Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Vet Microbiol ; 245: 108707, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32456815

ABSTRACT

Feline viral rhinotracheitis is a prevalent disease among cats caused by feline herpesvirus 1 (FHV-1). microRNAs (miRNAs), which serve as important regulatory factors in the host, participate in the regulation of the host innate immune response to virus infection. However, the roles of miRNAs in the FHV-1 life cycle remain unclear. In this study, we found that a new miRNA, miR-101, could suppress FHV-1 replication. FHV-1 infection upregulated the expression level of miR-101 in a cGAS-dependent manner. Furthermore, miR-101 could significantly enhance type I interferon antiviral signaling by targeting suppressor of cytokine signaling 5 (SOCS5), a negative regulator of the JAK-STAT pathway. Likewise, knockdown of cellular SOCS5 also suppressed FHV-1 replication due to the enhancement of IFN-I-induced signaling cascades. Taken together, our data demonstrated a new strategy for miR-101-mediated defense against FHV-1 infection by enhancing IFN-I antiviral signaling and increased the knowledge of miRNAs regulating innate immune signaling pathways.


Subject(s)
Herpesviridae Infections/veterinary , Host-Pathogen Interactions , MicroRNAs/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Varicellovirus/physiology , Virus Replication , Animals , Cats , Cell Line , Herpesviridae Infections/virology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/immunology , Varicellovirus/pathogenicity
2.
Int J Med Sci ; 17(6): 745-750, 2020.
Article in English | MEDLINE | ID: mdl-32218696

ABSTRACT

Both the herpes zoster virus and suid herpesvirus type 1 (SuHV-1) belong to the Varicellovirus genus of the α-herpesviridae subfamily. They may cause opportunistic infections especially in patients with kidney diseases, varying from latent illness to overt lethality. Under these circumstances, impaired renal function is both the culprit for and victim of the infection. However, fulminant eruption of severe skin herpes zoster in lupus nephritis (LN) patients under prolonged immunosuppressive therapy is rare and even more rarely seen is the SuHV-1 encephalitis in human. Facing the evolution of these rare infections, we hence chose to review the clinical pathogenicity of these two viruses which were cognate in origin but distinct in virulence. As such, we began with the first of the two above viral diseases and proceeded with peculiar renal involvement, unique clinical symptoms and pertinent lethal risk. Of importance, LN was used to exemplify the reciprocally detrimental interactions between impaired renal function and suppressed immune response. Then in a manner similar to the gradient overlay, SuHV-1 encephalitis was discussed focusing on its neurotropic features, specific MRI findings and exclusive test of high throughput sequencing. Our report highlighted novel presentations of the Varicellovirus genus infection by providing a productive multidisciplinary communication with pointed disclosure of the renal involvement. It may therefore be of great medical relevance and educational value for clinicians, especially the unseasoned ones, to foresee and manage similar cases in susceptible patients.


Subject(s)
Herpes Zoster/epidemiology , Herpesvirus 1, Suid/pathogenicity , Infectious Encephalitis/epidemiology , Kidney Diseases/epidemiology , Animals , Herpes Zoster/complications , Herpes Zoster/genetics , Herpes Zoster/virology , Herpesviridae Infections/complications , Herpesviridae Infections/epidemiology , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Infectious Encephalitis/complications , Infectious Encephalitis/genetics , Infectious Encephalitis/virology , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Diseases/virology , Lupus Nephritis/complications , Lupus Nephritis/epidemiology , Lupus Nephritis/genetics , Lupus Nephritis/virology , Opportunistic Infections/complications , Opportunistic Infections/epidemiology , Opportunistic Infections/genetics , Opportunistic Infections/virology , Swine/virology , Varicellovirus/pathogenicity
3.
Virus Genes ; 56(1): 49-57, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31776852

ABSTRACT

Feline herpesvirus type 1 (FHV-1) is a widespread cause of respiratory and ocular disease in domestic cats. A spectrum of disease severity is observed in host animals, but there has been limited prior investigation into viral genome factors which could be responsible. Stocks of FHV-1 were established from oropharyngeal swabs obtained from twenty-five cats with signs of infection housed in eight animal shelters around the USA. A standardized numerical host clinical disease severity scoring scheme was used for each cat from which an isolate was obtained. Illumina MiSeq was used to sequence the genome of each isolate. Genomic homogeneity among isolates was relatively high. A general linear model for fixed effects determined that only two synonymous single nucleotide polymorphisms across two genes (UL37/39) in the same isolate (from one host animal with a low disease severity score) were significantly associated (p ≤ 0.05) with assigned host respiratory and total disease severity score. No variants in any isolate were found to be significantly associated with assigned host ocular disease severity score. A concurrent analysis of missense mutations among the viral isolates identified three genes as being primarily involved in the observed genomic variation, but none were significantly associated with host disease severity scores. An ancestral state likelihood reconstruction was performed and determined that there was no evidence of a connection between host disease severity score and viral evolutionary state. We conclude from our results that the spectrum of host disease severity observed with FHV-1 is unlikely to be primarily related to viral genomic variations, and is instead due to host response and/or other factors.


Subject(s)
Cat Diseases/virology , Herpesviridae Infections/veterinary , Varicellovirus/genetics , Varicellovirus/pathogenicity , Animals , Cats , Female , Genome, Viral , Genomics , Herpesviridae Infections/virology , Male , Mutation , Phylogeny , Polymorphism, Single Nucleotide , Varicellovirus/classification , Varicellovirus/isolation & purification , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Vet Pathol ; 54(6): 922-932, 2017 11.
Article in English | MEDLINE | ID: mdl-28812530

ABSTRACT

Feline herpesvirus type 1 (FeHV-1) is one of the etiological agents of feline respiratory disease. FeHV-1 is an epitheliotropic and cytopathic virus that mainly causes rhinitis and conjunctivitis, although pneumonia is also occasionally seen. In this study, the authors investigated the pathogenesis of FeHV-1-associated pneumonia, comparing natural cases with viral infection of tracheal ring and cell cultures in vitro, using histology, immunohistology, double immunofluorescence, and transmission electron microscopy as investigative tools. The results confirm that FeHV-1 targets both respiratory epithelial cells and pneumocytes and indicate that FeHV-1 pneumonia is the consequence of continuous cell-to-cell viral spread from the upper airways via the trachea into the lungs. They provide strong evidence that FeHV-1-infected cells die primarily via apoptosis, following loss of cell-to-cell contact, rounding, and detachment. However, virus-induced lesions in vivo are dominated by marked neutrophil infiltration and extensive necrosis with less prominent apoptosis; in the airways, the tissue necrosis can extend into the submucosa. The necrosis appears to result from virus-induced neutrophil influx and release of proteolytic enzymes, such as matrix metalloproteinase-9, from the neutrophils.


Subject(s)
Cat Diseases/virology , Herpesviridae Infections/veterinary , Pneumonia, Viral/veterinary , Varicellovirus/pathogenicity , Animals , Apoptosis , Cat Diseases/pathology , Cats , Disease Models, Animal , Female , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Lung/pathology , Lung/virology , Male , Mice , Necrosis/pathology , Neutrophils/pathology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Trachea/pathology , Trachea/virology
5.
J Vet Med Sci ; 79(8): 1426-1436, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28717112

ABSTRACT

This study aimed to investigate the neuropathogenesis of equine herpes virus 9 (EHV-9) by studying the effects of a single point mutation introduced in two different EHV-9 genes. The two EHV-9 mutants, 14R and 19R, were generated carrying a point mutation in two separate EHV-9 genes. These mutants, along with the wild-type EHV-9, were used to infect a hamster model. The EHV-9- and 19R-infected groups showed earlier and more severe clinical signs of infection than the 14R-infected group. The white blood cells (WBCs) count was significantly increased in both EHV-9- and 19R-infected groups compared to the 14R-infected group at the 4th day post infection (DPI). Viremia was also detected earlier in both EHV-9- and 19R-infected groups than 14R-infected group. There were differences in the anterograde transmission pattern of both EHV-9 and 19R compared to 14R inside the brain. Serum TNF-α, IL-6 and IFN-γ levels were significantly increased in both EHV-9- and 19R-infected groups compared to the 14R-infected group. Histopathological and immunohistochemical analyses revealed that the mean group scores for the entire brain were significantly higher in both EHV-9- and 19R- infected groups than 14R-infected group. Collectively, these results confirm that the gene product of Open Reading Frame 19 (ORF19) plays an important role in EHV-9 neuropathogenicity and that the mutation in ORF19 is responsible for the attenuation of EHV-9.


Subject(s)
Central Nervous System Infections/virology , Herpesviridae Infections/virology , Point Mutation , Varicellovirus/genetics , Animals , Cricetinae , Male , Varicellovirus/pathogenicity
6.
J Virol ; 90(23): 10823-10843, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27681124

ABSTRACT

Primary infection with varicella-zoster virus (VZV), a neurotropic alphaherpesvirus, results in varicella. VZV establishes latency in the sensory ganglia and can reactivate later in life to cause herpes zoster. The relationship between VZV and its host during acute infection in the sensory ganglia is not well understood due to limited access to clinical specimens. Intrabronchial inoculation of rhesus macaques with simian varicella virus (SVV) recapitulates the hallmarks of VZV infection in humans. We leveraged this animal model to characterize the host-pathogen interactions in the ganglia during both acute and latent infection by measuring both viral and host transcriptomes on days postinfection (dpi) 3, 7, 10, 14, and 100. SVV DNA and transcripts were detected in sensory ganglia 3 dpi, before the appearance of rash. CD4 and CD8 T cells were also detected in the sensory ganglia 3 dpi. Moreover, lung-resident T cells isolated from the same animals 3 dpi also harbored SVV DNA and transcripts, suggesting that T cells may be responsible for trafficking SVV to the ganglia. Transcriptome sequencing (RNA-Seq) analysis showed that cessation of viral transcription 7 dpi coincides with a robust antiviral innate immune response in the ganglia. Interestingly, a significant number of genes that play a critical role in nervous system development and function remained downregulated into latency. These studies provide novel insights into host-pathogen interactions in the sensory ganglia during acute varicella and demonstrate that SVV infection results in profound and sustained changes in neuronal gene expression. IMPORTANCE: Many aspects of VZV infection of sensory ganglia remain poorly understood, due to limited access to human specimens and the fact that VZV is strictly a human virus. Infection of rhesus macaques with simian varicella virus (SVV), a homolog of VZV, provides a robust model of the human disease. Using this model, we show that SVV reaches the ganglia early after infection, most likely by T cells, and that the induction of a robust innate immune response correlates with cessation of virus transcription. We also report significant changes in the expression of genes that play an important role in neuronal function. Importantly, these changes persist long after viral replication ceases. Given the homology between SVV and VZV, and the genetic and physiological similarities between rhesus macaques and humans, our results provide novel insight into the interactions between VZV and its human host and explain some of the neurological consequences of VZV infection.


Subject(s)
Ganglia, Sensory/metabolism , Ganglia, Sensory/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Varicellovirus/pathogenicity , Acute Disease , Animals , Axonal Transport , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chickenpox/virology , DNA, Viral/genetics , DNA, Viral/metabolism , Disease Models, Animal , Ganglia, Sensory/immunology , Gene Expression , Herpesviridae Infections/immunology , Herpesvirus 3, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Macaca mulatta , Neurogenesis , Varicellovirus/genetics , Varicellovirus/physiology , Virus Replication
7.
J Pathol ; 235(2): 298-311, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25255989

ABSTRACT

Varicelloviruses in primates comprise the prototypic human varicella-zoster virus (VZV) and its non-human primate homologue, simian varicella virus (SVV). Both viruses cause varicella as a primary infection, establish latency in ganglionic neurons and reactivate later in life to cause herpes zoster in their respective hosts. VZV is endemic worldwide and, although varicella is usually a benign disease in childhood, VZV reactivation is a significant cause of neurological disease in the elderly and in immunocompromised individuals. The pathogenesis of VZV infection remains ill-defined, mostly due to the species restriction of VZV that impedes studies in experimental animal models. SVV infection of non-human primates parallels virological, clinical, pathological and immunological features of human VZV infection, thereby providing an excellent model to study the pathogenesis of varicella and herpes zoster in its natural host. In this review, we discuss recent studies that provided novel insight in both the virus and host factors involved in the three elementary stages of Varicellovirus infection in primates: primary infection, latency and reactivation.


Subject(s)
Herpesviridae Infections/virology , Primates , Varicellovirus/pathogenicity , Animals , Biopsy , Disease Models, Animal , Genotype , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Host-Pathogen Interactions , Humans , Pathology, Molecular/methods , Predictive Value of Tests , Varicellovirus/genetics , Varicellovirus/immunology , Virology/methods , Virulence , Virus Activation , Virus Latency
8.
Vet Pathol ; 51(3): 581-90, 2014 May.
Article in English | MEDLINE | ID: mdl-23804999

ABSTRACT

This study aimed to clarify the timing and infectivity of equine herpesvirus 9 (EHV-9) infection in BALB/c-nu/nu mice and their immunocompetent counterpart (BALB/c). Following intranasal inoculation with 10(5) PFU of EHV-9, specimens from 8 mice per group were collected at different times postinoculation (PI) and assessed using histopathology, immunohistochemistry for viral antigen, and quantitative real-time polymerase chain reaction for ORF30 gene expression. In BALB/c-nu/nu mice, EHV-9 antigen was abundant in olfactory epithelia of all inoculated animals, and in the olfactory bulb of 1 animal. In contrast, only 1 BALB/c mouse per time point had rhinitis, with mild to moderate immunopositivity starting from 12 to 48 h PI, followed by a gradual virus clearance at 72 h PI. Statistically, significant differences were noted in the immunohistochemistry reactions between the 2 mouse strains, indicating that BALB/c-nu/nu is more susceptible to infection. Relative expression levels of ORF30 gene in olfactory epithelia were significantly different between the 2 groups, with the exception of 12 h PI, when BALB/c-nu/nu animals showed dramatic increases in ORF30 gene expression level until 48 h PI, followed by a decline in expression level until the end of experiment. In contrast, the expression level in brains showed no differences between mouse strain except at 96 h PI. In both strains, the highest messenger RNA expression was detected at 48 h PI, followed by a decline in BALB/c mice, proving a rapid clearance of virus in BALB/c and a gradual slowing down of the increased expression levels in BALB/c-nu/nu.


Subject(s)
Disease Susceptibility/pathology , Herpesviridae Infections/veterinary , Mice, Inbred BALB C , Mice, Nude , Rodent Diseases/metabolism , Rodent Diseases/virology , Varicellovirus/pathogenicity , Administration, Intranasal , Animals , Antigens, Viral/metabolism , Cattle , Cell Line , DNA Primers/genetics , Herpesviridae Infections/metabolism , Immunohistochemistry/veterinary , Mice , Olfactory Mucosa/virology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
9.
Virol J ; 10: 278, 2013 Sep 08.
Article in English | MEDLINE | ID: mdl-24010815

ABSTRACT

BACKGROUND: Varicella zoster virus (VZV) is a neurotropic alphaherpesvirus that infects humans and results in chickenpox and herpes zoster. A number of VZV genes remain functionally uncharacterized and since VZV is an obligate human pathogen, rigorous evaluation of VZV mutants in vivo remains challenging. Simian varicella virus (SVV) is homologous to VZV and SVV infection of rhesus macaques (RM) closely mimics VZV infection of humans. Recently the SVV genome was cloned as a bacterial artificial chromosome (BAC) and BAC-derived SVV displayed similar replication kinetics as wild-type (WT) SVV in vitro. METHODS: RMs were infected with BAC-derived SVV or WT SVV at 4x10(5) PFU intrabronchially (N=8, 4 per group, sex and age matched). We collected whole blood (PBMC) and bronchoalveolar lavage (BAL) at various days post-infection (dpi) and sensory ganglia during latent infection (>84 dpi) at necropsy and compared disease progression, viral replication, immune response and the establishment of latency. RESULTS: Viral replication kinetics and magnitude in bronchoalveolar lavage cells and whole blood as well as rash severity and duration were similar in RMs infected with SVV BAC or WT SVV. Moreover, SVV-specific B and T cell responses were comparable between BAC and WT-infected animals. Lastly, we measured viral DNA in sensory ganglia from both cohorts of infected RMs during latent infection. CONCLUSIONS: SVV BAC is as pathogenic and immunogenic as WT SVV in vivo. Thus, the SVV BAC genetic system combined with the rhesus macaque animal model can further our understanding of viral ORFs important for VZV pathogenesis and the development of second-generation vaccines.


Subject(s)
Chickenpox/pathology , Chickenpox/virology , Chromosomes, Artificial, Bacterial , Varicellovirus/genetics , Varicellovirus/pathogenicity , Animals , Blood/virology , Bronchoalveolar Lavage Fluid/virology , Disease Models, Animal , Ganglia, Sensory/virology , Macaca mulatta , Virus Latency
10.
J Cell Biochem ; 114(12): 2809-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23836554

ABSTRACT

Caprine herpesvirus type 1 (CpHV-1), like other members of the alpha subfamily of herpesviruses, establishes latent infections in trigeminal ganglion neurons. Our groups previously demonstrated that CpHV-1 induces apoptosis in goat peripheral blood mononuclear cells and in an epithelial bovine cell line, but the ability of CpHV-1 to induce apoptosis in neuronal cells remains unexplored. In this report, the susceptibility of Neuro 2A cells to infection by CpHV-1 was examined. Following infection of cultured cells with CpHV-1, expression of cell death genes was evaluated using real-time PCR and Western blot assays. Analysis of virus-infected cells revealed activation of caspase-8, a marker for the extrinsic pathway of apoptosis, and caspase-9, a marker for the intrinsic pathway of apoptosis at 12 and 24 h post-infection. Significant increase in the levels of cleaved caspase-3 was also observed at the acme of cytopathic effect at 24 h post-infection. In particular, at 3 and 6 h post-infection, several proapototic genes were under-expressed. At 12 h post-infection several proapototic genes such as caspases, TNF, Cd70, and Traf1 were over expressed while Bcl2a1a, Fadd, and TNF genes were underexpressed. In conclusion, the simultaneous activation of caspase-8 and caspase-9 suggests that CpHV-1 can trigger the death-receptor pathway and the mitochondrial pathway separately and in parallel. Our findings are significant because this is the first published study showing the effect of CpHV-1 infection in neuronal cells in terms of gene expression and apoptosis modulation.


Subject(s)
Apoptosis/genetics , Herpesviridae Infections/genetics , Neurons/metabolism , Varicellovirus/genetics , Animals , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cattle , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression Regulation , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Leukocytes, Mononuclear/metabolism , Neurons/pathology , Neurons/virology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Varicellovirus/pathogenicity
11.
Vet Pathol ; 50(1): 56-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22508701

ABSTRACT

By using a new member of the neurotropic equine herpesviruses, EHV-9, which induced encephalitis in various species via various routes, an ocular infection model was developed in suckling hamsters. The suckling hamsters were inoculated with EHV-9 via the conjunctival route and were sacrificed after 6, 12, 24, 36, 48, 72, 96, 120, and 144 hours (h) post inoculation (PI). Three horizontal sections of the brains, including the eyes and cranial cavity, were examined histologically to assess the viral kinetics and time-course neuropathological alterations using a panoramic view. At 6 to 24 h PI, there were various degrees of necrosis in the conjunctival epithelial cells, as well as frequent mononuclear cell infiltrations in the lamina propria and the tarsus of the eyelid, and frequent myositis of the eyelid muscles. At 96 h PI, encephalitis was observed in the brainstem at the level of the pons and cerebellum. EHV-9 antigen immunoreactivity was detected in the macrophages circulating in the eyelid and around the fine nerve endings supplying the eyelid, the nerves of the extraocular muscles, and the lacrimal glands from 6 h to 144 h PI. At 96 h PI, the viral antigen immunoreactivity was detected in the brainstem at the level of the pons and cerebellum. These results suggest that EHV-9 invaded the brain via the trigeminal nerve in addition to the abducent, oculomotor, and facial nerves. This conjunctival EHV-9 suckling hamster model may be useful in assessing the neuronal spread of neuropathogenic viruses via the eyes to the brain.


Subject(s)
Disease Models, Animal , Encephalitis, Viral/veterinary , Eye Infections, Viral/veterinary , Herpesviridae Infections/veterinary , Horse Diseases/virology , Varicellovirus/pathogenicity , Animals , Animals, Suckling , Antigens, Viral/analysis , Brain/pathology , Brain/virology , Conjunctiva/pathology , Cricetinae , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Eye/pathology , Eye/virology , Eye Infections, Viral/pathology , Eye Infections, Viral/virology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Horse Diseases/pathology , Horses , Immunohistochemistry , Kinetics , Mesocricetus , Necrosis , Time Factors , Trigeminal Nerve/virology , Varicellovirus/immunology
12.
Braz. j. microbiol ; 43(4): 1632-1640, Oct.-Dec. 2012. ilus, tab
Article in English | LILACS | ID: lil-665851

ABSTRACT

Suid herpesvirus 1 (SuHV-1) is the causative agent of pseudorabies (PR), a disease of great importance due to the huge losses it causes in the swine industry. The aim of this study was to determine a method for genotyping SuHV-1 based on partial sequences of the gene coding for glycoprotein C (gC) and to elucidate the possible reasons for the variability of this region. A total of 109 gCsequences collected from GenBank were divided into five major groups after reconstruction of a phylogenetic tree by Bayesian inference. The analysis showed that a portion of gC (approximately 671 bp) is under selective pressure at various points that coincide with regions of protein disorder. It was also possible to divide SuHV-1 into five genotypes that evolved under different selective pressures. These genotypes are not specific to countries or continents, perhaps due to multiple introduction events related to the importation of swine.


Subject(s)
Animals , Genetic Variation , Glycoproteins/genetics , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/pathogenicity , Pseudorabies/genetics , Base Sequence/genetics , Varicellovirus/genetics , Varicellovirus/pathogenicity , Genetics, Microbial , Genotype , Methods , Virulence
13.
J Comp Pathol ; 146(2-3): 211-22, 2012.
Article in English | MEDLINE | ID: mdl-21741053

ABSTRACT

The pathogenesis and kinetics of oral infection by equine herpesvirus (EHV)-9 were studied in mice and hamsters. After oral inoculation of 10(5) plaque-forming units (PFU) of virus, 1-week-old suckling hamsters showed varying severity of neurological disease from 72 hours post inoculation (hpi) and all of these animals had died by 96 hpi. Four-week-old ICR mice inoculated orally with 4 × 10(4)PFU of virus showed no clinical signs, but they developed erosive and ulcerative gastritis from 36 hpi. Varying degrees of encephalitis were seen in infected mice and hamsters, and the hamsters also developed myelitis by 96 hpi. Immunohistochemistry performed on whole body sections of suckling hamsters revealed the kinetics of spread of the virus to the central nervous system. EHV-9 antigen was detected initially in macrophages of the oral and lingual submucosa. At 36 hpi virus antigen was detected in the nerve fibres and pseudounipolar neurons of the trigeminal ganglion and at 96 hpi antigen was present in the myenteric plexuses of the intestine. Virus antigen was also detected in the liver, lungs and heart of affected animals. EHV-9 DNA was detected by polymerase chain reaction in the brain, blood and spinal cord of suckling hamsters at 36, 48 and 96 hpi. These findings show that EHV-9 may spread via the trigeminal nerve when mice and hamsters are inoculated orally with virus.


Subject(s)
Brain/virology , Encephalitis, Viral/virology , Herpesviridae Infections/virology , Varicellovirus/pathogenicity , Animals , Antigens, Viral , Brain/pathology , Cricetinae , Encephalitis, Viral/pathology , Herpesviridae Infections/pathology , Mesocricetus , Mice
14.
Vet Pathol ; 48(3): 558-64, 2011 May.
Article in English | MEDLINE | ID: mdl-20574073

ABSTRACT

The infectivity and pathology of equine herpesvirus 9 (EHV-9), a new neurotropic equine herpesvirus isolated from gazelles, was studied in hamsters experimentally infected via nasal, ocular, oral, intravenous (IV), or peritoneal routes. Clinically, all animals inoculated by the nasal route and ~25% inoculated by the oral and peritoneal routes showed neurological signs on days 3, 6, and 9 postinoculation (PI), respectively. Neurological signs were not observed in animals administered EHV-9 by the IV and ocular routes. With the exception of animals administered EHV-9 by the IV route, all infected animals had lymphocytic meningoencephalitis. Although there were a number of differences in the severity and distribution of the lesions depending on the route of inoculation, the basic features of lymphocytic meningoencephalitis caused by EHV-9 were common. Lesions consisted of neuronal necrosis, perivascular aggregates of lymphocytes, plasma cells, and neutrophils, gliosis, intranuclear inclusion bodies, and diffuse lymphocytic infiltrates in the meninges. Viral antigen was detected in degenerated neurons in infected animals inoculated by the nasal, ocular, oral, and peritoneal routes. The distribution of EHV-9 antigen was somewhat dependent on inoculation route. There were no microscopic abnormalities or viral antigen in animals treated by the IV route. This study provides new data about experimental EHV-9 infection in hamsters through routes other than the IV route. These results suggest that in the animals infected by the oral, ocular, and peritoneal routes, EHV-9 might travel to the brain through nerves, other than by the olfactory route, after initial propagation at the site of viral entry.


Subject(s)
Herpesviridae Infections/virology , Meningoencephalitis/virology , Varicellovirus/pathogenicity , Animals , Cricetinae , Herpesviridae Infections/pathology , Male , Meningoencephalitis/pathology , Mesocricetus , Varicellovirus/classification
15.
PLoS Pathog ; 5(11): e1000657, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19911054

ABSTRACT

Simian varicella virus (SVV), the etiologic agent of naturally occurring varicella in primates, is genetically and antigenically closely related to human varicella zoster virus (VZV). Early attempts to develop a model of VZV pathogenesis and latency in nonhuman primates (NHP) resulted in persistent infection. More recent models successfully produced latency; however, only a minority of monkeys became viremic and seroconverted. Thus, previous NHP models were not ideally suited to analyze the immune response to SVV during acute infection and the transition to latency. Here, we show for the first time that intrabronchial inoculation of rhesus macaques with SVV closely mimics naturally occurring varicella (chickenpox) in humans. Infected monkeys developed varicella and viremia that resolved 21 days after infection. Months later, viral DNA was detected only in ganglia and not in non-ganglionic tissues. Like VZV latency in human ganglia, transcripts corresponding to SVV ORFs 21, 62, 63 and 66, but not ORF 40, were detected by RT-PCR. In addition, as described for VZV, SVV ORF 63 protein was detected in the cytoplasm of neurons in latently infected monkey ganglia by immunohistochemistry. We also present the first in depth analysis of the immune response to SVV. Infected animals produced a strong humoral and cell-mediated immune response to SVV, as assessed by immunohistology, serology and flow cytometry. Intrabronchial inoculation of rhesus macaques with SVV provides a novel model to analyze viral and immunological mechanisms of VZV latency and reactivation.


Subject(s)
Disease Models, Animal , Herpesviridae Infections , Macaca mulatta/virology , Varicellovirus/pathogenicity , Animals , B-Lymphocytes/immunology , Cell Proliferation , DNA, Viral/analysis , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Herpesvirus 3, Human , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Varicellovirus/genetics , Viral Load
16.
Comp Med ; 59(5): 482-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19887033

ABSTRACT

Simian varicella virus (SVV; Cercopithecine herpesvirus 9) is a naturally occurring herpesvirus of nonhuman primates. Here we present the clinical, pathologic, and virologic findings from 2 cases of SVV in adult female pigtailed macaques (Macaca nemestrina). The initial case presented with hyperthermia and a diffuse inguinal rash which spread centripetally, progressing to vesiculoulcerative dermatitis of the trunk, face, and extremities. At 96 h after presentation, the animal was anorexic and lethargic and had oral and glossal ulcerations. Euthanasia was elected in light of the macaque's failure to respond to clinical treatment. Seven days after the first case was identified, a second macaque presented with a vesicular rash and was euthanized. Gross necropsy lesions for both cases included vesicular, ulcerative dermatitis with mucocutaneous extension and hepatic necrosis; the initial case also demonstrated necrohemorrhagic gastroenterocolitis and multifocal splenic necrosis. Histology confirmed herpetic viral infection with abundant intranuclear inclusion bodies. Immunofluorescence assays detected antibodies specific for SVV. PCR assays of vesicular fluid, tissue, and blood confirmed SVV and excluded varicella-zoster virus (Human herpesvirus 3). Serology for Macacine herpesvirus 1 (formerly Cercopithecine herpesvirus 1), poxvirus (monkeypox), and rubella was negative. Banked serum samples confirmed SVV exposure and seroconversion. Investigation into the epidemiology of the seroconversion demonstrated a SVV colony prevalence of 20%. The described cases occurred in animals with reconstituted immune systems (after total-body irradiation) and demonstrate the clinical effects of infection with an endemic infectious agent in animals with a questionable immune status.


Subject(s)
Herpesviridae Infections/veterinary , Monkey Diseases/virology , Varicellovirus/pathogenicity , Animals , Antibodies, Viral/blood , Dermatitis/pathology , Dermatitis/veterinary , Dermatitis/virology , Enterocolitis/pathology , Enterocolitis/veterinary , Enterocolitis/virology , Fatal Outcome , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Intranuclear Inclusion Bodies/pathology , Intranuclear Inclusion Bodies/virology , Liver/pathology , Liver/virology , Macaca nemestrina , Monkey Diseases/immunology , Monkey Diseases/pathology , Necrosis , RNA, Viral/analysis , Seroepidemiologic Studies , Spleen/pathology , Spleen/virology
17.
Virus Res ; 145(2): 321-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19699769

ABSTRACT

Cervid herpesvirus 2 (CvHV2) has never been isolated from reindeer in Norway, but serological data and investigations by PCR indicate that the virus is endemic in the country, with horizontal and vertical transmission, systemic spread, and latency in the trigeminal ganglion. In this study two seropositive reindeer, one of which was pregnant, were administered dexamethasone, to reactivate CvHV2 latent infection. One control animal received sterile water. All animals including the control reactivated, as shown by amplification of CvHV2 DNA from nasal swabs. The pregnant animal showed lesions in the lip mucosa 10 days after the first dexamethasone injection and CvHV2 was visualized by electron microscopy and isolated from those lesions, as well as from nasal and vaginal swabs. On day 13 she aborted and CvHV2 was isolated from both the aborted calf and the mother. CvHV2 was isolated from the other animal administered dexamethasone. Despite amplification of viral DNA in the control animal, it was never possible to isolate the virus. Molecular characterization of the new isolates confirmed these to be CvHV2, and similar to the previous known strain Salla82. Present results represent the first isolation of CvHV2 in Norway and reconfirm that this virus can cause systemic infections in reindeer even after reactivation episodes, and infect the fetus in utero despite a prompt immune response. While it is not possible to atribute the abortion to CvHV2 alone, present data together with previous reports of vertical transmission of CvHV2 and neonatal death, point to an abortogenic potential, which should be further investigated.


Subject(s)
Abortion, Veterinary/virology , Herpesviridae Infections/veterinary , Varicellovirus/pathogenicity , Viremia/veterinary , Virus Activation , Aborted Fetus/virology , Animals , DNA, Viral/chemistry , DNA, Viral/genetics , Dexamethasone/administration & dosage , Female , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Immunologic Factors/administration & dosage , Lip/pathology , Lip/virology , Male , Microscopy, Electron , Molecular Sequence Data , Nasal Cavity/virology , Norway , Phylogeny , Pregnancy , Reindeer , Sequence Analysis, DNA , Sequence Homology , Vagina/virology , Varicellovirus/isolation & purification , Viremia/virology , Virion/ultrastructure
18.
Virus Res ; 145(2): 227-35, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19619589

ABSTRACT

It is known that Caprine Herpesvirus 1 (CpHV-1) causes apoptosis in mitogen-stimulated as well as not stimulated caprine peripheral blood mononuclear cells (PBMC). Initial experiments in Madin Darby bovine kidney (MDBK) cells revealed that CpHV-1 infection induced apoptotic features like chromatin condensation and DNA laddering. Thus, to characterize in more detail this apoptotic process, activation of caspase-8, -9 and -3 in MDBK cells CpHV-1 infected was investigated and demonstrated. In addition, CpHV-1 infection resulted in disruption of mitochondrial membrane potential, cytochrome c release and alterations in the pro- and anti-apoptotic proteins of Bcl-2 family. Proteolytic cleavage of poly(ADP-ribose) polymerases (PARP), confirming the activation of downstream caspases, was also observed. Our data indicated that a "cross-talk" between the death-receptor (extrinsic) pathway and the mitochondrial (intrinsic) pathway occurred in CpHV-1-induced apoptosis in vitro.


Subject(s)
Apoptosis , Varicellovirus/pathogenicity , Animals , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cattle , Cell Line , Cytochromes c/metabolism , Membrane Potential, Mitochondrial , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation
19.
J Virol ; 83(18): 9273-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19605493

ABSTRACT

Simian varicella virus (SVV) causes varicella in primates, becomes latent in ganglionic neurons, and reactivates to produce zoster. SVV produces a cytopathic effect in monkey kidney cells in tissue culture. To study the mechanism by which SVV-infected cells die, we examined markers of apoptosis 24 to 64 h postinfection (hpi). Western blot analysis of virus-infected cell lysates revealed a significant increase in the levels of the cleaved active form of caspase-3, accompanied by a parallel increase in caspase-3 activity at 40 to 64 hpi. Caspase-9, a marker for the intrinsic pathway, was activated significantly in SVV-infected cells at all time points, whereas trace levels of the active form of caspase-8, an extrinsic pathway marker, was detected only at 64 hpi. Bcl-2 expression at the mRNA and protein levels was decreased by 50 to 70% throughout the course of virus infection. Release of cytochrome c, an activator of caspase-9, from mitochondria into the cytoplasm was increased by 200% at 64 hpi. Analysis of Vero cells infected with SVV expressing green fluorescent protein (SVV-GFP) at 64 hpi revealed colocalization of the active forms of caspase-3 and caspase-9 and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining with GFP. A significant decrease in the bcl-2 mRNA levels along with an abundance of mRNA specific for SVV genes 63, 40, and 21 was seen in the fraction of Vero cells that were infected with SVV-GFP. Together, these findings indicate that SVV induces apoptosis in cultured Vero cells through the intrinsic pathway in which Bcl-2 is downregulated.


Subject(s)
Apoptosis , Kidney/virology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Varicellovirus/pathogenicity , Animals , Caspases/metabolism , Chickenpox/pathology , Chlorocebus aethiops , Down-Regulation/genetics , Haplorhini , Kidney/pathology , RNA, Messenger/analysis , Time Factors , Vero Cells
20.
Vet Microbiol ; 138(3-4): 266-72, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19411146

ABSTRACT

Enzyme-linked immunosorbent assays (ELISAs) were developed to detect caprine herpervirus-1 (CpHV-1)-specific IgG1 and IgG2 in sera from 43 naturally infected goats. The analysis of the IgG subclasses showed a dual pattern of distribution in seropositive goats with a major group of animals (36 out of 43) exhibiting significantly higher levels of IgG2 over IgG1 and a minor group (7 out of 43) possessing equal levels of IgG1 and IgG2. Four goats were experimentally infected with a virulent CpHV-1 Ba.1 strain by the intranasal or the intravaginal route and the kinetics of appearance of CpHV-1-specific IgG, IgG1 and IgG2 in the serum were studied. Two weeks following infection, both IgG1 and IgG2 levels increased although convalescent sera (i.e., collected 5-8 weeks post-infection) showed a clear prevalence of the IgG2 subclass. To determine the contribution of the different IgG subclasses to herpesvirus immunity, serum neutralization (SN) assays were performed in both naturally and experimentally infected goats. The kinetics of SN showed that neutralization activity was mainly associated to the IgG1 subclass and this was also confirmed in naturally infected goats. The results are discussed from the standpoint that the profile of the IgG subclasses is instrumental to study immune responses to CpHV-1 and that vaccination strategies may benefit from this information.


Subject(s)
Goat Diseases/blood , Immunoglobulin G/blood , Varicellovirus/immunology , Animals , Female , Goat Diseases/immunology , Goat Diseases/virology , Goats , Immunoglobulin G/classification , Male , Varicellovirus/pathogenicity , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...