Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429336

ABSTRACT

Human herpesvirus 6B (HHV-6B) DNA is frequently detected in human samples. Diagnostic assays distinguishing HHV-6B reactivation from latency are limited. This has impaired strategies to diagnose and treat HHV-6B-associated diseases. We used RNA sequencing to characterize and compare the HHV-6B transcriptome in multiple sample types, including (i) whole blood from hematopoietic cell transplant (HCT) recipients with and without HHV-6B plasma viremia, (ii) tumor tissue samples from subjects with large B cell lymphoma infected with HHV-6B, (iii) lymphoblastoid cell lines (LCLs) from subjects with inherited chromosomally integrated HHV-6B or latent infection with HHV-6B, and (iv) HHV-6B Z29 infected SupT1 CD4+ T cells. We demonstrated substantial overlap in the HHV-6B transcriptome observed in in vivo and in vitro samples, although there was variability in the breadth and quantity of gene expression across samples. The HHV-6B viral polymerase gene U38 was the only HHV-6B transcript detected in all next-generation RNA sequencing (RNA-seq) data sets and was one of the most highly expressed genes. We developed a novel reverse transcription-PCR assay targeting HHV-6B U38, which identified U38 mRNA in all tested whole-blood samples from patients with concurrent HHV-6B viremia. No HHV-6B U38 transcripts were detected by RNA-seq or reverse transcription-real-time quantitative PCR (RT-qPCR) in whole-blood samples from subjects without HHV-6B plasma detection or from latently infected LCLs. A RT-qPCR assay for HHV-6B U38 may be useful to identify lytic HHV-6B infection in nonplasma samples and samples from individuals with inherited chromosomally integrated HHV-6B. This study also demonstrates the feasibility of transcriptomic analyses for HCT recipients.IMPORTANCE Human herpesvirus 6B (HHV-6B) is a DNA virus that infects most children within the first few years of life. After primary infection, HHV-6B persists as a chronic, latent infection in many cell types. Additionally, HHV-6B can integrate into germ line chromosomes, resulting in individuals with viral DNA in every nucleated cell. Given that PCR to detect viral DNA is the mainstay for diagnosing HHV-6B infection, the characteristics of HHV-6B infection complicate efforts to distinguish between latent and active viral infection, particularly in immunocompromised patients who have frequent HHV-6B reactivation. In this study, we used RNA sequencing to characterize the HHV-6B gene expression profile in multiple sample types, and our findings identified evidence-based targets for diagnostic tests that distinguish between latent and active viral infection.


Subject(s)
Herpesvirus 6, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Roseolovirus Infections/diagnosis , Transcriptome , Viral Proteins/genetics , Viremia/diagnosis , Virus Activation , Virus Latency , Adult , Aged , Biomarkers/analysis , Case-Control Studies , Cytokines/blood , DNA, Viral , Female , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/virology , Male , Middle Aged , Roseolovirus Infections/genetics , Roseolovirus Infections/virology , Viremia/genetics , Viremia/virology
2.
PLoS Pathog ; 14(12): e1007441, 2018 12.
Article in English | MEDLINE | ID: mdl-30557332

ABSTRACT

KSHV is endemic in Uganda and the HIV epidemic has dramatically increased the incidence of Kaposi sarcoma (KS). To investigate the role of KSHV in the development of KS, we obtained KS biopsies from ART-naïve, HIV-positive individuals in Uganda and analyzed the tumors using RNAseq to globally characterize the KSHV transcriptome. Phylogenetic analysis of ORF75 sequences from 23 tumors revealed 6 distinct genetic clusters with KSHV strains exhibiting M, N or P alleles. RNA reads mapping to specific unique coding sequence (UCDS) features were quantitated using a gene feature file previously developed to globally analyze and quantitate KSHV transcription in infected endothelial cells. A pattern of high level expression was detected in the KSHV latency region that was common to all KS tumors. The clear majority of transcription was derived from the downstream latency transcript promoter P3(LTd) flanking ORF72, with little evidence of transcription from the P1(LTc) latency promoter, which is constitutive in KSHV-infected lymphomas and tissue-culture cells. RNAseq data provided evidence of alternate P3(LTd) transcript editing, splicing and termination resulting in multiple gene products, with 90% of the P3(LTd) transcripts spliced to release the intronic source of the microRNAs K1-9 and 11. The spliced transcripts encode a regulatory uORF upstream of Kaposin A with alterations in intervening repeat sequences yielding novel or deleted Kaposin B/C-like sequences. Hierarchical clustering and PCA analysis of KSHV transcripts revealed three clusters of tumors with different latent and lytic gene expression profiles. Paradoxically, tumors with a latent phenotype had high levels of total KSHV transcription, while tumors with a lytic phenotype had low levels of total KSHV transcription. Morphologically distinct KS tumors from the same individual showed similar KSHV gene expression profiles suggesting that the tumor microenvironment and host response play important roles in the activation level of KSHV within the infected tumor cells.


Subject(s)
Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/genetics , Transcriptome/genetics , Virus Latency/genetics , Gene Expression Profiling/methods , Genes, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Promoter Regions, Genetic/genetics , Sarcoma, Kaposi/virology , Sequence Analysis, RNA , Uganda
3.
PLoS One ; 13(11): e0205632, 2018.
Article in English | MEDLINE | ID: mdl-30444879

ABSTRACT

Macaque RFHV and LCV are close homologs of human KSHV and EBV, respectively. No experimental model of RFHV has been developed due to the lack of a source of culturable infectious virus. Screening of macaques at the Washington National Primate Research Center detected RFHV in saliva of SIV-infected macaques from previous vaccine studies. A pilot experimental infection of two naïve juvenile pig-tailed macaques was initiated by inoculation of saliva from SIV-infected pig-tailed and cynomolgus macaque donors, which contained high levels of DNA (> 10(6) genomes/ml) of the respective species-specific RFHV strain. Both juvenile recipients developed SIV and RFHV infections with RFHV DNA detected transiently in saliva and/or PBMC around week 16 post-infection. One juvenile macaque was infected with the homologous RFHVMn from whole saliva of a pig-tailed donor, which had been inoculated into the cheek pouch. This animal became immunosuppressed, developing simian AIDS and was euthanized 23 weeks after inoculation. The levels of RFHV DNA in saliva and PBMC remained below the level of detection after week 17, showing no reactivation of the RFHVMn infection during the rapid development of AIDS. The other juvenile macaque was infected with the heterologous RFHVMf from i.v. inoculation of purified virions from saliva of a cynomolgus donor. The juvenile recipient remained immunocompetent, developing high levels of persistent anti-RFHV and -SIV antibodies. After the initial presence of RFHVMf DNA in saliva and PBMC decreased to undetectable levels by week 19, all attempts to reactivate the infection through additional inoculations, experimental infection with purified SRV-2 or SIV, or immunosuppressive treatments with cyclosporine or dexamethasone were unsuccessful. An heterologous LCV transmission was also detected in this recipient, characterized by continual high levels of LCVMf DNA from the cynomolgus donor in both saliva (> 10(6) genomes/ml) and PBMC (> 10(4) genomes/million cells), coupled with high levels of anti-LCV antibodies. The macaque was sacrificed 209 weeks after the initial inoculation. Low levels of LCVMf DNA were detected in salivary glands, tonsils and other lymphoid organs, while RFHVMf DNA was below the level of detection. These results show successful co-transmission of RFHV and LCV from saliva and demonstrate differential lytic activation of the different gammaherpesvirus lineages due to presumed differences in biology and tropism and control by the host immune system. Although this initial pilot transmission study utilized only two macaques, it provides the first evidence for experimental transmission of the macaque homolog of KSHV, setting the stage for larger transmission studies to examine the differential activation of rhadinovirus and lymphocryptovirus infections and the pathological effects of immunosuppression.


Subject(s)
Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Simian Immunodeficiency Virus/genetics , Viral Proteins/genetics , Animals , Epstein-Barr Virus Infections/transmission , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/pathogenicity , Humans , Leukocytes, Mononuclear/virology , Macaca mulatta/virology , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/pathogenicity , Washington
4.
Virology ; 519: 106-120, 2018 06.
Article in English | MEDLINE | ID: mdl-29689462

ABSTRACT

We developed a set of rabbit antisera to characterize infections by the macaque RV2 rhadinovirus homologs of KSHV. We analyzed tissues from rhesus and pig-tailed macaques naturally infected with rhesus rhadinovirus (RRV) or Macaca nemestrina rhadinovirus 2 (MneRV2). Our study demonstrates that RV2 rhadinoviruses have a tropism for epithelial cells, lymphocytes and gonadal germ cells in vivo. We observed latent infections in both undifferentiated and differentiated epithelial cells with expression of the latency marker, LANA. Expression of the early (ORF59) and late (glycoprotein B) lytic markers were detected in highly differentiated cells in epithelial ducts in oral, renal, dermal and gastric mucosal tissue as well as differentiated germ cells in male and female gonads. Our data provides evidence that epithelial and germ cell differentiation in vivo induces rhadinovirus reactivation and suggests that infected epithelial and germ cells play a role in transmission and dissemination of RV2 rhadinovirus infections in vivo.


Subject(s)
Epithelial Cells/virology , Germ Cells/virology , Germinal Center/cytology , Herpesviridae Infections/virology , Herpesvirus 8, Human/physiology , Lymphocytes/virology , Rhadinovirus/physiology , Animals , Antigens, Viral/genetics , Gastrointestinal Tract/virology , Germinal Center/immunology , Germinal Center/virology , Gonads/virology , Herpesvirus 8, Human/genetics , Immunity, Innate , Macaca mulatta , Macaca nemestrina , Nuclear Proteins/genetics , Rabbits , Rhadinovirus/genetics , Sequence Homology , Skin/cytology , Skin/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Tropism , Virus Latency
5.
PLoS One ; 13(2): e0192659, 2018.
Article in English | MEDLINE | ID: mdl-29425228

ABSTRACT

Kaposi's sarcoma herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS). Both KSHV and HIV infections are endemic in Uganda, where KS is among the most common cancers in HIV-infected individuals. Recent studies examined the use of small RNAs as biomarkers of disease, including microRNAs (miRNAs), with viral and tumor-derived miRNAs being detected in exosomes from individuals with KSHV-associated malignancies. In the current study, the host and viral extracellular mature miRNA expression profiles were analyzed in blood of KS-negative individuals in Uganda, comparing those with or without KSHV detectable from the oropharynx. We observed increased levels of cellular oncogenic miRNAs and decreased levels of tumor-suppressor miRNAs in plasma of infected individuals exhibiting oral KSHV shedding. These changes in host oncomiRs were exacerbated in people co-infected with HIV, and partially reversed after 2 years of anti-retroviral therapy. We also detected KSHV miRNAs in plasma of KSHV infected individuals and determined that their expression levels correlated with KSHV plasma viremia. Deep sequencing revealed an expected profile of small cellular RNAs in plasma, with miRNAs constituting the major RNA biotype. In contrast, the composition of small RNAs in exosomes was highly atypical with high levels of YRNA and low levels of miRNAs. Mass spectrometry analysis of the exosomes revealed eleven different peptides derived from the malaria parasite, Plasmodium falciparum, and small RNA sequencing confirmed widespread plasmodium co-infections in the Ugandan cohorts. Proteome analysis indicated an exosomal protein profile consistent with erythrocyte and keratinocyte origins for the plasma exosomes. A strong correlation was observed between the abundance of Plasmodium proteins and cellular markers of malaria. As Plasmodium falciparum is an endemic pathogen in Uganda, our study shows that co-infection with other pathogens, such as KSHV, can severely impact the small RNA repertoire, complicating the use of exosome miRNAs as biomarkers of disease.


Subject(s)
Gene Expression Profiling , Herpesvirus 8, Human/physiology , Malaria, Falciparum/virology , MicroRNAs/genetics , Plasmodium falciparum/isolation & purification , Viremia , Virus Shedding , High-Throughput Nucleotide Sequencing , Humans
6.
J Virol ; 91(24)2017 12 15.
Article in English | MEDLINE | ID: mdl-28978712

ABSTRACT

The latency-associated nuclear antigen (LANA) of the Kaposi's sarcoma-associated herpesvirus (KSHV) performs a variety of functions to establish and maintain KSHV latency. During latency, LANA localizes to discrete punctate spots in the nucleus, where it tethers viral episomes to cellular chromatin and interacts with nuclear components to regulate cellular and viral gene expression. Using highly sensitive tyramide signal amplification, we determined that LANA localizes to the cytoplasm in different cell types undergoing the lytic cycle of replication after de novo primary infection and after spontaneous, tetradecanoyl phorbol acetate-, or open reading frame 50 (ORF50)/replication transactivator (RTA)-induced activation. We confirmed the presence of cytoplasmic LANA in a subset of cells in lytically active multicentric Castleman disease lesions. The induction of cellular migration by scratch-wounding confluent cell cultures, culturing under subconfluent conditions, or induction of cell differentiation in primary cultures upregulated the number of cells permissive for primary lytic KSHV infection. The induction of lytic replication was characterized by high-level expression of cytoplasmic LANA and nuclear ORF59, a marker of lytic replication. Subcellular fractionation studies revealed the presence of multiple isoforms of LANA in the cytoplasm of ORF50/RTA-activated Vero cells undergoing primary infection. Mass spectrometry analysis demonstrated that cytoplasmic LANA isoforms were full length, containing the N-terminal nuclear localization signal. These results suggest that trafficking of LANA to different subcellular locations is a regulated phenomenon, which allows LANA to interact with cellular components in different compartments during both the latent and the replicative stages of the KSHV life cycle.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) causes AIDS-related malignancies, including lymphomas and Kaposi's sarcoma. KSHV establishes lifelong infections using its latency-associated nuclear antigen (LANA). During latency, LANA localizes to the nucleus, where it connects viral and cellular DNA complexes and regulates gene expression, allowing the virus to maintain long-term infections. Our research shows that intact LANA traffics to the cytoplasm of cells undergoing permissive lytic infections and latently infected cells in which the virus is induced to replicate. This suggests that LANA plays important roles in the cytoplasm and nuclear compartments of the cell during different stages of the KSHV life cycle. Determining cytoplasmic function and mechanism for regulation of the nuclear localization of LANA will enhance our understanding of the biology of this virus, leading to therapeutic approaches to eliminate infection and block its pathological effects.


Subject(s)
Antigens, Viral/metabolism , Cytoplasm/virology , Herpesvirus 8, Human/physiology , Nuclear Proteins/metabolism , Sarcoma, Kaposi/virology , Virus Replication , Animals , Antigens, Viral/genetics , Cell Line , Chlorocebus aethiops , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/metabolism , Mass Spectrometry , Nuclear Proteins/genetics , Protein Isoforms , Vero Cells , Virus Latency
7.
Antimicrob Agents Chemother ; 55(6): 2696-703, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21402841

ABSTRACT

Kaposi's sarcoma (KS) is the most common HIV-associated cancer worldwide and is associated with high levels of morbidity and mortality in some regions. Antiretroviral (ARV) combination regimens have had mixed results for KS progression and resolution. Anecdotal case reports suggest that protease inhibitors (PIs) may have effects against KS that are independent of their effect on HIV infection. As such, we evaluated whether PIs or other ARVs directly inhibit replication of Kaposi's sarcoma-associated herpesvirus (KSHV), the gammaherpesvirus that causes KS. Among a broad panel of ARVs tested, only the PI nelfinavir consistently displayed potent inhibitory activity against KSHV in vitro as demonstrated by an efficient quantitative assay for infectious KSHV using a recombinant virus, rKSHV.294, which expresses the secreted alkaline phosphatase. This inhibitory activity of nelfinavir against KSHV replication was confirmed using virus derived from a second primary effusion lymphoma cell line. Nelfinavir was similarly found to inhibit in vitro replication of an alphaherpesvirus (herpes simplex virus) and a betaherpesvirus (human cytomegalovirus). No activity was observed with nelfinavir against vaccinia virus or adenovirus. Nelfinavir may provide unique benefits for the prevention or treatment of HIV-associated KS and potentially other human herpesviruses by direct inhibition of replication.


Subject(s)
HIV Protease Inhibitors/pharmacology , Herpesvirus 8, Human/drug effects , Nelfinavir/pharmacology , Virus Replication/drug effects , Adenoviridae/drug effects , Adenoviridae/physiology , Alkaline Phosphatase/genetics , Animals , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Vaccinia virus/drug effects , Vaccinia virus/physiology , Vero Cells
8.
Virology ; 410(1): 17-29, 2011 Feb 05.
Article in English | MEDLINE | ID: mdl-21084105

ABSTRACT

We previously found that KSHV (HHV-8) lytic activation occurs during differentiation of oral keratinocytes in organotypic raft cultures. To further investigate the spatial and temporal aspects of KSHV lytic activation and the roles of integrins, cadherins, and calcium, we used rKSHV.219-infected primary oral keratinocytes in submerged, suspension, and direct suprabasal plating, models of differentiation. We found that early keratinocyte differentiation did not activate lytic KSHV in cells attached to a substratum, with activation only occurring in suprabasal cells. Temporally, KSHV lytic expression occurred between the expression of early and late differentiation markers. Keratinocytes differentiated in suspension culture, which mimics substratum loss that occurs with stratification, activated lytic KSHV. This lytic activation was inhibited by integrin engagement, showing that integrins are a control point for KSHV reactivation. A role for cadherins was not found. Elevated extracellular calcium was necessary, but not sufficient, for lytic activation.


Subject(s)
Cadherins/metabolism , Calcium/metabolism , Herpesvirus 8, Human/physiology , Integrins/metabolism , Keratinocytes/cytology , Keratinocytes/virology , Cell Differentiation , Cells, Cultured , Gene Expression Regulation, Viral , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mouth Mucosa/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism
9.
J Immunol ; 180(5): 3417-25, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292568

ABSTRACT

Little is known about what effector populations are associated with the control of human herpesvirus 8 (HHV-8) infection in vivo. We compared T lymphocyte subsets among HIV-HHV-8+ and HIV-HHV-8- infected human individuals. alphabeta+ T cells from HHV-8-infected individuals displayed a significantly higher percentage of differentiated effector cells among both CD4+ and CD8+ T cell subsets. HHV-8 infection was associated with significant expansion of gammadelta+ Vdelta1 T cells expressing a differentiated effector cell phenotype in peripheral blood. In vitro stimulation of PBMC from HHV-8-infected individuals with either infectious viral particles or different HHV-8 viral proteins resulted in gammadelta Vdelta1 T cell activation. In addition, gammadelta Vdelta1 T cells displayed a strong reactivity against HHV-8-infected cell lines and prevented the release of infectious viral particles following the induction of lyric replication. These data indicate that gammadelta T cells play a role in both innate and adaptive T cell responses against HHV-8 in immunocompetent individuals.


Subject(s)
Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 8, Human/immunology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Adult , Animals , Antiviral Agents/metabolism , Base Sequence , Cell Differentiation/immunology , Cell Line, Transformed , Cell Line, Tumor , Chlorocebus aethiops , Chronic Disease , Clone Cells , Herpesviridae Infections/pathology , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Male , Middle Aged , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Vero Cells
10.
J Virol ; 80(19): 9481-96, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16973553

ABSTRACT

Varicella-zoster virus (VZV) glycoprotein E (gE) is a multifunctional protein important for cell-cell spread, envelopment, and possibly entry. In contrast to other alphaherpesviruses, gE is essential for VZV replication. Interestingly, the N-terminal region of gE, comprised of amino acids 1 to 188, was shown not to be conserved in the other alphaherpesviruses by bioinformatics analysis. Mutational analysis was performed to investigate the functions associated with this unique gE N-terminal region. Linker insertions, serine-to-alanine mutations, and deletions were introduced in the gE N-terminal region in the VZV genome, and the effects of these mutations on virus replication and cell-cell spread, gE trafficking and localization, virion formation, and replication in vivo in the skin were analyzed. In summary, mutagenesis of the gE N-terminal region identified a new functional region in the VZV gE ectodomain essential for cell-cell spread and the pathogenesis of VZV skin tropism and demonstrated that different subdomains of the unique N-terminal region had specific roles in viral replication, cell-cell spread, and secondary envelopment.


Subject(s)
Glycoproteins/metabolism , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/pathogenicity , Skin Diseases, Infectious/metabolism , Skin Diseases, Infectious/virology , Viral Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Cell Line, Tumor , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/ultrastructure , Herpesvirus 3, Human/ultrastructure , Humans , Kinetics , Mice , Mice, SCID , Microscopy, Electron , Molecular Sequence Data , Mutation/genetics , Sequence Alignment , Skin Diseases, Infectious/pathology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/ultrastructure , Xenograft Model Antitumor Assays
11.
J Virol ; 78(23): 13293-305, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542680

ABSTRACT

The protein product of varicella-zoster virus (VZV) ORF47 is a serine/threonine protein kinase and tegument component. Evaluation of two recombinants of the Oka strain, rOka47DeltaC, with a C-terminal truncation of ORF47, and rOka47D-N, with a point mutation in the conserved kinase motif, showed that ORF47 kinase function was necessary for optimal VZV replication in human skin xenografts in SCID mice but not in cultured cells. We now demonstrate that rOka47DeltaC and rOka47D-N mutants do not infect human T-cell xenografts. Differences in the growth of kinase-defective ORF47 mutants allowed an examination of requirements for VZV pathogenesis in skin and T cells in vivo. Although virion assembly was reduced and no virion transport to cell surfaces was observed, epidermal cell fusion persisted, and VZV polykaryocytes were generated by rOka47DeltaC and rOka47D-N in skin. Virion assembly was also impaired in vitro, but VZV-induced cell fusion continued to cause syncytia in cultured cells infected with rOka47DeltaC or rOka47D-N. Intracellular trafficking of envelope glycoprotein E and the ORF47 and IE62 proteins, components of the tegument, was aberrant without ORF47 kinase activity. In summary, normal VZV virion assembly appears to require ORF47 kinase function. Cell fusion was induced by ORF47 mutants in skin, and cell-cell spread occurred even though virion formation was deficient. VZV-infected T cells do not undergo cell fusion, and impaired virion assembly by ORF47 mutants was associated with a complete elimination of T-cell infectivity. These observations suggest a differential requirement for cell fusion and virion formation in the pathogenesis of VZV infection in skin and T cells.


Subject(s)
Herpesvirus 3, Human/pathogenicity , Membrane Fusion , Skin/virology , T-Lymphocytes/virology , Virion/physiology , Virus Replication , Animals , Humans , Male , Mice , Protein Kinases/physiology , Skin Transplantation , Transplantation, Heterologous , Viral Envelope Proteins/physiology
12.
J Med Virol ; 70 Suppl 1: S95-102, 2003.
Article in English | MEDLINE | ID: mdl-12627496

ABSTRACT

ORF47, a serine/threonine protein kinase encoded by varicella-zoster virus (VZV), has often been compared to the ubiquitous cellular kinase, casein kinase II (CKII). However, no direct comparison of the two protein kinases has been carried out. Herein, we show that the ORF47 kinase was resistant to heparin, while CKII activity is profoundly inhibited by the acidic molecule in vitro. ORF47 required the presence of polyamines (aliphatic, positively-charged molecules) for in vitro activity. When polyamines were depleted from MeWo cells prior to VZV infection by pretreatment with D,L-alpha-difluoromethylornithine, VZV replication was reduced by 80%. Finally, the substrate specificity of the ORF47 kinase was defined using an in vitro assay. The ORF47 kinase phosphorylated maltose-binding protein, the mouse IgG2A heavy chain, the rabbit IgG heavy chain, casein, VZV ORF62, and VZV ORF63. The ORF47 kinase failed to phosphorylate an ORF62 truncation mutant, glutathione-S-transferase, or VZV gB. In contrast, CKII weakly phosphorylated VZV gB in vitro. By analyzing the sequences of these substrates, the minimal ORF47 consensus sequence was deduced to be the following motif: S/T-X-D/E-D/E, with a marked preference for additional acidic amino acids in the -1 and +1 position.


Subject(s)
Herpesvirus 3, Human/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Casein Kinase II , Cell Line , Consensus Sequence , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , HeLa Cells , Heparin/pharmacology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Humans , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Proteins/metabolism , Substrate Specificity , Virus Replication/drug effects
13.
J Clin Microbiol ; 40(7): 2526-32, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12089274

ABSTRACT

Fragments of glycoprotein G (gG-2(281-594His)), comprising residues 281 to 594 of herpes simplex virus type 2 (HSV-2), glycoprotein G of HSV-1 (gG-1(t26-189His)), and glycoprotein D of HSV-1 (gD-1(1-313)), were expressed in the baculovirus expression system to develop an assay for the detection of HSV-1 and HSV-2 type-specific antibodies. The expression of the gG-1(t26-189His) and gG-2(281-594His) fragments was analyzed by Western blotting using monoclonal antibodies LP10 and AP1, respectively. The molecular masses of the major products of gG-1(t26-189His) and the fragment of gG-2(281-594His) were 36 to 39 kDa and 64 to 72 kDa, respectively. Human sera positive for HSV-1 reacted with gG-1(t26-189His), sera positive for HSV-2 reacted with the gG-2(281-594His) fragment, and sera positive for both types reacted with gG-1(t26-189His) and gG-2(281-594His) in Western blotting. The human sera recognized polypeptides of gG-2(281-594His) with molecular masses of 57 to 67 and 120 to 150 kDa and additional faint bands of 21, 29, and 45 kDa. The recombinant gG-1(t26-189His) and the recombinant gG-2(281-594His) fragment were used as type-specific antigens for the detection of HSV-1- and HSV-2-specific antibody responses in human sera, respectively. As type-common antigens, an extract of HSV-1-infected Vero cells and recombinant gD-1(1-313) were used. An enzyme-linked immunosorbent assay to detect type-specific antibodies was developed, and the sensitivity and specificity were evaluated by comparison with commercial tests by using sera obtained from different sources. The sensitivity and specificity were 91.5 and 95.5%, respectively, compared to the Gull assay. The gG-2(281-594His) fragment can be obtained in relatively large quantities at low cost.


Subject(s)
Antibodies, Viral/analysis , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Animals , Antibody Specificity , Baculoviridae/genetics , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/statistics & numerical data , Gene Expression , Genes, Viral , Herpesvirus 1, Human/immunology , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Sensitivity and Specificity , Vero Cells , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...