Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Virology ; 491: 96-105, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891237

ABSTRACT

Varicella-zoster virus (VZV) is the causative agent of both chickenpox (varicella) and shingles (zoster). VZV survives host defenses, even with an intact immune system, and disseminates in the host before causing disease. To date, several diverse immunomodulatory strategies used by VZV to undermine host immunity have been identified; however, few studies have addressed the complement evasion strategies used by this virus. Here, we show that expression of CD59, which is a key member of host regulators of complement activation (RCA), is significantly upregulated in response to VZV infection in human T cells and dorsal root ganglia (DRG) but not in human skin xenografts in SCID-hu mice in vivo. This is the first report demonstrating that VZV infection upregulates host CD59 expression in a tissue-specific manner in vivo, which may aid VZV in complement evasion and pathogenesis.


Subject(s)
CD59 Antigens/genetics , Chickenpox/genetics , Herpesvirus 3, Human/physiology , Animals , CD59 Antigens/metabolism , Chickenpox/metabolism , Chickenpox/pathology , Chickenpox/virology , Disease Models, Animal , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/virology , Herpesvirus 3, Human/genetics , Host-Pathogen Interactions , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Male , Mice , Mice, SCID , Thymus Gland/metabolism , Thymus Gland/pathology , Thymus Gland/virology
2.
PLoS Pathog ; 11(5): e1004901, 2015 May.
Article in English | MEDLINE | ID: mdl-25973608

ABSTRACT

Varicella zoster virus (VZV) causes chickenpox in humans and, subsequently, establishes latency in the sensory ganglia from where it reactivates to cause herpes zoster. Infection of rhesus macaques with simian varicella virus (SVV) recapitulates VZV pathogenesis in humans thus representing a suitable animal model for VZV infection. While the type I interferon (IFN) response has been shown to affect VZV replication, the virus employs counter mechanisms to prevent the induction of anti-viral IFN stimulated genes (ISG). Here, we demonstrate that SVV inhibits type I IFN-activated signal transduction via the JAK-STAT pathway. SVV-infected rhesus fibroblasts were refractory to IFN stimulation displaying reduced protein levels of IRF9 and lacking STAT2 phosphorylation. Since previous work implicated involvement of the VZV immediate early gene product ORF63 in preventing ISG-induction we studied the role of SVV ORF63 in generating resistance to IFN treatment. Interestingly, SVV ORF63 did not affect STAT2 phosphorylation but caused IRF9 degradation in a proteasome-dependent manner, suggesting that SVV employs multiple mechanisms to counteract the effect of IFN. Control of SVV ORF63 protein levels via fusion to a dihydrofolate reductase (DHFR)-degradation domain additionally confirmed its requirement for viral replication. Our results also show a prominent reduction of IRF9 and inhibition of STAT2 phosphorylation in VZV-infected cells. In addition, cells expressing VZV ORF63 blocked IFN-stimulation and displayed reduced levels of the IRF9 protein. Taken together, our data suggest that varicella ORF63 prevents ISG-induction both directly via IRF9 degradation and indirectly via transcriptional control of viral proteins that interfere with STAT2 phosphorylation. SVV and VZV thus encode multiple viral gene products that tightly control IFN-induced anti-viral responses.


Subject(s)
Herpesviridae Infections/metabolism , Host-Pathogen Interactions , Interferon Type I/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Varicellovirus/physiology , Animals , Cell Line , Cercopithecinae , Chickenpox/immunology , Chickenpox/metabolism , Chickenpox/pathology , Chickenpox/virology , DNA, Recombinant/metabolism , Gene Expression Regulation, Viral , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 3, Human/immunology , Herpesvirus 3, Human/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Phosphorylation , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , Proteolysis , Recombinant Proteins/metabolism , STAT Transcription Factors/genetics , Varicellovirus/immunology
3.
PLoS Pathog ; 10(1): e1003896, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24497829

ABSTRACT

Varicella zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterized by epidermal skin blistering. Using a calcium-induced keratinocyte differentiation model we investigated the interaction between epidermal differentiation and VZV infection. RNA-seq analysis showed that VZV infection has a profound effect on differentiating keratinocytes, altering the normal process of epidermal gene expression to generate a signature that resembles patterns of gene expression seen in both heritable and acquired skin-blistering disorders. Further investigation by real-time PCR, protein analysis and electron microscopy revealed that VZV specifically reduced expression of specific suprabasal cytokeratins and desmosomal proteins, leading to disruption of epidermal structure and function. These changes were accompanied by an upregulation of kallikreins and serine proteases. Taken together VZV infection promotes blistering and desquamation of the epidermis, both of which are necessary to the viral spread and pathogenesis. At the same time, analysis of the viral transcriptome provided evidence that VZV gene expression was significantly increased following calcium treatment of keratinocytes. Using reporter viruses and immunohistochemistry we confirmed that VZV gene and protein expression in skin is linked with cellular differentiation. These studies highlight the intimate host-pathogen interaction following VZV infection of skin and provide insight into the mechanisms by which VZV remodels the epidermal environment to promote its own replication and spread.


Subject(s)
Cell Differentiation , Chickenpox/metabolism , Gene Expression Regulation, Viral/physiology , Herpesvirus 3, Human/physiology , Keratinocytes/metabolism , RNA, Viral/biosynthesis , Viral Proteins/biosynthesis , Virus Replication/physiology , Chickenpox/genetics , Female , Humans , Keratinocytes/pathology , Keratinocytes/virology , Male , RNA, Viral/genetics , Sequence Analysis, RNA , Viral Proteins/genetics
4.
J Virol ; 88(2): 890-902, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198400

ABSTRACT

Autophagy and the effects of its inhibition or induction were investigated during the entire infectious cycle of varicella-zoster virus (VZV), a human herpesvirus. As a baseline, we first enumerated the number of autophagosomes per cell after VZV infection compared with the number after induction of autophagy following serum starvation or treatment with tunicamycin or trehalose. Punctum induction by VZV was similar in degree to punctum induction by trehalose in uninfected cells. Treatment of infected cells with the autophagy inhibitor 3-methyladenine (3-MA) markedly reduced the viral titer, as determined by assays measuring both cell-free virus and infectious foci (P < 0.0001). We next examined a virion-enriched band purified by density gradient sedimentation and observed that treatment with 3-MA decreased the amount of VZV gE, while treatment with trehalose increased the amount of gE in the same band. Because VZV gE is the most abundant glycoprotein, we selected gE as a representative viral glycoprotein. To further investigate the role of autophagy in VZV glycoprotein biosynthesis as well as confirm the results obtained with 3-MA inhibition, we transfected cells with ATG5 small interfering RNA to block autophagosome formation. VZV-induced syncytium formation was markedly reduced by ATG5 knockdown (P < 0.0001). Further, we found that both expression and glycan processing of VZV gE were decreased after ATG5 knockdown, while expression of the nonglycosylated IE62 tegument protein was unchanged. Taken together, our cumulative results not only documented abundant autophagy within VZV-infected cells throughout the infectious cycle but also demonstrated that VZV-induced autophagy facilitated VZV glycoprotein biosynthesis and processing.


Subject(s)
Autophagy , Chickenpox/physiopathology , Herpesvirus 3, Human/physiology , Protein Biosynthesis , Viral Envelope Proteins/genetics , Autophagy-Related Protein 5 , Chickenpox/genetics , Chickenpox/metabolism , Chickenpox/virology , Herpesvirus 3, Human/genetics , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Viral Envelope Proteins/metabolism , Virus Replication
5.
J Virol ; 88(2): 1381-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227856

ABSTRACT

Programmed cell death (apoptosis) is an important host defense mechanism against intracellular pathogens, such as viruses. Accordingly, viruses have evolved multiple mechanisms to modulate apoptosis to enhance replication. Varicella-zoster virus (VZV) induces apoptosis in human fibroblasts and melanoma cells. We found that VZV triggered the phosphorylation of the proapoptotic proteins Bim and BAD but had little or no effect on other Bcl-2 family members. Since phosphorylation of Bim and BAD reduces their proapoptotic activity, this may prevent or delay apoptosis in VZV-infected cells. Phosphorylation of Bim but not BAD in VZV-infected cells was dependent on activation of the MEK/extracellular signal-regulated kinase (ERK) pathway. Cells knocked down for Bim showed delayed VZV plaque formation, resulting in longer survival of VZV-infected cells and increased replication of virus, compared with wild-type cells infected with virus. Conversely, overexpression of Bim resulted in earlier plaque formation, smaller plaques, reduced virus replication, and increased caspase 3 activity. Inhibition of caspase activity in VZV-infected cells overexpressing Bim restored levels of virus production similar to those seen with virus-infected wild-type cells. Previously we showed that VZV ORF12 activates ERK and inhibits apoptosis in virus-infected cells. Here we found that VZV ORF12 contributes to Bim and BAD phosphorylation. In summary, VZV triggers Bim phosphorylation; reduction of Bim levels results in longer survival of VZV-infected cells and increased VZV replication.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Chickenpox/metabolism , Down-Regulation , Herpesvirus 3, Human/physiology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Virus Replication , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Chickenpox/genetics , Chickenpox/virology , Herpesvirus 3, Human/genetics , Humans , MAP Kinase Signaling System , Membrane Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Viral Plaque Assay , Viral Proteins/genetics , Viral Proteins/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
6.
PLoS Pathog ; 9(8): e1003512, 2013.
Article in English | MEDLINE | ID: mdl-23935496

ABSTRACT

Varicella-zoster virus (VZV) is a neurotropic human alphaherpesvirus that causes varicella upon primary infection, establishes latency in multiple ganglionic neurons, and can reactivate to cause zoster. Live attenuated VZV vaccines are available; however, they can also establish latent infections and reactivate. Studies of VZV latency have been limited to the analyses of human ganglia removed at autopsy, as the virus is strictly a human pathogen. Recently, terminally differentiated human neurons have received much attention as a means to study the interaction between VZV and human neurons; however, the short life-span of these cells in culture has limited their application. Herein, we describe the construction of a model of normal human neural progenitor cells (NHNP) in tissue-like assemblies (TLAs), which can be successfully maintained for at least 180 days in three-dimensional (3D) culture, and exhibit an expression profile similar to that of human trigeminal ganglia. Infection of NHNP TLAs with cell-free VZV resulted in a persistent infection that was maintained for three months, during which the virus genome remained stable. Immediate-early, early and late VZV genes were transcribed, and low-levels of infectious VZV were recurrently detected in the culture supernatant. Our data suggest that NHNP TLAs are an effective system to investigate long-term interactions of VZV with complex assemblies of human neuronal cells.


Subject(s)
Chickenpox/metabolism , Herpesvirus 3, Human/physiology , Models, Biological , Neural Stem Cells/virology , Virus Latency/physiology , Cell Line, Tumor , Chickenpox/pathology , Female , Genes, Immediate-Early/physiology , Humans , Male , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Neurons/virology , Time Factors , Transcription, Genetic/physiology
7.
Int J Dermatol ; 51(7): 809-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22715825

ABSTRACT

BACKGROUND: Herpes virus infections are well known infectious complications of pemphigus and bullous pemphigoid. We describe pathologic findings utilizing autopsy tissue from several organs from a patient affected by a new variant of endemic pemphigus in El Bagre, Colombia, South America. CASE REPORT: We describe a patient by a new variant of endemic pemphigus foliaceus from El Bagre that was receiving high-dosage immunosuppressants when hospitalized and died suddenly following contact with a second patient affected by chicken pox. MATERIALS AND METHODS: We performed studies utilizing hematoxylin and eosin, immunohistochemistry, and direct immunofluorescence techniques on tissues from several organs. RESULTS: We detected the presence of varicella zoster virus, as well as strong positivity for α-1 antitrypsin in the heart, kidneys, spleen, liver, skin, brain, lungs, pancreas, small and large intestines, and skeletal muscle. In regard to structural damage in the kidney and heart, we believe the observed damage is associated with the presence of autoantibodies to these organs, since both of them are rich in plakins and El Bagre-EPF patients present significant antibodies to plakin molecules. CONCLUSION: In patients with endemic pemphigus foliaceus, we recommend complete isolation of the patient when receiving high dosages of systemic immunosuppressive agents. We further suggest the clinical possibility of a synergistic, fatal interaction between active pemphigus foliaceus, varicella zoster virus, herpes simplex virus, immunosuppressive agents, and a systemic activation of α-1 antitrypsin. Thus, we suggest adequate bed spacing, barrier nursing, and preventative testing for α-1 antitrypsin activation are warranted in these patients to address these complications.


Subject(s)
Chickenpox/complications , Herpesvirus 3, Human , Immunosuppressive Agents/therapeutic use , Pemphigus/complications , Pemphigus/drug therapy , alpha 1-Antitrypsin/metabolism , Acyclovir/therapeutic use , Adult , Antiviral Agents/therapeutic use , Azathioprine/therapeutic use , Chickenpox/drug therapy , Chickenpox/metabolism , Endemic Diseases , Fatal Outcome , Humans , Immunosuppressive Agents/adverse effects , Male , Pemphigus/enzymology , Prednisone/therapeutic use
8.
J Virol ; 86(6): 3143-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238304

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are a family of serine-threonine protein kinases involved in many cellular processes, including cell proliferation, differentiation, inflammation, and cell death. Activation of several MAPKs, including extracellular signal-regulated kinase 1 and 2 (ERK1/2), p38, and c-Jun N-terminal kinase (JNK), results in stimulation of activator protein 1 (AP-1), which promotes gene transcription. Previous studies have demonstrated that varicella-zoster virus (VZV) infection activates ERK1/2, p38, and JNK to promote viral replication, but the underlying mechanism(s) is unclear. To identify viral proteins responsible for the activation of MAPK, we used a proteomic approach to screen viral proteins for AP-1 promoter activation by an AP-1-luciferase reporter assay. We found that VZV ORF12 protein, located in the tegument of virions, enhances AP-1 reporter activity. This effect of ORF12 protein was markedly inhibited by a MAPK/ERK kinase 1 and 2 (MEK1/2) inhibitor (U0126), partially blocked by a p38 inhibitor (SB202190), but not inhibited by a JNK inhibitor (SP600125). Expression of VZV ORF12 protein in cells resulted in phosphorylation of ERK1/2 and p38 but not JNK. Infection of cells with a VZV ORF12 deletion mutant resulted in reduced levels of phosphorylated ERK1/2 (p-ERK1/2) compared to infection with wild-type VZV. Furthermore, deletion of ORF12 rendered VZV-infected cells more susceptible to staurosporine-induced apoptosis. In conclusion, VZV ORF12 protein activates the AP-1 pathway by selectively triggering the phosphorylation of ERK1/2 and p38. Cells infected with a VZV ORF12 deletion mutant have reduced levels of p-ERK1/2 and are more susceptible to apoptosis than cells infected with wild-type VZV.


Subject(s)
Apoptosis , Chickenpox/enzymology , Chickenpox/physiopathology , Herpesvirus 3, Human/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Viral Structural Proteins/metabolism , Cell Line , Chickenpox/metabolism , Chickenpox/virology , Herpesvirus 3, Human/genetics , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Open Reading Frames , Phosphorylation , Viral Structural Proteins/genetics
9.
J Biol Chem ; 286(20): 17921-33, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21385879

ABSTRACT

Innate cellular immunity is the immediate host response against pathogens, and activation of innate immunity also modulates the induction of adaptive immunity. The nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) are a family of intracellular receptors that recognize conserved patterns associated with intracellular pathogens, but information about their role in the host defense against DNA viruses is limited. Here we report that varicella-zoster virus (VZV), an alphaherpesvirus that is the causative agent of varicella and herpes zoster, induces formation of the NLRP3 inflammasome and the associated processing of the proinflammatory cytokine IL-1ß by activated caspase-1 in infected cells. NLRP3 inflammasome formation was induced in VZV-infected human THP-1 cells, which are a transformed monocyte cell line, primary lung fibroblasts, and melanoma cells. Absent in melanoma gene-2 (AIM2) is an interferon-inducible protein that can form an alternative inflammasome complex with caspase-1 in virus-infected cells. Experiments in VZV-infected melanoma cells showed that NLRP3 protein recruits the adaptor protein ASC and caspase-1 to form an NLRP3 inflammasome complex independent of AIM2 protein and in the absence of free radical reactive oxygen species release. NLRP3 was also expressed extensively in infected skin xenografts in the severe combined immunodeficiency mouse model of VZV pathogenesis in vivo. We conclude that NLRP3 inflammasome formation is an innate cellular response to infection with this common pathogenic human herpesvirus.


Subject(s)
Chickenpox/metabolism , Herpes Zoster/metabolism , Herpesvirus 3, Human/metabolism , Immunity, Innate , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cell Line, Tumor , Chickenpox/genetics , Chickenpox/immunology , DNA-Binding Proteins , Enzyme Activation/genetics , Enzyme Activation/immunology , Herpes Zoster/genetics , Herpes Zoster/immunology , Herpesvirus 3, Human/immunology , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, SCID , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Skin Transplantation , Transplantation, Heterologous
10.
J Virol ; 84(23): 12454-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861271

ABSTRACT

Studies of varicella-zoster virus gene expression during latency require the acquisition of human ganglia at autopsy. Concerns have been raised that the virus might reactivate immediately after death. Because features of varicella-zoster virus latency are similar in primate and human ganglia, we examined virus gene expression in tissues either processed immediately or kept at 4°C for 30 h before necropsy of two monkeys inoculated with simian varicella-zoster virus and euthanized 117 days later. Virus transcription and the detection of open reading frame (ORF) 63 protein in the cytoplasm of neurons were comparable. Thus, a 30-h delay after death did not affect varicella-zoster virus expression in latently infected ganglia.


Subject(s)
Chickenpox/physiopathology , Ganglia/metabolism , Gene Expression Regulation, Viral/physiology , Herpesvirus 3, Human/physiology , Immediate-Early Proteins/metabolism , Viral Envelope Proteins/metabolism , Virus Latency/physiology , Animals , Chickenpox/metabolism , Herpesvirus 3, Human/metabolism , Immunohistochemistry , Macaca mulatta , Neurons/virology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Viremia/blood
11.
Proc Natl Acad Sci U S A ; 107(1): 282-7, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966293

ABSTRACT

Varicella-zoster virus (VZV) is an alphaherpesvirus that infects skin, lymphocytes, and sensory ganglia. VZV glycoprotein E (gE) has a unique N-terminal region (aa1-188), which is required for replication and includes domains involved in secondary envelopment, efficient cell-cell spread, and skin infection in vivo. The nonconserved N-terminal region also mediates binding to the insulin-degrading enzyme (IDE), which is proposed to be a VZV receptor. Using viral mutagenesis to make the recombinant rOka-DeltaP27-G90, we showed that amino acids in this region are required for gE/IDE binding in infected cells; this deletion reduced cell-cell spread in vitro and skin infection in vivo. However, a gE point mutation, linker insertions, and partial deletions in the aa27-90 region, and deletion of a large portion of the unique N-terminal region, aa52-187, had similar or more severe effects on VZV replication in vitro and in vivo without disrupting the gE/IDE interaction. VZV replication in T cells in vivo was not impaired by deletion of gE aa27-90, suggesting that these gE residues are not essential for VZV T cell tropism. However, the rOka-DeltaY51-P187 mutant failed to replicate in T cell xenografts as well as skin in vivo. VZV tropism for T cells and skin, which is necessary for its life cycle in the human host, requires this nonconserved region of the N-terminal region of VZV gE.


Subject(s)
Chickenpox/physiopathology , Herpesvirus 3, Human/pathogenicity , Viral Envelope Proteins/metabolism , Animals , Cell Line, Tumor , Chickenpox/metabolism , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Humans , Mice , Mice, SCID , Mutagenesis , Protein Structure, Tertiary , Skin/cytology , Skin/pathology , Skin/virology , Skin Diseases/pathology , Skin Diseases/virology , Skin Transplantation , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transplantation, Heterologous , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virus Replication/genetics
12.
J Virol ; 83(15): 7560-72, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19457996

ABSTRACT

Varicella-zoster virus (VZV) open reading frame 61 (ORF61) encodes a protein that transactivates viral and cellular promoters in transient-transfection assays and is the ortholog of herpes simplex virus ICP0. In this report, we mapped the ORF61 promoter and investigated its regulation by viral and cellular proteins in transient-expression experiments and by mutagenesis of the VZV genome (parent Oka strain). The 5' boundary of the minimal ORF61 promoter required for IE62 transactivation was mapped to position -95 relative to the mRNA start site, and three noncanonical GT-rich Sp1-binding sites were documented to occur within the region comprising positions -95 to -45. Contributions of the three Sp1-binding-site motifs, designated Sp1a, Sp1b, and Sp1c, to ORF61 expression and viral replication were varied despite their similar sequences. Two sites, Sp1a and Sp1c, functioned synergistically. When both sites were mutated in the pOka genome to produce pOka-61proDeltaSp1ac, the mutant virus expressed significantly less ORF61 protein. Using this mutant to investigate ORF61 functions resulted in reductions in the expression levels of IE proteins, viral kinases ORF47 and ORF66, and the major glycoprotein gE, with the most impact on gE. Virion morphogenesis appeared to be intact despite minimal ORF61 expression. Pretreating melanoma cells with sodium butyrate enhanced titers of pOka-61proDeltaSp1ac but not pOka, suggesting that ORF61 has a role in histone deacetylase inhibition. Growth of pOka-61proDeltaSp1ac was impaired in SCIDhu skin xenografts, indicating that the regulation of the ORF61 promoter by Sp1 family proteins is important for ORF61 expression in vivo and that ORF61 contributes to VZV virulence at skin sites of replication.


Subject(s)
Chickenpox/virology , Gene Expression Regulation, Viral , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/pathogenicity , Promoter Regions, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Animals , Binding Sites , Chickenpox/metabolism , Disease Models, Animal , Herpesvirus 3, Human/chemistry , Herpesvirus 3, Human/genetics , Humans , Mice , Mice, SCID , Skin/metabolism , Skin/virology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Viral Proteins/chemistry
13.
J Virol ; 83(15): 7495-506, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19474103

ABSTRACT

Glycoprotein B (gB), the most conserved protein in the family Herpesviridae, is essential for the fusion of viral and cellular membranes. Information about varicella-zoster virus (VZV) gB is limited, but homology modeling showed that the structure of VZV gB was similar to that of herpes simplex virus (HSV) gB, including the putative fusion loops. In contrast to HSV gB, VZV gB had a furin recognition motif ([R]-X-[KR]-R-|-X, where | indicates the position at which the polypeptide is cleaved) at residues 491 to 494, thought to be required for gB cleavage into two polypeptides. To investigate their contribution, the putative primary fusion loop or the furin recognition motif was mutated in expression constructs and in the context of the VZV genome. Substitutions in the primary loop, W180G and Y185G, plus the deletion mutation Delta491RSRR494 and point mutation 491GSGG494 in the furin recognition motif did not affect gB expression or cellular localization in transfected cells. Infectious VZV was recovered from parental Oka (pOka)-bacterial artificial chromosomes that had either the Delta491RSRR494 or 491GSGG494 mutation but not the point mutations W180G and Y185G, demonstrating that residues in the primary loop of gB were essential but gB cleavage was not required for VZV replication in vitro. Virion morphology, protein localization, plaque size, and replication were unaffected for the pOka-gBDelta491RSRR494 or pOka-gB491GSGG494 virus compared to pOka in vitro. However, deletion of the furin recognition motif caused attenuation of VZV replication in human skin xenografts in vivo. This is the first evidence that cleavage of a herpesvirus fusion protein contributes to viral pathogenesis in vivo, as seen for fusion proteins in other virus families.


Subject(s)
Chickenpox/virology , Furin/metabolism , Herpesvirus 3, Human/pathogenicity , Mutagenesis , Skin/virology , Viral Envelope Proteins/genetics , Virus Replication , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line, Tumor , Chickenpox/metabolism , Chickenpox/pathology , Herpesvirus 3, Human/chemistry , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Humans , In Vitro Techniques , Mice , Mice, SCID , Molecular Sequence Data , Mutation , Protein Binding , Sequence Alignment , Skin/metabolism , Skin/pathology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
14.
Inflammation ; 30(1-2): 38-43, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17171437

ABSTRACT

The acute involution of the thymus is induced by either exogenous or endogenous factors, including some infections (infection type involution). The present study was focused on both detection and immunocytochemical analysis of NGF immunopositive mast cells in child thymus with acute infection-induced involution. Autopsy thymus specimens from children with infection diseases (Sepsis, Encephalomyelitis, Varicella) were examined at light and electron microscopic level and compared to normal infantile thymuses. We observed a redistribution of NGF immunopositive mast cells in infection-affected child thymus, which lobular architecture was collapsed. A positive correlation between the degree of the involutive changes, increased distribution and enhanced NGF immunoreactivity of mast cells was defined. The possible involvement of NGF immunopositive mast cells in the process of acute thymus involution is discussed.


Subject(s)
Chickenpox/metabolism , Encephalomyelitis/metabolism , Mast Cells/chemistry , Nerve Growth Factor/analysis , Sepsis/metabolism , Thymus Gland/chemistry , Case-Control Studies , Chickenpox/enzymology , Chickenpox/pathology , Child , Child, Preschool , Encephalomyelitis/enzymology , Encephalomyelitis/pathology , Female , Humans , Immunohistochemistry , Infant , Male , Mast Cells/enzymology , Mast Cells/ultrastructure , Microscopy, Immunoelectron , Receptor, trkA/analysis , Sepsis/enzymology , Sepsis/pathology , Thymus Gland/enzymology , Thymus Gland/ultrastructure , Tryptases/analysis
15.
J Virol ; 80(19): 9772-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16973581

ABSTRACT

Open reading frame 4 (ORF4) of varicella-zoster virus (VZV) encodes an immediate-early protein that is believed to be important for viral infectivity and establishing latency. Evidence suggests that VZV-specific T cells are crucial in the control of viral replication, but there are no data addressing the existence of potential ORF4 protein-specific CD4+ T cells. We tested the hypothesis that VZV ORF4 protein-specific CD4+ T cells could be identified and characterized within the peripheral blood of healthy immune donors following primary infection. Gamma interferon (IFN-gamma) immunosorbent assays were used to screen peripheral blood mononuclear cells obtained from healthy seropositive donors for responses to overlapping ORF4 peptides, viral lysate, and live vaccine. High frequencies of ORF4 protein-specific T cells were detected ex vivo in individuals up to 52 years after primary infection. Several immunogenic regions of the ORF4 protein were identified, including a commonly recognized epitope which was restricted through HLA-DRB1*07. Total ORF4 protein-specific responses comprised 19.7% and 20.7% of the total lysate and vaccine responses, respectively, and were dominated by CD4+ T cells. Indeed, CD4+ T cells were found to dominate the overall virus-specific IFN-gamma cellular immune response both ex vivo and after expansion in vitro. In summary, we have identified an ORF4 protein as a novel target antigen for persistent VZV-specific CD4+ T cells, with implications for disease pathogenesis and future vaccine development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Herpesvirus 3, Human/immunology , Immediate-Early Proteins/immunology , Viral Proteins/immunology , Adult , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chickenpox/immunology , Chickenpox/metabolism , Epitopes, B-Lymphocyte/immunology , HLA-DR Antigens/immunology , HLA-DRB1 Chains , Herpesvirus 3, Human/physiology , Herpesvirus Vaccines/immunology , Humans , Immediate-Early Proteins/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Viral Proteins/metabolism
16.
Cell ; 119(7): 915-26, 2004 Dec 29.
Article in English | MEDLINE | ID: mdl-15620351

ABSTRACT

Varicella zoster virus (VZV) is a highly infectious human pathogen; nevertheless, infectious virions are not released in vitro where infection is cell associated. Four VZV envelope glycoproteins contain mannose 6-phosphate (Man 6-P), and Man 6-P blocks infection of cells by cell-free VZV. Expression of antisense cDNA or siRNA-like transcripts were used to generate five stable human cell lines deficient in cation-independent mannose 6-phosphate receptors (MPRci). All 5 MPRci-deficient lines resisted infection by cell-free, but not cell-associated, VZV, secreted lysosomal enzymes, and released infectious virions when infected by cell-associated VZV. Intracellular MPRci thus appear to divert newly enveloped VZV to late endosomes, and plasmalemmal MPRci are necessary for entry by cell-free VZV. Biopsies from VZV-infected human skin supported the idea that because MPRci expression is naturally lost in maturing superficial epidermal cells, these cells do not divert VZV to endosomes and constitutively secrete infectious VZV.


Subject(s)
Chickenpox/metabolism , Epidermis/metabolism , Epidermis/virology , Herpes Zoster/metabolism , Herpesvirus 3, Human/metabolism , Receptor, IGF Type 2/metabolism , Cell Line, Tumor , Cells, Cultured , Chickenpox/pathology , Chickenpox/virology , DNA, Antisense/genetics , DNA, Antisense/metabolism , Endosomes/virology , Epidermis/pathology , Epidermis/ultrastructure , Herpes Zoster/pathology , Herpes Zoster/virology , Herpesvirus 3, Human/growth & development , Herpesvirus 3, Human/pathogenicity , Herpesvirus 3, Human/ultrastructure , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, IGF Type 2/deficiency , Receptor, IGF Type 2/genetics
17.
Arch Virol Suppl ; (17): 135-42, 2001.
Article in English | MEDLINE | ID: mdl-11339542

ABSTRACT

The limited supple of appropriate tissues for study has been an impediment to investigations of varicella zoster virus (VZV) latency. Human dorsal root ganglia (DRG) harboring latent virus are not plentiful and are not amenable to manipulation for studying the events surrounding the establishment, maintenance, and cessation of latency. An alternative to studies in human DRG is the rat model of latency, which appears to provide a reliable method of investigating VZV latency. Other alternatives include studies in other human tissues involved in VZV pathogenesis. In order to improve our understanding of the establishment and cessation of latency, we performed comparative immunohistochemical analysis of chickenpox and zoster skin lesions. This analysis revealed that during primary infection and reactivation productive VZV infection occurs in a variety of cell types and that the major VZV DNA binding protein, ORF29p, is present in peripheral axons early during the course of chickenpox. VZV latency was studied in the rat model by in situ hybridization and compared with similar studies performed in human DRG containing latent virus, confirming that VZV DNA persists in the same sites in DRG of the two species.


Subject(s)
Chickenpox/virology , Herpes Zoster/virology , Virus Activation , Virus Latency , Animals , Chickenpox/metabolism , Chickenpox/pathology , Ganglia, Spinal/pathology , Ganglia, Spinal/virology , Herpes Zoster/metabolism , Herpes Zoster/pathology , Herpesvirus 3, Human/growth & development , Herpesvirus 3, Human/isolation & purification , Herpesvirus 3, Human/metabolism , Herpesvirus 3, Human/physiology , Humans , Rats , Skin/pathology , Skin/virology
18.
Electrophoresis ; 17(3): 612-6, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8740187

ABSTRACT

Acute-phase serum proteins were analyzed by two-dimensional electrophoresis with isoelectric focusing in 3-10 immobilized pH gradients. Most spots were identified by reference to the plasma map in the SWISS-2DPAGE database. Serum amyloid A protein spots were identified by immunoblotting with specific antiserum and by matching determined with predicted values of electrophoretic parameters. Changes in the concentrations of alpha 1-antitrypsin, leucine-rich glycoprotein, haptoglobin, serum retinol-binding protein and transthyretin were quantitated by densitometry of silver-stained gels. Electrophoretic patterns from 18 patients with bacterial diseases and 16 patients with viral diseases were compared. The incidence of serum amyloid A protein spots was 18/18 in bacterial diseases and 6/16 in viral diseases. As the the other reactants studied, variations were simultaneous in bacterial disease and tended to be staggered in viral diseases.


Subject(s)
Acute-Phase Proteins/analysis , Bacterial Infections/blood , Electrophoresis, Gel, Two-Dimensional , Virus Diseases/blood , Chickenpox/blood , Chickenpox/metabolism , Child , Child, Preschool , Haemophilus Infections/blood , Haemophilus Infections/metabolism , Haemophilus influenzae/isolation & purification , Humans , Measles/blood , Measles/metabolism , Mumps/blood , Mumps/metabolism , Salmonella/isolation & purification , Salmonella Infections/blood , Salmonella Infections/metabolism , Streptococcal Infections/blood , Streptococcal Infections/metabolism , Streptococcus pyogenes/isolation & purification
19.
Microbiol Immunol ; 39(3): 217-20, 1995.
Article in English | MEDLINE | ID: mdl-7603367

ABSTRACT

The effect of peripheral blood mononuclear cells (PBMC) on expression of varicella-zoster virus (VZV) glycoproteins (Gps) was analyzed by flow cytometry. PBMC from VZV seropositive and seronegative donors and supernatant of PBMC co-cultured with VZV-infected human embryonic fibroblasts reduced VZV Gp expression. Neutralization of supernatant fluid with mixture of anti-interferons (IFN)-alpha, -beta, -gamma, and tumor necrosis factor (TNF)-alpha partially reduced inhibitory activity of supernatant on VZV Gp expression. Deletion of natural killer (NK) cells and adherent cells from PBMC reduced inhibitory activity of PBMC on VZV Gp expression. These results suggest that IFN-alpha, -beta, -gamma, TNF-alpha and other soluble factors released from NK cells and monocytes by co-cultivation with VZV-infected fibroblasts inhibit VZV Gp expression.


Subject(s)
Glycoproteins/biosynthesis , Herpesvirus 3, Human/metabolism , Leukocytes, Mononuclear/metabolism , Viral Envelope Proteins/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Chickenpox/metabolism , Cytokines/analysis , Cytokines/immunology , Flow Cytometry , Herpesvirus 3, Human/drug effects , Herpesvirus 3, Human/immunology , Humans , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology
20.
J Interferon Res ; 13(2): 111-20, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8509658

ABSTRACT

The pharmacokinetics and antiviral activity of recombinant human interferon-beta ser17 (Betaseron) were evaluated in African green monkeys. In one study, animals infected with simian varicella virus were administered Betaseron intravenously (i.v.), intramuscularly (i.m.), or subcutaneously (s.c.) at doses of 1 x 10(6) or 1 x 10(7) IU/kg twice daily for 10 days. In another study, infected animals received Betaseron s.c. at doses of 1 x 10(6) IU/kg twice daily, 2 x 10(6) IU/kg once daily, 4 x 10(6) IU/kg every other day, or 6 x 10(6) IU/kg every 3 days for 10 days. Following i.v. administration, mean clearance, steady-state volume of distribution, and terminal half-life values for Betaseron were 0.36 +/- 0.08 liters/hr.kg, 0.65 +/- 0.09 liters/kg, and 1.9 +/- 0.43 h, respectively. Although bioavailability following i.m. and s.c. administration was only 30-50%, antiviral activity, as measured by reduction in viremia and appearance of skin rash, was comparable for i.v., i.m., and s.c. administration of 1 x 10(6) IU/kg of Betaseron twice daily. With increasing dose (1 x 10(6) IU/kg to 1 x 10(7) IU/kg), both the area under the serum concentration-time curve (AUC) and antiviral activity of Betaseron tended to increase. When comparing various s.c. dosing regimens, there was significant accumulation of Betaseron in serum with repeated twice-daily dosing. However, no accumulation of Betaseron in serum was observed if the dosing interval was less frequent than once daily. Antiviral activity was greatest with twice-daily or once-daily s.c. administrations of Betaseron.


Subject(s)
Chickenpox/drug therapy , Interferon-beta/pharmacokinetics , Interferon-beta/therapeutic use , Monkey Diseases/drug therapy , Animals , Chickenpox/metabolism , Chlorocebus aethiops , Disease Models, Animal , Drug Administration Routes , Drug Administration Schedule , Drug Evaluation , Interferon beta-1a , Interferon beta-1b , Monkey Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...