinnatifida, has effects on healing or re-emergence of Herpetic infections, and additionally, to assess effects of GFS in vitro. Undaria is the most commonly eaten seaweed in Japan, and contains sulphated polyanions and other components with potential anti-viral activity. Herpes simplex virus type 1 (HSV-1) infections have lower reactivation rates and Herpes type 2 (HSV-2) infections have lower incidence in Japan than in the west. METHODS: Patients with active (15 subjects) or latent (6 subjects) Herpetic infections (HSV-1, 2, EBV, Zoster) were monitored for response to ingestion of GFS. GFS extract was tested in vitro for human T cell mitogenicity and anti-Herpes activity. RESULTS: Ingestion of GFS was associated with increased healing rates in patients with active infections. In addition, patients with latent infection remained asymptomatic whilst ingesting GFS. GFS extract inhibited Herpes viruses in vitro and was mitogenic to human T cells in vitro. CONCLUSIONS: Ingestion of GFS has inhibitory effects on reactivation and is associated with increased rate of healing after Herpetic outbreaks. GFS extract potently inhibited Herpes virus in vitro, and had mitogenic effects on human T cells.


Subject(s)
Complex Mixtures/therapeutic use , Herpesviridae Infections/drug therapy , Undaria , Virus Activation/drug effects , Adolescent , Adult , Aged , Child , Complex Mixtures/pharmacology , Complex Mixtures/toxicity , Epstein-Barr Virus Infections/drug therapy , Female , Herpes Genitalis/drug therapy , Herpes Simplex/drug therapy , Herpes Zoster/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/physiology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Humans , In Vitro Techniques , Male , Middle Aged , Mitogens/pharmacology , T-Lymphocytes/drug effects , Virus Latency/drug effects , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL