Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Virol ; 90(23): 10789-10799, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27681121

ABSTRACT

The interferon (IFN) response to viral pathogens is critical for host survival. In humans and mouse models, defects in IFN responses can result in lethal herpes simplex virus 1 (HSV-1) infections, usually from encephalitis. Although rare, HSV-1 can also cause fulminant hepatic failure, which is often fatal. Although herpes simplex encephalitis has been extensively studied, HSV-1 generalized infections and subsequent acute liver failure are less well understood. We previously demonstrated that IFN-αßγR-/- mice are exquisitely susceptible to liver infection following corneal infection with HSV-1. In this study, we used bone marrow chimeras of IFN-αßγR-/- (AG129) and wild-type (WT; 129SvEv) mice to probe the underlying IFN-dependent mechanisms that control HSV-1 pathogenesis. After infection, WT mice with either IFN-αßγR-/- or WT marrow exhibited comparable survival, while IFN-αßγR-/- mice with WT marrow had a significant survival advantage over their counterparts with IFN-αßγR-/- marrow. Furthermore, using bioluminescent imaging to maximize data acquisition, we showed that the transfer of IFN-competent hematopoietic cells controlled HSV-1 replication and damage in the livers of IFN-αßγR-/- mice. Consistent with this, the inability of IFN-αßγR-/- immune cells to control liver infection in IFN-αßγR-/- mice manifested as profoundly elevated aspartate transaminase (AST) and alanine transaminase (ALT) levels, indicative of severe liver damage. In contrast, IFN-αßγR-/- mice receiving WT marrow exhibited only modest elevations of AST and ALT levels. These studies indicate that IFN responsiveness of the immune system is a major determinant of viral tropism and damage during visceral HSV infections. IMPORTANCE: Herpes simplex virus 1 (HSV-1) infection is an incurable viral infection with the most significant morbidity and mortality occurring in neonates and patients with compromised immune systems. Severe pathologies from HSV include the blindness-inducing herpetic stromal keratitis, highly debilitating and lethal herpes simplex encephalitis, and generalized infections that can lead to herpes simplex virus-induced acute liver failure. While immune compromise is a known factor, the precise mechanisms that lead to generalized HSV infections are unknown. In this study, we used and developed a mouse model system in combination with real-time bioluminescence imaging to demonstrate the relative importance of the immune and nonimmune compartments for containing viral spread and promoting host survival after corneal infection. Our results shed light on the pathogenesis of HSV infections that lead to generalized infection and acute liver failure.


Subject(s)
Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Interferons/metabolism , Liver Failure, Acute/immunology , Animals , Disease Models, Animal , Female , Herpes Simplex/etiology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Humans , Immunocompromised Host , Interferons/deficiency , Interferons/genetics , Keratitis, Herpetic/etiology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/etiology , Liver Failure, Acute/virology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Radiation Chimera/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Virus Replication/immunology
2.
J Neurosci Methods ; 212(1): 173-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23068604

ABSTRACT

The immortalized human cerebral microvascular endothelial cell line HCMEC/D3 presents a less expensive and more logistically feasible alternative to primary human brain microvascular endothelial cells (HBMEC's) for use in constructing in vitro models of the blood brain barrier (BBB). However, the fidelity of the HCMEC/D3 cell line to primary HBMEC's in studies of immune transmigration has yet to be established. Flow cytometric analysis of primary human leukocyte migration across in vitro BBB's generated with either HCMEC/D3 or primary HBMEC's revealed that HCMEC/D3 maintains the immune barrier properties of primary HBMEC's. Leukocyte migration responses and inflammatory cytokine production were statistically indistinguishable between both endothelial cell types, and both cell types responded similarly to astrocyte coculture, stimulation of leukocytes with phorbol myristate acetate (PMA) and ionomycin, and inflammatory cytokine treatment. This report is the first to validate the HCMEC/D3 cell line in a neuroimmunological experimental system via direct comparison to primary HBMEC's, demonstrating remarkable fidelity in terms of barrier resistance, immune migration profiles, and responsiveness to inflammatory cytokines. Moreover, we report novel findings demonstrating that interaction effects between immune cells and resident CNS cells are preserved in HCMEC/D3, suggesting that important characteristics of neuroimmune interactions during CNS inflammation are preserved in systems utilizing this cell line. Together, these findings demonstrate that HCMEC/D3 is a valid and powerful tool for less expensive and higher throughput in vitro investigations of immune migration at the BBB.


Subject(s)
Blood-Brain Barrier/physiology , Cell Movement/physiology , Endothelial Cells/physiology , Leukocytes/physiology , Models, Biological , Analysis of Variance , Area Under Curve , Astrocytes/drug effects , Astrocytes/physiology , Blood-Brain Barrier/drug effects , Calcium Ionophores/pharmacology , Cell Movement/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Cytokines/metabolism , Cytokines/pharmacology , Diffusion Chambers, Culture , Flow Cytometry , Humans , Ionomycin/pharmacology , Leukocytes/drug effects , Phenobarbital/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
3.
J Virol ; 86(14): 7692-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553331

ABSTRACT

Herpes simplex viruses lacking the virion host shutoff function (Δvhs) are avirulent and hypersensitive to type I and type II interferon (IFN). In this study, we demonstrate that even in the absence of IFN responses in AG129 (IFN-αßγR(-/-)) mice, Δvhs remains highly attenuated via corneal infection but is fully virulent via intracranial infection. The data demonstrate that the interferon-independent inherent replication defect of Δvhs has a significant impact upon peripheral replication and neuroinvasion.


Subject(s)
Cornea/virology , Herpesvirus 1, Human/pathogenicity , Interferon Type I/immunology , Interferon-gamma/immunology , Keratitis, Herpetic/virology , Ribonucleases/metabolism , Viral Proteins/metabolism , Animals , Bone Marrow Cells/virology , Cornea/pathology , Dendritic Cells/virology , Herpesvirus 1, Human/physiology , Mice , Mice, Transgenic , Ribonucleases/genetics , Viral Proteins/genetics , Virus Replication
4.
J Virol ; 85(24): 12972-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994441

ABSTRACT

Innate immune deficiencies result in a spectrum of severe clinical outcomes following infection. In particular, there is a strong association between loss of the signal transducer and activator of transcription (Stat) pathway, breach of the blood-brain barrier (BBB), and virus-induced neuropathology. The gene signatures that characterize resistance, disease, and mortality in the virus-infected nervous system have not been defined. Herpes simplex virus type 1 (HSV-1) is commonly associated with encephalitis in humans, and humans and mice lacking Stat1 display increased susceptibility to HSV central nervous system (CNS) infections. In this study, two HSV-1 strains were used, KOS (wild type [WT]), and Δvhs, an avirulent recombinant lacking the virion host shutoff (vhs) function. In addition, two mouse strains were used: strain 129 (control) and a Stat1-deficient (Stat1(-/-)) strain. Using combinations of these virus and mouse strains, we established a model of infection resulting in three different outcomes: viral clearance without neurological disease (Δvhs infection of control mice), neurological disease followed by viral clearance (Δvhs infection of Stat1(-/-) mice and WT infection of control mice), or neurological disease followed by death (WT infection of Stat1(-/-) mice). Through the use of functional genomics on the infected brain stems, we determined gene signatures that were representative of the three infection outcomes. We demonstrated a pathological signature in the brain stem of Stat1-deficient mice characterized by upregulation of transcripts encoding chemokine receptors, inflammatory markers, neutrophil chemoattractants, leukocyte adhesion proteins, and matrix metalloproteases. Additionally, there was a greater than 100-fold increase in the inflammatory markers interleukin 1ß (IL-1ß) and IL-6. Consistent with this gene signature, we demonstrated profound CNS inflammation with a concomitant lethal breach of the BBB. Taken together, our results indicated an essential role for normal Stat1-dependent signaling in mediating a nonpathological immune response to viral CNS infection.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , STAT1 Transcription Factor/immunology , Animals , Brain Stem/pathology , Brain Stem/virology , Cytokines/biosynthesis , Gene Expression Profiling , Histocytochemistry , Keratitis, Herpetic/complications , Mice , Mice, Knockout , Microarray Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
5.
PLoS One ; 6(9): e24018, 2011.
Article in English | MEDLINE | ID: mdl-21915277

ABSTRACT

Pivotal components of the IFN response to virus infection include the IFN receptors (IFNR), and the downstream factor signal transducer and activator of transcription 1 (Stat1). Mice deficient for Stat1 and IFNR (Stat1(-/-) and IFNαßγR(-/-) mice) lack responsiveness to IFN and exhibit high sensitivity to various pathogens. Here we examined herpes simplex virus type 1 (HSV-1) pathogenesis in Stat1(-/-) mice and in IFNαßγR(-/-) mice following corneal infection and bioluminescent imaging. Two divergent and paradoxical patterns of infection were observed. Mice with an N-terminal deletion in Stat1 (129Stat1(-/-) (N-term)) had transient infection of the liver and spleen, but succumbed to encephalitis by day 10 post-infection. In stark contrast, infection of IFNαßγR(-/-) mice was rapidly fatal, with associated viremia and fulminant infection of the liver and spleen, with infected infiltrating cells being primarily of the monocyte/macrophage lineage. To resolve the surprising difference between Stat1(-/-) and IFNαßγR(-/-) mice, we infected an additional Stat1(-/-) strain deleted in the DNA-binding domain (129Stat1(-/-) (DBD)). These 129Stat1(-/-) (DBD) mice recapitulated the lethal pattern of liver and spleen infection seen following infection of IFNαßγR(-/-) mice. This lethal pattern was also observed when 129Stat1(-/-) (N-term) mice were infected and treated with a Type I IFN-blocking antibody, and immune cells derived from 129Stat1(-/-) (N-term) mice were shown to be responsive to Type I IFN. These data therefore show significant differences in viral pathogenesis between two commonly-used Stat1(-/-) mouse strains. The data are consistent with the hypothesis that Stat1(-/-) (N-term) mice have residual Type I IFN receptor-dependent IFN responses. Complete loss of IFN signaling pathways allows viremia and rapid viral spread with a fatal infection of the liver. This study underscores the importance of careful comparisons between knockout mouse strains in viral pathogenesis, and may also be relevant to the causation of HSV hepatitis in humans, a rare but frequently fatal infection.


Subject(s)
Herpes Simplex/pathology , Luminescent Measurements/methods , Receptors, Interferon/deficiency , STAT1 Transcription Factor/deficiency , Animals , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpesvirus 1, Human/pathogenicity , Liver/pathology , Liver/virology , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptors, Interferon/genetics , STAT1 Transcription Factor/genetics , Spleen/pathology , Spleen/virology
6.
J Virol ; 84(19): 9685-94, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660188

ABSTRACT

The initiation of the immune response at the cellular level relies on specific recognition molecules to rapidly signal viral infection via interferon (IFN) regulatory factor 3 (IRF-3)-dependent pathways. The absence of IRF-3 would be expected to render such pathways inoperative and thereby significantly affect viral infection. Unexpectedly, a previous study found no significant change in herpes simplex virus (HSV) pathogenesis in IRF-3(-/-) mice following intravenous HSV type 1 (HSV-1) challenge (K. Honda, H. Yanai, H. Negishi, M. Asagiri, M. Sato, T. Mizutani, N. Shimada, Y. Ohba, A. Takaoka, N. Yoshida, and T. Taniguchi, Nature 434:772-777, 2005). In contrast, the present study demonstrated that IRF-3(-/-) mice are significantly more susceptible to HSV infection via the corneal and intracranial routes. Following corneal infection with 2 x 10(6) PFU of HSV-1 strain McKrae, 50% of wild-type mice survived, compared to 10% of IRF-3-deficient mice. Significantly increased viral replication and inflammatory cytokine production were observed in brain tissues of IRF-3(-/-) mice compared to control mice, with a concomitant deficit in production of both IFN-beta and IFN-alpha. These data demonstrate a critical role for IRF-3 in control of central nervous system infection following HSV-1 challenge. Furthermore, this work underscores the necessity to evaluate multiple routes of infection and animal models in order to fully determine the role of host resistance factors in pathogenesis.


Subject(s)
Central Nervous System Viral Diseases/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/pathogenicity , Interferon Regulatory Factor-3/physiology , Animals , Brain Stem/immunology , Cytokines/biosynthesis , Disease Models, Animal , Female , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions/immunology , Inflammation Mediators/metabolism , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Type I/biosynthesis , Keratitis, Herpetic/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Replication
7.
J Virol ; 83(5): 2075-87, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19109391

ABSTRACT

Humans and mice lacking the interferon signaling molecule Stat1 are sensitive to a variety of pathogens due to their presumed inability to mount a strong innate immune response. The herpes simplex virus type 1 (HSV-1) virion host shutoff (vhs) protein is a multifunctional immunomodulator that counteracts the innate immune response and viruses lacking vhs are attenuated and effective live vaccines in animal models. To investigate the interplay of viruses with an immunocompromised host, we performed functional genomics analyses on control and Stat1(-/-) mouse corneas infected with wild-type or vhs-null viruses. In control mice, correlative with viral growth, both viruses induced a transient increase in immunomodulators, followed by viral clearance. In contrast, infection of the Stat1(-/-) mice induced a heightened and prolonged induction of inflammatory modulators for both viruses, manifesting as a significant immune cell infiltrate and ocular disease. Moreover, while wild-type virus infection of Stat1(-/-) was always lethal, vhs-null infection was rarely lethal. There was a significant increase in Stat3- and interleukin-6 (IL-6)-dependent transcription in Stat1(-/-) mice, implicating the Stat3 and IL-6 pathways in the observed ocular pathology. Further, infected Stat1(-/-) mice showed phosphorylated Stat3 in the corneal epithelium. Our data show a role for vhs in evading innate host responses and a role for Stat1 in limiting virus infection and for facilitating an appropriate nonpathological inflammatory response.


Subject(s)
Cornea/virology , Herpesvirus 1, Human/immunology , Keratitis, Herpetic/immunology , STAT1 Transcription Factor/deficiency , Animals , Cornea/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Genomics , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions , Immunity, Innate , Inflammation/immunology , Inflammation/virology , Interleukin-6/immunology , Keratitis, Herpetic/pathology , Male , Mice , Oligonucleotide Array Sequence Analysis , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Virus Replication
8.
J Virol ; 82(12): 6052-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400863

ABSTRACT

Mice lacking the Stat1 interferon signaling gene were infected with herpes simplex virus type 1 (HSV-1) or an attenuated recombinant lacking virion host shutoff (Delta vhs). Delta vhs virus-infected Stat1(-/-) mice showed levels of replication equivalent to that of the wild-type virus-infected control mice but reduced relative to wild-type virus-infected Stat1(-/-) mice. Stat1 deficiency relieves the immunomodulatory deficiency of Delta vhs virus, but not its inherent growth defect. Also Vhs is dispensable for reactivation.


Subject(s)
STAT1 Transcription Factor/genetics , Simplexvirus/pathogenicity , Animals , Female , Male , Mice , Mice, Congenic , Mice, Knockout , Simplexvirus/physiology , Virulence , Virus Latency , Virus Replication
9.
J Virol ; 82(11): 5527-35, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367525

ABSTRACT

Herpes simplex virus mutants lacking the vhs protein are severely attenuated in animal models of pathogenesis and exhibit reduced growth in primary cell culture. As a result of these properties, viruses with vhs deleted have been proposed as live-attenuated vaccines. Despite these findings and their implications for vaccines, the mechanisms by which vhs promotes infection in cell culture and in vivo are not understood. In this study we demonstrate that vhs-deficient viruses replicate to reduced levels in interferon (IFN)-primed cells and that this deficit has both IFN-dependent and IFN-independent components. Furthermore, vhs-defective viruses induce increased and physiologically active levels of IFN, increased amounts of IFN-stimulated transcripts, and more phosphorylated eIF2alpha. In addition, we demonstrate greater accumulation of viral RNAs following infection with a vhs-deficient virus. This leads to the hypothesis that attenuation of viruses lacking vhs may be attributed to increased levels of double-stranded RNA, a potent pathogen-associated molecular pattern. Together these data show that vhs likely functions to reduce innate immune responses and thereby acts as a critical determinant of viral pathogenesis.


Subject(s)
Simplexvirus/genetics , Simplexvirus/metabolism , Virion/genetics , Virion/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling , Gene Expression Regulation , Interferon-alpha/pharmacology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Mice, Knockout , Mutation/genetics , Phosphorylation , RNA, Messenger/genetics , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Transcription, Genetic/genetics
10.
J Virol ; 80(15): 7600-12, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16840339

ABSTRACT

Herpes simplex virus type 1 (HSV-1) mutants lacking the ICP34.5 gene are severely attenuated in mouse models and have a significant growth defect in confluent mouse embryo fibroblasts. Previously, ICP34.5 was demonstrated to have a crucial role in evading the innate immune response to infection by mediating the dephosphorylation of eIF2alpha, a translation initiation factor phosphorylated by PKR during the antiviral response. To further understand the role of ICP34.5 in evasion of the antiviral response, we used transcriptional profiling to examine host cell gene expression in both wild-type and ICP34.5-null virus-infected mouse embryo fibroblasts over a time course of infection. Our study revealed that cells responded to infection within 3 h through PKR-dependent eIF2alpha phosphorylation and that the majority of up-regulated genes at 3 h postinfection were involved in the antiviral response. HSV-1 counters this response through early expression of ICP34.5 and dephosphorylation of eIF2alpha. By 12 h postinfection, the differences between the number and functional classification of genes differentially up- and down-regulated between wild-type and ICP34.5-null virus-infected cells were maximal. Specifically, in wild-type virus-infected cells, the majority of changed genes were involved in metabolic and biosynthetic processes, while in ICP34.5-null virus-infected cells, mostly antiviral genes were up-regulated. Further, ICP34.5-null virus-infected cells produced greater amounts of beta interferon than wild-type virus-infected cells. These results indicate that ICP34.5 expression and function at early times postinfection have a pivotal role in the ability of HSV-1 to gain control of the host cell and maintain an environment for successful viral replication.


Subject(s)
Fibroblasts/virology , Genomics , Herpes Simplex , Herpesvirus 1, Human/growth & development , Viral Proteins/physiology , Animals , Chlorocebus aethiops , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Phosphorylation , Signal Transduction , Vero Cells/metabolism , Vero Cells/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/physiology , eIF-2 Kinase/genetics , eIF-2 Kinase/physiology
11.
J Virol ; 79(2): 997-1007, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15613328

ABSTRACT

The cytoplasmic tails of all three major varicella-zoster virus (VZV) glycoproteins, gE, gH, and gB, harbor functional tyrosine-based endocytosis motifs that mediate internalization. The aim of the present study was to examine whether endocytosis from the plasma membrane is a cellular route by which VZV glycoproteins are delivered to the final envelopment compartment. In this study, we demonstrated that internalization of the glycoproteins occurred in the first 24 h postinfection but was reduced later in infection. Using surface biotinylation of VZV-infected cells followed by a glutathione cleavage assay, we showed that endocytosis was independent of antibody binding to gE, gH, and gB. Subsequently, with this assay, we demonstrated that biotinylated gE, gH, and gB retrieved from the cell surface were incorporated into nascent virus particles isolated after density gradient sedimentation. To confirm and extend this finding, we repeated the above sedimentation step and specifically detected envelopes decorated with Streptavidin-conjugated gold beads on a majority of complete virions through examination by transmission electron microscopy. In addition, a gE-gI complex and a gE-gH complex were found on the virions. Therefore, the above studies established that VZV subsumed a postendocytosis trafficking pathway as one mechanism by which to deliver viral glycoproteins to the site of virion assembly in the cytoplasm. Furthermore, since a recombinant VZV genome lacking only endocytosis-competent gE cannot replicate, these results supported the conclusion that the endocytosis-envelopment pathway is an essential component of the VZV life cycle.


Subject(s)
Endocytosis , Herpesvirus 3, Human/metabolism , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Virion/metabolism , Biotinylation , Cell Line, Tumor , Cell Membrane/metabolism , Herpesvirus 3, Human/ultrastructure , Humans , Microscopy, Electron
12.
J Virol ; 78(6): 2884-96, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990707

ABSTRACT

The gH glycoprotein of varicella-zoster virus (VZV) is a major fusogen. The realigned short cytoplasmic tail of gH (18 amino acids) harbors a functional endocytosis motif (YNKI) that mediates internalization in both VZV-infected and transfected cells (T. J. Pasieka, L. Maresova, and C. Grose, J. Virol. 77: 4194-4202, 2003). During subsequent confocal microscopy studies of endocytosis-deficient gH mutants, we observed that cells transfected with the gH tail mutants exhibited marked fusion. Therefore, we postulated that VZV gH endocytosis served to regulate cell-to-cell fusion. Subsequent analyses of gH+gL transfection fusion assays by the Kolmogorov-Smirnov statistical test demonstrated that expression of the endocytosis-deficient gH mutants resulted in a statistically significant enhancement of cell-to-cell fusion (P < 0.0001) compared to wild-type gH. On the other hand, coexpression of VZV gE, another endocytosis-competent VZV glycoprotein, was able to temper the fusogenicity of the gH endocytosis mutants by facilitating internalization of the mutant gH protein from the cell surface. When the latter results were similarly analyzed, there was no longer any enhanced fusion by the endocytosis-deficient gH mutant protein. In summary, these studies support a role for gH endocytosis in regulating the cell surface expression of gH and thereby regulating gH-mediated fusion. The data also confirm and extend prior observations of a gE-gH interaction during viral glycoprotein trafficking in a VZV transfection system.


Subject(s)
Cell Fusion , Endocytosis/physiology , Gene Expression Regulation, Viral , Herpesvirus 3, Human/pathogenicity , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Animals , Giant Cells , HeLa Cells , Herpesvirus 3, Human/physiology , Humans , Membrane Glycoproteins/genetics , Mice , Microscopy, Confocal , Mutation , Viral Envelope Proteins/genetics , Viral Proteins/genetics
13.
J Virol Methods ; 111(2): 157-61, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12880931

ABSTRACT

Fusion and syncytium formation induced by the expression of fusogenic viral glycoproteins are common cytopathic effects. Numerous laboratories have studied mechanisms of fusion, generally with each laboratory utilizing a different method to measure polykaryote formation. There is little consensus as to which methods are preferred for measuring and comparing fusion. The Kolmogorov-Smirnov two-sample test was used to document a statistical difference in syncytium formation between two forms of a varicella-zoster virus (VZV) fusogenic glycoprotein. This test is widely applied toward the analysis of large human populations in which interest centers on testing the hypothesis that the distributions of two sub-populations are identical. We suggest that the Kolmogorov-Smirnov test is a preferable method to assess whether differences in distribution of syncytia size between two fusogenic glycoproteins are meaningful statistically.


Subject(s)
Giant Cells/physiology , Herpesvirus 3, Human/pathogenicity , Membrane Fusion , Virology/statistics & numerical data , Cytopathogenic Effect, Viral , HeLa Cells , Humans , Statistics, Nonparametric , Viral Envelope Proteins/metabolism , Virology/methods
14.
J Virol ; 77(7): 4191-204, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12634377

ABSTRACT

The trafficking of varicella-zoster virus (VZV) gH was investigated under both infection and transfection conditions. In initial endocytosis assays performed in infected cells, the three glycoproteins gE, gI, and gB served as positive controls for internalization from the plasma membrane. Subsequently, we discovered that gH in VZV-infected cells was also internalized and followed a similar trafficking pattern. This observation was unexpected because all herpesvirus gH homologues have short endodomains not known to contain trafficking motifs. Further investigation demonstrated that VZV gH, when expressed alone with its chaperone gL, was capable of endocytosis in a clathrin-dependent manner, independent of gE, gI, or gB. Upon inspection of the short gH cytoplasmic tail, we discovered a putative tyrosine-based endocytosis motif (YNKI). When the tyrosine was replaced with an alanine, endocytosis of gH was blocked. Utilizing an endocytosis assay dependent on biotin labeling, we further documented that endocytosis of VZV gH was antibody independent. In control experiments, we showed that gE, gI, and gB also internalized in an antibody-independent manner. Alignment analysis of the VZV gH cytoplasmic tail to other herpesvirus gH homologues revealed two important findings: (i) herpes simplex virus type 1 and 2 homologues lacked an endocytosis motif, while all other alphaherpesvirus gH homologues contained a potential motif, and (ii) the VZV gH and simian varicella virus gH cytoplasmic tails were likely longer in length (18 amino acids) than predicted in the original sequence analyses (12 and 16 amino acids, respectively). The longer tails provided the proper context for a functional endocytosis motif.


Subject(s)
Herpesvirus 3, Human/physiology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Viral Proteins/chemistry , Viral Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Antibodies, Viral , Base Sequence , Clathrin/metabolism , DNA, Viral/genetics , Endocytosis , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/immunology , Kinetics , Membrane Glycoproteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Sequence Alignment , Transfection , Tyrosine/chemistry , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...