Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
Viruses ; 10(5)2018 05 16.
Article in English | MEDLINE | ID: mdl-29772651

ABSTRACT

Feline immunodeficiency virus (FIV) is an important cat pathogen worldwide whose biological and pathophysiological properties resemble those of human immunodeficiency virus type 1 (HIV-1). Therefore, the study of FIV not only benefits its natural host but is also useful for the development of antiviral strategies directed against HIV-1 infections in humans. FIV assembly results from the multimerization of a single but complex viral polypeptide, the Gag precursor. In this review, we will first give an overview of the current knowledge of the proteins encoded by the FIV pol, env, rev, vif, and orf-A genes, and then we will describe and discuss in detail the critical roles that each of the FIV Gag domains plays in virion morphogenesis. Since retroviral assembly is an attractive target for therapeutic interventions, gaining a better understanding of this process is highly desirable.


Subject(s)
Gene Products, gag/chemistry , Gene Products, gag/metabolism , Immunodeficiency Virus, Feline/physiology , Virion/physiology , Virus Assembly , Amino Acid Sequence , Animals , Antigens, Viral/chemistry , Antigens, Viral/physiology , Immunodeficiency Virus, Feline/chemistry , Immunodeficiency Virus, Feline/genetics , Models, Molecular , Protein Conformation , Virion/metabolism
2.
Cytokine ; 107: 35-42, 2018 07.
Article in English | MEDLINE | ID: mdl-29175261

ABSTRACT

OBJECT: Th17 cytokines have been identified in several types of human cancers. In this pilot study, the expression of Th17 cytokines profiling in enteroviruses 71 (EV71) associated colorectal cancer (CRC) were explored. METHODS: 66 patients with CRC were enrolled in this study; immune- histochemical analyses were performed on cancerous tissues and adjacent non- cancerous tissues of the patients. Serum Th17 cytokines of CRC patients and healthy controls were measured using a Luminex 200 analyzer. RESULTS: Cancerous tissues had more positive EV71 antigen expression than adjacent non- cancerous tissues. In TNM II-III CRC, 59.9% of cancerous tissues were observed to be EV71 positive; on the contrary, 65.2% of the adjacent non- cancerous epithelium was EV71 negative. In TNM I CRC, all adjacent non- cancerous epithelium was virus negative, but in TNM IV, half of adjacent non- cancerous tissues were virus positive. Serum IL-10 were significantly higher in CRC patients than in healthy controls, and IL-10 concentrations in the EV71 positive group were higher than those of the EV71 negative group, with the highest IL-10 levels being observed in CRC patients with strong positive group (P < 0.05). Similar results were found for IL-21 and IL-23. IL-17 levels were higher in CRC patients than in healthy controls, there was no significant difference in IL-17 between the viral positive and viral negative groups (P > 0.05). CONCLUSION: Persistent existing EV71 viral antigens in intestinal tissues are positively associated with TNM III/IV CRC. EV71 latent infection recruits Th17 cells in the colorectal tumor site, stimulating Th17 cytokine production that closely associated with CRC carcinogenesis.


Subject(s)
Antigens, Viral/immunology , Colorectal Neoplasms/immunology , Cytokines/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Th17 Cells/immunology , Adult , Aged , Aged, 80 and over , Antigens, Viral/genetics , Antigens, Viral/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/virology , Cytokines/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Female , Host-Pathogen Interactions/immunology , Humans , Male , Middle Aged , Neoplasm Staging , Pilot Projects , Th17 Cells/metabolism
3.
PLoS Pathog ; 10(12): e1004554, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25503639

ABSTRACT

Coevolution of viruses and their hosts represents a dynamic molecular battle between the immune system and viral factors that mediate immune evasion. After the abandonment of smallpox vaccination, cowpox virus infections are an emerging zoonotic health threat, especially for immunocompromised patients. Here we delineate the mechanistic basis of how cowpox viral CPXV012 interferes with MHC class I antigen processing. This type II membrane protein inhibits the coreTAP complex at the step after peptide binding and peptide-induced conformational change, in blocking ATP binding and hydrolysis. Distinct from other immune evasion mechanisms, TAP inhibition is mediated by a short ER-lumenal fragment of CPXV012, which results from a frameshift in the cowpox virus genome. Tethered to the ER membrane, this fragment mimics a high ER-lumenal peptide concentration, thus provoking a trans-inhibition of antigen translocation as supply for MHC I loading. These findings illuminate the evolution of viral immune modulators and the basis of a fine-balanced regulation of antigen processing.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antigens, Viral/genetics , Cowpox virus/genetics , Frameshift Mutation/genetics , Genome, Viral/genetics , ATP-Binding Cassette Transporters/immunology , ATP-Binding Cassette Transporters/physiology , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Antigens, Viral/physiology , Endoplasmic Reticulum , Feedback, Physiological/physiology , HeLa Cells , Histocompatibility Antigens Class I/immunology , Humans , Molecular Sequence Data , Sf9 Cells
4.
Proc Natl Acad Sci U S A ; 111(32): 11816-21, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071216

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects tumor cells and persists as a multiple-copy, extrachromosomal, circular episome. To persist, the viral genome must replicate with each cell cycle. The KSHV latency-associated nuclear antigen (LANA) mediates viral DNA replication and persistence, but little is known regarding the underlying mechanisms. We find that LANA recruits replication factor C (RFC), the DNA polymerase clamp [proliferating cell nuclear antigen (PCNA)] loader, to drive DNA replication efficiently. Mutated LANA lacking RFC interaction was deficient for LANA-mediated DNA replication and episome persistence. RFC depletion had a negative impact on LANA's ability to replicate and maintain viral DNA in cells containing artificial KSHV episomes or in infected cells, leading to loss of virus. LANA substantially increased PCNA loading onto DNA in vitro and recruited RFC and PCNA to KSHV DNA in cells. These findings suggest that PCNA loading is a rate-limiting step in DNA replication that is incompatible with viral survival. LANA enhancement of PCNA loading permits efficient virus replication and persistence, revealing a previously unidentified mechanism for KSHV latency.


Subject(s)
Antigens, Viral/physiology , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/pathogenicity , Nuclear Proteins/physiology , Replication Protein C/physiology , Virus Replication/physiology , Cell Line, Tumor , DNA Replication/physiology , DNA, Viral/biosynthesis , DNA, Viral/genetics , DNA-Directed DNA Polymerase/physiology , Gene Knockdown Techniques , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Humans , Proliferating Cell Nuclear Antigen/physiology , Replication Protein C/antagonists & inhibitors , Replication Protein C/genetics , Sarcoma, Kaposi/physiopathology , Sarcoma, Kaposi/virology , Virus Latency/physiology
5.
Nat Commun ; 5: 4394, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25008037

ABSTRACT

Binding of antibodies to their cognate antigens is fundamental for adaptive immunity. Molecular engineering of antibodies for therapeutic and diagnostic purposes emerges to be one of the major technologies in combating many human diseases. Despite its importance, a detailed description of the nanomechanical process of antibody-antigen binding and dissociation on the molecular level is lacking. Here we utilize high-speed atomic force microscopy to examine the dynamics of antibody recognition and uncover a principle; antibodies do not remain stationary on surfaces of regularly spaced epitopes; they rather exhibit 'bipedal' stochastic walking. As monovalent Fab fragments do not move, steric strain is identified as the origin of short-lived bivalent binding. Walking antibodies gather in transient clusters that might serve as docking sites for the complement system and/or phagocytes. Our findings could inspire the rational design of antibodies and multivalent receptors to exploit/inhibit steric strain-induced dynamic effects.


Subject(s)
Antigen-Antibody Complex/physiology , Antigens, Bacterial/physiology , Antigens, Viral/physiology , Epitopes/physiology , Immunoglobulin G/physiology , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/physiology , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/physiology , Epitopes/chemistry , Epitopes/immunology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Microscopy, Atomic Force , Protein Binding/physiology , Stochastic Processes
6.
J Immunol ; 193(3): 1258-67, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973451

ABSTRACT

Hemorrhagic manifestations occur frequently accompanying a wide range of dengue disease syndromes. Much work has focused on the contribution of immune factors to the pathogenesis of hemorrhage, but how dengue virus (DENV) participates in the pathogenic process has never been explored. Although there is no consensus that apoptosis is the basis of vascular permeability in human dengue infections, we showed in dengue hemorrhage mouse model that endothelial cell apoptosis is important to hemorrhage development in mice. To explore the molecular basis of the contribution of DENV to endothelial cell death, we show in this study that DENV protease interacts with cellular IκBα and IκBß and cleaves them. By inducing IκBα and IκBß cleavage and IκB kinase activation, DENV protease activates NF-κB, which results in endothelial cell death. Intradermal inoculation of DENV protease packaged in adenovirus-associated virus-9 induces endothelial cell death and dermal hemorrhage in mice. Although the H51 activity site is not involved in the interaction between DENV protease and IκB-α/ß, the enzymatic activity is critical to the ability of DENV protease to induce IκBα and IκBß cleavage and trigger hemorrhage development. Moreover, overexpression of IκBα or IκBß protects endothelial cells from DENV-induced apoptosis. In this study, we show that DENV protease participates in the pathogenesis of dengue hemorrhage and discover IκBα and IκBß to be the new cellular targets that are cleaved by DENV protease.


Subject(s)
Apoptosis/immunology , Dengue/immunology , Endothelium, Vascular/immunology , Hemorrhage/immunology , I-kappa B Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Serine Endopeptidases/metabolism , Animals , Antigens, Viral/metabolism , Antigens, Viral/physiology , Capillary Permeability/immunology , Cell Death/immunology , Cell Line , Dengue/enzymology , Dengue/pathology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , HEK293 Cells , Hemorrhage/pathology , Hemorrhage/virology , Humans , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Serine Endopeptidases/physiology
7.
Cancer Res ; 74(7): 2026-37, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24523441

ABSTRACT

Notch signaling has been implicated in the pathogenesis of Kaposi sarcoma. Kaposi sarcoma is an angioproliferative neoplasm that originates from Kaposi sarcoma-associated herpesvirus (KSHV) infection. Previously, we showed that the KSHV LANA protein can stabilize intracellular Notch in KSHV-infected tumor cells and promote cell proliferation. However, whether Notch signaling functions in pathologic angiogenesis of Kaposi sarcoma remains largely unknown. Hey1, an essential downstream effector of the Notch signaling pathway, has been demonstrated to play a fundamental role in vascular development. In the present study, we performed whole transcriptome, paired-end sequencing on three patient-matched clinical Kaposi sarcoma specimens and their corresponding adjacent stroma samples, with an average depth of 42 million reads per sample. Dll4, Hey1, and HeyL displayed significant upregulation in Kaposi sarcoma. Further verification based on immunohistochemistry analysis demonstrated that Hey1 was indeed highly expressed in Kaposi sarcoma lesions. Using the Matrigel plug assay, we showed that downregulation of Hey1 and γ-secretase inhibitor treatment caused dramatic reduction in the formation of new blood vessels in mice. Interestingly, LANA was responsible for the elevated level of Hey1 through inhibition of its degradation. Importantly, Hey1 stabilized by LANA promoted the neoplastic vasculature. Taken together, our data suggest that hijacking of the proangiogenic property of Hey1 by LANA is an important strategy utilized by KSHV to achieve pathologic angiogenesis and that Hey1 is a potential therapeutic target in Kaposi sarcoma.


Subject(s)
Antigens, Viral/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Neovascularization, Pathologic/etiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Sarcoma, Kaposi/blood supply , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, Notch/physiology , Repressor Proteins/analysis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Sarcoma, Kaposi/therapy
8.
Oncogene ; 32(32): 3676-85, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-22964633

ABSTRACT

Kaposi's Sarcoma Herpesvirus (KSHV) is the causative agent of Kaposi's Sarcoma (KS) and two rare lymphoproliferative disorders, primary effusion lymphoma (PEL) and the plasmablastic variant of multicentric Castleman's disease (MCD). The KSHV latency-associated nuclear antigen-1 (LANA), required for the replication and maintenance of latent viral episomal DNA, is involved in the transcriptional regulation of viral and cellular genes and interacts with different cellular proteins, including the tumour suppressor p53. Here, we report that LANA also recruits the p53-related nuclear transcription factor p73, which influences cellular processes like DNA damage response, cell cycle progression and apoptosis. Both the full-length isoform TAp73α, as well as its dominant negative regulator ΔNp73α, interact with LANA. LANA affects TAp73α stability and sub-nuclear localisation, as well as TAp73α-mediated transcriptional activation of target genes. We observed that the small-molecule inhibitor Nutlin-3, which disrupts the interaction of p53 and p73 with MDM2, induces apoptotic cell death in p53 wild-type, as well as p53-mutant PEL cell lines, suggesting a possible involvement of p73. The small-molecule RETRA, which activates p73 in the context of mutant p53, leads to the induction of apoptosis in p53-mutant PEL cell lines. RNAi-mediated knockdown of p73 confirmed that these effects depend on the presence of the p73 protein. Furthermore, both Nutlin-3 and RETRA disrupt the LANA-p73 interaction in different PEL cell lines. These results suggest that LANA modulates p73 function and that the LANA-p73 interaction may represent a therapeutic target to interfere with the survival of latently KSHV-infected cells.


Subject(s)
Antigens, Viral/physiology , DNA-Binding Proteins/physiology , Lymphoma, Primary Effusion/pathology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , Antigens, Viral/chemistry , Apoptosis , Binding Sites , Catechols/pharmacology , Cell Survival , DNA Damage , HEK293 Cells , HeLa Cells , Humans , Imidazoles/pharmacology , Lymphoma, Primary Effusion/drug therapy , Nuclear Proteins/chemistry , Piperazines/pharmacology , Thiazoles/pharmacology , Tumor Protein p73 , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/physiology
9.
PLoS Pathog ; 8(9): e1002906, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22969427

ABSTRACT

The Latency-Associated Nuclear Antigen (LANA), encoded by ORF73, is a conserved gene among the γ2-herpesviruses (rhadinoviruses). The Kaposi's Sarcoma-Associated Herpesvirus (KSHV) LANA is consistently expressed in KSHV-associated malignancies. In the case of the rodent γ2-herpesvirus, murine gammaherpesvirus 68 (MHV68), the LANA homolog (mLANA) is required for efficient virus replication, reactivation from latency and immortalization of murine fetal liver-derived B cells. To gain insights into mLANA function(s), knowing that KSHV LANA binds DNA and can modulate transcription of a variety of promoters, we sought out and identified a mLANA-responsive promoter which maps to the terminal repeat (TR) of MHV68. Notably, mLANA strongly repressed activity from this promoter. We extended these analyses to demonstrate direct, sequence-specific binding of recombinant mLANA to TR DNA by DNase I footprinting. To assess whether the DNA-binding and/or transcription modulating function is important in the known mLANA phenotypes, we generated an unbiased library of mLANA point mutants using error-prone PCR, and screened a large panel of mutants for repression of the mLANA-responsive promoter to identify loss of function mutants. Notably, among the mutant mLANA proteins recovered, many of the mutations are in a predicted EBNA-1-like DNA-binding domain. Consistent with this prediction, those tested displayed loss of DNA binding activity. We engineered six of these mLANA mutants into the MHV68 genome and tested the resulting mutant viruses for: (i) replication fitness; (ii) efficiency of latency establishment; and (iii) reactivation from latency. Interestingly, each of these mLANA-mutant viruses exhibited phenotypes similar to the mLANA-null mutant virus, indicating that DNA-binding is critical for mLANA function.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/physiology , DNA/metabolism , Gammaherpesvirinae/genetics , Mutagenesis/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Protein Interaction Domains and Motifs/genetics , Animals , Antigens, Viral/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Viral , HEK293 Cells , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/chemistry , Protein Interaction Domains and Motifs/physiology , Repression, Psychology , Terminal Repeat Sequences/genetics
10.
J Virol ; 86(18): 9683-95, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740392

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes sustained latent persistence in susceptible cells. This is dependent on the latency-associated nuclear antigen (LANA). Understanding how LANA transcription is regulated thus aids our fundamental understanding of KSHV biology. Two hundred ninety-four base pairs are sufficient to regulate LANA transcription in response to the viral RTA protein and RBPjκ. The same region controls K14/viral G-protein-coupled receptor (vGPCR) transcription in the opposite direction. We used a quantitative analysis in conjunction with specific nucleotide substitutions and defined gain-of-function and loss-of-function RTA mutants to dissect this region. We used a bidirectional reporter driving red and green luciferase to study the LANApi and K14p promoters simultaneously. This established that LANApi/K14p functions as a canonical bidirectional promoter. Both were TATA dependent. K14p was favored by ∼50-fold in this context. Eliminating the distal LANApi TATA box increased maximal output and lowered the induction threshold (T) of K14p even further. Two RBPjκ binding sites were independently required; however, at high concentrations of RTA, direct interactions with an RTA-responsive element (RRE) could complement the loss of one RBPjκ binding site. Intracellular Notch (ICN) was no longer able to activate RBPjκ in the viral context. This suggests a model whereby KSHV alters ICN-RBPjκ gene regulation. When the architecture of this pair of head-to-head RBPjκ binding sites is changed, the sites now respond exclusively to the viral transactivator RTA and no longer to the host mediator ICN.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/physiology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Virus/physiology , Base Sequence , Cell Line , Genome, Viral , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Models, Biological , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Viral/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Virus/genetics , Trans-Activators/genetics , Trans-Activators/physiology , Virus Latency/genetics , Virus Latency/physiology
11.
Blood ; 119(15): 3638-45, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22374699

ABSTRACT

The coagulation system provides physiologic host defense, but it can also be exploited by pathogens for infection. On the HSV1 surface, host-cell-derived tissue factor (TF) and virus-encoded glycoprotein C (gC) can stimulate protease activated receptor 1 (PAR1)-enhanced infection by triggering thrombin production. Using novel engineered HSV1 variants deficient in either TF and/or gC, in the present study, we show that activated coagulation factors X (FXa) or VII (FVIIa) directly affect HSV1 infection of human umbilical vein endothelial cells in a manner that is dependent on viral TF and gC. The combination of FXa and FVIIa maximally enhanced infection for TF(+)/gC(+) HSV1 and receptor desensitization and Ab inhibition demonstrated that both proteases act on PAR2. Inhibitory TF Abs showed that the required TF source was viral. Individually, TF or gC partly enhanced the effect of FXa, but not FVIIa, revealing gC as a novel PAR2 cofactor for FVIIa. In sharp contrast, thrombin enhanced infection via PAR1 independently of viral TF and gC. Thrombin combined with FXa/FVIIa enhanced infection, suggesting that PAR1 and PAR2 are independently involved in virus propagation. These results show that HSV1 surface cofactors promote cellular PAR2-mediated infection, indicating a novel mode by which pathogens exploit the initiation phase of the host hemostatic system.


Subject(s)
Herpes Simplex/pathology , Receptor, PAR-2/metabolism , Thromboplastin/physiology , Viral Envelope Proteins/physiology , Antigens, Surface/metabolism , Antigens, Viral/metabolism , Antigens, Viral/physiology , Blood Coagulation Factors/metabolism , Cells, Cultured , Coenzymes/metabolism , Coenzymes/physiology , Disease Progression , Herpes Simplex/enzymology , Herpes Simplex/metabolism , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/virology , Humans , Signal Transduction , Thromboplastin/metabolism , Thromboplastin/pharmacology , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/pharmacology
12.
Semin Ophthalmol ; 26(4-5): 246-50, 2011.
Article in English | MEDLINE | ID: mdl-21958170

ABSTRACT

Recurrent herpes stromal keratitis (HSK) is one of the leading causes of blindness in the developed world. Cyokines characteristic of Th1 cells (in particular IFN-γ and IL-2) have been shown to dominate in HSK in addition to mechanisms by nonspecific, antigen-independent effector cells such as neutrophils, basophils, and monocytes. More recently, the migration and maturation of dendritic cells (DC) within the corneal stroma of patients with HSK have been recognized as contributors to recurrent disease, suggesting a role for delayed type hypersensitivity (DTH) in the immunopathogenesis of HSK. The role of DC and DTH in recurrent HSK has not been studied extensively and experimental models of recurrent HSK focusing on DTH as the pathogenesis and viral particles as the triggering antigen may contribute to better understanding of the disease.


Subject(s)
Hypersensitivity, Delayed/immunology , Keratitis, Herpetic/immunology , Animals , Antigens, Viral/physiology , Dendritic Cells/physiology , Herpesvirus 1, Human/physiology , Humans , Recurrence , Th1 Cells/physiology , Virus Activation
13.
Clin Cancer Res ; 17(10): 3056-63, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21372216

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous human γ-herpesvirus that establishes a life-long asymptomatic infection in immunocompetent hosts. It is also found to be frequently associated with a broad spectrum of B-cell lymphomas predominantly seen in immunodeficient patients. Despite many resemblances, these EBV-linked lymphoproliferative disorders display heterogeneity at the clinical and the molecular level. Moreover, EBV-associated lymphoproliferative diseases differ in their differential expression patterns of the EBV-encoded latent antigens, which are directly related to their interactions with the host. EBV-driven primary B-cell immortalization is linked to the cooperative functions of these latent proteins, which are critical for perturbing many important cell-signaling pathways maintaining B-cell proliferation. Additionally, it is used as a surrogate model to explore the underlying mechanisms involved in the development of B-cell neoplasms. Recent discoveries have revealed that a number of sophisticated mechanisms are exploited by EBV during cancer progression. This finding will be instrumental in the design of novel approaches for therapeutic interventions against EBV-associated B-cell lymphomas. This review limits the discussion to the biology and pathogenesis of EBV-associated B-cell lymphomas and the related clinical implications.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/etiology , Antigens, Viral/genetics , Antigens, Viral/physiology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Viral/genetics , Humans , Lymphoma, B-Cell/complications , Lymphoma, B-Cell/virology , Models, Biological , Prognosis , Virus Latency/genetics , Virus Latency/physiology
14.
PLoS Pathog ; 7(2): e1001275, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347341

ABSTRACT

EBNA3C, one of the Epstein-Barr virus (EBV)-encoded latent antigens, is essential for primary B-cell transformation. Cyclin D1, a key regulator of G1 to S phase progression, is tightly associated and aberrantly expressed in numerous human cancers. Previously, EBNA3C was shown to bind to Cyclin D1 in vitro along with Cyclin A and Cyclin E. In the present study, we provide evidence which demonstrates that EBNA3C forms a complex with Cyclin D1 in human cells. Detailed mapping experiments show that a small N-terminal region which lies between amino acids 130-160 of EBNA3C binds to two different sites of Cyclin D1- the N-terminal pRb binding domain (residues 1-50), and C-terminal domain (residues 171-240), known to regulate Cyclin D1 stability. Cyclin D1 is short-lived and ubiquitin-mediated proteasomal degradation has been targeted as a means of therapeutic intervention. Here, we show that EBNA3C stabilizes Cyclin D1 through inhibition of its poly-ubiquitination, and also increases its nuclear localization by blocking GSK3ß activity. We further show that EBNA3C enhances the kinase activity of Cyclin D1/CDK6 which enables subsequent ubiquitination and degradation of pRb. EBNA3C together with Cyclin D1-CDK6 complex also efficiently nullifies the inhibitory effect of pRb on cell growth. Moreover, an sh-RNA based strategy for knock-down of both cyclin D1 and EBNA3C genes in EBV transformed lymphoblastoid cell lines (LCLs) shows a significant reduction in cell-growth. Based on these results, we propose that EBNA3C can stabilize as well as enhance the functional activity of Cyclin D1 thereby facilitating the G1-S transition in EBV transformed lymphoblastoid cell lines.


Subject(s)
Antigens, Viral/physiology , Cyclin D1/metabolism , Cyclin D1/physiology , G1 Phase/genetics , S Phase/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cell Transformation, Viral/genetics , Cells, Cultured , Cyclin D1/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens , Gene Expression Regulation , Herpesvirus 4, Human/physiology , Humans , Protein Binding , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Protein Stability , Protein Structure, Tertiary/physiology , Ubiquitination , Up-Regulation/genetics
15.
J Immunol ; 186(3): 1828-39, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187441

ABSTRACT

Dendritic cell (DC)-based immunotherapy has potential for treating infections and malignant tumors, but the functional capacity of DC must be assessed in detail, especially maturation and Ag-specific CTL priming. Recent reports suggest that DC that are provided with continuous maturation signals in vivo after transfer into patients are required to elicit the full DC functions. We demonstrate in this study that the rSendai virus vector (SeV) is a novel and ideal stimulant, providing DC with a continuous maturation signal via viral RNA synthesis in the cytosol, resulting in full maturation of monocyte-derived DC(s). Both RIG-I-dependent cytokine production and CD4 T cell responses to SeV-derived helper Ags are indispensable for overcoming regulatory T cell suppression to prime melanoma Ag recognized by T cell-1-specific CTL in the regulatory T cell abundant setting. DC stimulated via cytokine receptors, or TLRs, do not show these functional features. Therefore, SeV-infected DC have the potential for DC-directed immunotherapy.


Subject(s)
Cell Differentiation/immunology , Cytosol/immunology , DEAD-box RNA Helicases/physiology , Dendritic Cells/immunology , RNA, Viral/biosynthesis , Sendai virus/immunology , Signal Transduction/immunology , Virus Replication/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Line, Transformed , Coculture Techniques , Cytosol/metabolism , Cytosol/virology , Cytotoxicity Tests, Immunologic , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dendritic Cells/pathology , Dendritic Cells/virology , Epitopes, T-Lymphocyte/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , RNA, Viral/genetics , Receptors, Immunologic , Sendai virus/genetics , Signal Transduction/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Virus Replication/genetics
16.
PLoS One ; 5(11): e13979, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21085583

ABSTRACT

Epstein-Barr virus (EBV) is able to drive the transformation of B-cells, resulting in the generation of lymphoblastoid cell lines (LCLs) in vitro. EBV nuclear proteins EBNA3A and EBNA3C are necessary for efficient transformation, while EBNA3B is dispensable. We describe a transcriptome analysis of BL31 cells infected with a series of EBNA3-knockout EBVs, including one deleted for all three EBNA3 genes. Using Affymetrix Exon 1.0 ST microarrays analysed with the MMBGX algorithm, we have identified over 1000 genes whose regulation by EBV requires one of the EBNA3s. Remarkably, a third of the genes identified require more than one EBNA3 for their regulation, predominantly EBNA3C co-operating with either EBNA3B, EBNA3A or both. The microarray was validated by real-time PCR, while ChIP analysis of a selection of co-operatively repressed promoters indicates a role for polycomb group complexes. Targets include genes involved in apoptosis, cell migration and B-cell differentiation, and show a highly significant but subtle alteration in genes involved in mitosis. In order to assess the relevance of the BL31 system to LCLs, we analysed the transcriptome of a set of EBNA3B knockout (3BKO) LCLs. Around a third of the genes whose expression level in LCLs was altered in the absence of EBNA3B were also altered in 3BKO-BL31 cell lines.Among these are TERT and TCL1A, implying that EBV-induced changes in the expression of these genes are not required for B-cell transformation. We also identify 26 genes that require both EBNA3A and EBNA3B for their regulation in LCLs. Together, this shows the complexity of the interaction between EBV and its host, whereby multiple EBNA3 proteins co-operate to modulate the behaviour of the host cell.


Subject(s)
Antigens, Viral/genetics , Chromatin/metabolism , Gene Expression Profiling , Antigens, Viral/physiology , Cell Line, Tumor , Cluster Analysis , Epigenomics , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/physiology , Gene Knockout Techniques , HEK293 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/genetics , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
17.
Vet Res Commun ; 34 Suppl 1: S63-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20512626

ABSTRACT

Flow cytometry is a useful tool to determine both the phenotype and function of immune cells during vaccination and viral infection. Since cytokine release is a hallmark of cell activation, detection of intracellular interferon (IFN)gamma in cytotoxic CD8alpha+ cells under physiological conditions and after Aujeszky's Disease Virus (ADV)- or Porcine Circovirus type 2 (PCV2)-swine interaction was carried out. Blood samples were collected from healthy 10-month-old pigs, ADV- or PCV2-vaccinated pigs, and PCV2-infected pigs; the levels of total IFNgamma+ and CD8alpha+IFNgamma+ cells were evaluated after PMA-ionomycin and virus-specific in vitro stimulation. High CD8alpha+IFNgamma+ cell levels were detected in adult pigs, whereas lower virus-specific cell fractions were observed after ADV or PCV2 vaccination as well as after PCV2 natural infection due to restricted activation. Such results support the use of cytokine intracellular staining to monitor virus-specific cell-mediated immunity.


Subject(s)
Antigens, Viral/physiology , Circovirus/physiology , Herpesvirus 1, Suid/physiology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/virology , Animals , Cells, Cultured , Circovirus/classification , Flow Cytometry , Interferon-gamma/genetics , Ionomycin , Pseudorabies/prevention & control , Swine , Viral Vaccines/immunology
18.
Virology ; 403(1): 85-91, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20447672

ABSTRACT

Site-directed mutagenesis of residues in the BC loop (residues 329-333) of the envelope (E) protein domain III in a West Nile virus (WNV) infectious clone and in plasmids encoding recombinant WNV and dengue type 2 virus domain III proteins demonstrated a critical role for residues in this loop in the function and antigenicity of the E protein. This included a strict requirement for the tyrosine at residue 329 of WNV for virus viability and E domain III folding. The absence of an equivalent residue in this region of yellow fever group viruses and most tick-borne flavivirus suggests there is an evolutionary divergence in the molecular mechanisms of domain III folding employed by different flaviviruses.


Subject(s)
Antigens, Viral/physiology , Viral Envelope Proteins/physiology , Virus Attachment , West Nile virus/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Antigens, Viral/genetics , Antigens, Viral/immunology , Chlorocebus aethiops , Female , Humans , Mice , Microbial Viability , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutralization Tests , Protein Folding , Protein Structure, Tertiary , Sequence Alignment , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , West Nile virus/genetics , West Nile virus/immunology
19.
Vet Res ; 41(3): 36, 2010.
Article in English | MEDLINE | ID: mdl-20167197

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the causative agent of a highly contagious acute vesicular disease affecting cloven-hoofed animals, including cattle, sheep and pigs. The current vaccine induces a rapid humoral response, but the duration of the protective antibody response is variable, possibly associated with a variable specific CD4+ T cell response. We investigated the use of heat shock protein 70 (Hsp70) as a molecular chaperone to target viral antigen to the Major Histocompatibility Complex (MHC) class II pathway of antigen presenting cells and generate enhanced MHC II-restricted CD4+ T cell responses in cattle. Monocytes and CD4+ T cells from FMDV vaccinated cattle were stimulated in vitro with complexes of Hsp70 and FMDV peptide, or peptide alone. Hsp70 was found to consistently improve the presentation of a 25-mer FMDV peptide to CD4+ T cells, as measured by T cell proliferation. Complex formation was required for the enhanced effects and Hsp70 alone did not stimulate proliferation. This study provides further evidence that Hsp70:peptide complexes can enhance antigen-specific CD4+ T cell responses in vitro for an important pathogen of livestock.


Subject(s)
Antigens, Viral/physiology , Foot-and-Mouth Disease Virus/physiology , HSP70 Heat-Shock Proteins/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , Cattle , Cattle Diseases/prevention & control , Cattle Diseases/virology , Cells, Cultured , Foot-and-Mouth Disease/prevention & control , Viral Vaccines
20.
J Gen Virol ; 91(Pt 4): 829-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20089802

ABSTRACT

Viruses that establish lifelong latent infections must ensure that the viral genome is maintained within the latently infected cell throughout the life of the host, yet at the same time must also be capable of avoiding elimination by the immune surveillance system. Gammaherpesviruses, which include the human viruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, establish latent infections in lymphocytes. Infection of this dynamic host-cell population requires that the viruses have appropriate strategies for enabling the viral genome to persist while these cells go through rounds of mitosis, but at the same time must avoid detection by host CD8(+) cytotoxic T lymphocytes (CTLs). The majority of gammaherpesviruses studied have been found to encode a specific protein that is critical for maintenance of the viral genome within latently infected cells. This protein is termed the genome maintenance protein (GMP). Due to its vital role in long-term latency, this offers the immune system a crucial target for detection and elimination of virus-infected cells. GMPs from different gammaherpesviruses have evolved related strategies that allow the protein to be present within latently infected cells, but to remain effectively hidden from circulating CD8(+) CTLs. In this review, I will summarize the role of the GMPs and highlight the available data describing the immune-evasion properties of these proteins.


Subject(s)
Gammaherpesvirinae/immunology , Genome, Viral , Immune Evasion , Viral Proteins/physiology , Animals , Antigens, Viral/physiology , Epstein-Barr Virus Nuclear Antigens/immunology , Epstein-Barr Virus Nuclear Antigens/physiology , Gammaherpesvirinae/classification , Humans , Nuclear Proteins/physiology , T-Lymphocytes, Cytotoxic/immunology , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...