Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 16(6): e1008597, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32511265

RESUMO

During infection of neurons by alphaherpesviruses including Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) viral nucleocapsids assemble in the cell nucleus, become enveloped in the cell body then traffic into and down axons to nerve termini for spread to adjacent epithelia. The viral membrane protein US9p and the membrane glycoprotein heterodimer gE/gI play critical roles in anterograde spread of both HSV-1 and PRV, and several models exist to explain their function. Biochemical studies suggest that PRV US9p associates with the kinesin-3 motor KIF1A in a gE/gI-stimulated manner, and the gE/gI-US9p complex has been proposed to recruit KIF1A to PRV for microtubule-mediated anterograde trafficking into or along the axon. However, as loss of gE/gI-US9p essentially abolishes delivery of alphaherpesviruses to the axon it is difficult to determine the microtubule-dependent trafficking properties and motor-composition of Δ(gE/gI-US9p) particles. Alternatively, studies in HSV-1 have suggested that gE/gI and US9p are required for the appearance of virions in the axon because they act upstream, to help assemble enveloped virions in the cell body. We prepared Δ(gE/gI-US9p) mutant, and control parental PRV particles from differentiated cultured neuronal or porcine kidney epithelial cells and quantitated the efficiency of virion assembly, the properties of microtubule-dependent transport and the ability of viral particles to recruit kinesin motors. We find that loss of gE/gI-US9p has no significant effect upon PRV particle assembly but leads to greatly diminished plus end-directed traffic, and enhanced minus end-directed and bidirectional movement along microtubules. PRV particles prepared from infected differentiated mouse CAD neurons were found to be associated with either kinesin KIF1A or kinesin KIF5C, but not both. Loss of gE/gI-US9p resulted in failure to recruit KIF1A and KF5C, but did not affect dynein binding. Unexpectedly, while KIF5C was expressed in undifferentiated and differentiated CAD neurons it was only found associated with PRV particles prepared from differentiated cells.


Assuntos
Herpesvirus Suídeo 1 , Peptídeos e Proteínas de Sinalização Intracelular , Cinesinas/metabolismo , Lipoproteínas , Microtúbulos/metabolismo , Pseudorraiva , Proteínas do Envelope Viral , Proteínas Virais , Liberação de Vírus , Animais , Transporte Biológico Ativo , Linhagem Celular , Deleção de Genes , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinesinas/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Microtúbulos/genética , Microtúbulos/virologia , Pseudorraiva/genética , Pseudorraiva/metabolismo , Pseudorraiva/patologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
Vet Microbiol ; 244: 108666, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32402347

RESUMO

This study describes an Aujeszky's disease case in an adult male red fox found in an urban area in Central Italy, that exhibited a fatal infection with neurological lesions, but neither itching nor skin lesions. Diagnostic examinations included histology, and parasitological, bacteriological and virological analyses. Detection of parasitic enteric pathogens, bacteria, E. coli, Leptospira spp., rabies, canine distemper virus, parvovirus, hepatitis E virus and pseudorabies virus (PrV) was performed. Results showed the presence of a gE-deleted PrVthat was closely related to the NIA-3 strain but differed from the PrV strains currently circulating in wild boars and domestic pigs in Italy. All the results led to the conclusion that the fox suffered from Aujeszky's disease caused by a gE-deleted PrV strain closely related to a vaccine strain. The epidemiological link between the PrV vaccine strain and fox infection remains unclear. It could involve vaccinated pigs as a primary source of infection by direct or indirect contact with the red fox or less likely it could be related to improper use of the vaccine in the fox.


Assuntos
Raposas/virologia , Deleção de Genes , Herpesvirus Suídeo 1/genética , Pseudorraiva/diagnóstico , Pseudorraiva/patologia , Proteínas do Envelope Viral/genética , Animais , Herpesvirus Suídeo 1/patogenicidade , Itália , Masculino , Vacinas contra Pseudorraiva , Vacinas Virais/imunologia
3.
Antiviral Res ; 173: 104652, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751590

RESUMO

Both classical swine fever (CSF) and pseudorabies are highly contagious, economically significant diseases of swine in China. Although vaccination with the C-strain against classical swine fever virus (CSFV) is widely carried out and severe outbreaks of CSF seldom occur in China, CSF is sporadic in many pig herds and novel sub-subgenotypes of CSFV endlessly emerge. Thus, new measures are needed to eradicate CSFV from Chinese farms. The emergence of a pseudorabies virus (PRV) variant also posed a new challenge for the control of swine pseudorabies. Here, the recombinant PRV strain JS-2012-ΔgE/gI-E2 expressing E2 protein of CSFV was developed by inserting the E2 expression cassette into the intergenic region between the gG and gD genes of the gE/gI-deletion PRV variant strain JS-2012-ΔgE/gI. The recombinant virus was stable when passaged in vitro. A single vaccination of JS-2012-ΔgE/gI-E2 via intramuscular injection fully protected against lethal challenges of PRV and CSFV. Vaccination of piglets with the recombinant JS-2012-ΔgE/gI-E2 in the presence of high levels of maternally derived antibodies (Abs) to PRV can provide partial protection against lethal challenge of CSFV. Vaccination of the recombinant PRV JS-2012-ΔgE/gI-E2 strain did not induce the production of Abs to the gE protein of PRV or to the CSFV proteins other than E2. Thus, JS-2012-ΔgE/gI-E2 appears to be a promising recombinant marker vaccine candidate against PRV and CSFV for the control and eradication of the PRV variant and CSFV.


Assuntos
Peste Suína Clássica/prevenção & controle , Expressão Gênica , Vetores Genéticos/genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/imunologia , Pseudorraiva/prevenção & controle , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/imunologia , Peste Suína Clássica/imunologia , Peste Suína Clássica/patologia , Vírus da Febre Suína Clássica/genética , Vírus da Febre Suína Clássica/imunologia , Ordem dos Genes , Herpesvirus Suídeo 1/patogenicidade , Pseudorraiva/imunologia , Pseudorraiva/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Suínos , Doenças dos Suínos/prevenção & controle , Vacinação , Vacinas Virais/genética , Vacinas Virais/imunologia
4.
Mol Neurodegener ; 14(1): 8, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30736827

RESUMO

BACKGROUND: Neurotropic virus-based tracers have been extensively applied in mapping and manipulation of neural circuits. However, their neurotropic and neurotoxic properties remain to be fully characterized. METHODS: Through neural circuit tracing, we systematically compared the neurotropism discrepancy among different multi-trans-synaptic and mono-synaptic retrograde viral tracers including pseudorabies virus (PRV), rabies virus (RV), and the newly engineered retro adeno-associated virus (rAAV2-retro) tracers. The (single-cell) RNA sequencing analysis was utilized for seeking possible attribution to neurotropism discrepancy and comparing cell toxicity caused by viral infection between glycoprotein-deleted RV (RV-∆G) and rAAV2-retro. Viral toxicity induced microglia activation and neuronal protein change were evaluated by immunohistochemistry. RESULTS: Multi-trans-synaptic retrograde viral tracers, PRV and RV, exhibit differential neurotropism when they were used for central neural circuit tracing from popliteal lymph nodes. Mono-synaptic retrograde tracers, including RV-∆G and rAAV2-retro, displayed discrepant neurotropic property, when they were applied to trace the inputs of lateral hypothalamic area and medial preoptic nucleus. rAAV2-retro demonstrated preference in cerebral cortex, whereas RV-∆G prefers to label basal ganglia and hypothalamus. Remarkably, we detected a distinct preference for specific cortical layer of rAAV2-retro in layer 5 and RV-∆G in layer 6 when they were injected into dorsal lateral geniculate nucleus to label corticothalamic neurons in primary visual cortex. Complementation of TVA receptor gene in RV-resistant neurons enabled EnvA-pseudotyped RV infection, supporting receptors attribution to viral neurotropism. Furthermore, both RV-∆G and rAAV2-retro exerted neurotoxic influence at the injection sites and retrogradely labeled sites, while the changes were more profound for RV-∆G infection. Finally, we demonstrated a proof-of-concept strategy for more comprehensive high-order circuit tracing of a specific target nucleus by combining rAAV2-retro, RV, and rAAV tracers. CONCLUSIONS: Different multi-trans-synaptic and mono-synaptic retrograde viral tracers exhibited discrepant neurotropism within certain brain regions, even cortical layer preference. More neurotoxicity was observed under RV-∆G infection as compared with rAAV2-retro. By combining rAAV2-retro, RV, and rAAV tracers, high-order circuit tracing can be achieved. Our findings provide important reference for appropriate application of viral tracers to delineate the landscape and dissect the function of neural network.


Assuntos
Encéfalo/virologia , Dependovirus , Corantes Fluorescentes , Herpesvirus Suídeo 1 , Vírus da Raiva , Animais , Proteínas Luminescentes , Camundongos , Infecções por Parvoviridae/patologia , Pseudorraiva/patologia , Raiva/patologia , Tropismo Viral
5.
Viruses ; 10(9)2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150559

RESUMO

Pseudorabies virus (PRV) is one of the most important pathogens of swine, resulting in devastating disease and economic losses worldwide. Nevertheless, there are currently no antiviral drugs available for PRV infection. Resveratrol (Res) was identified to exert its antiviral activity by inhibiting the PRV replication in preliminary investigations. In our previous study, we found that Res has anti-PRV activity in vitro. Here, we show that Res can effectively reduce the mortality and increase the growth performance of PRV-infected piglets. After Res treatment, the viral loads significantly (p < 0.001) decreased. Pathological symptoms, particularly inflammation in the brain caused by PRV infection, were significantly (p < 0.001) relieved by the effects of Res. In Res-treated groups, higher levels of cytokines in serum, including interferon gama, interleukin 12, tumor necrosis factor-alpha and interferon alpha were observed at 7 days post infection. These results indicated that Res possesses potent inhibitory activity against PRV-infection through inhibiting viral reproduction, alleviating PRV-induced inflammation and enhancing animal immunity, suggesting that Res is expected to be a new alternative control measure for PRV infection.


Assuntos
Antivirais/farmacologia , Herpesvirus Suídeo 1/efeitos dos fármacos , Pseudorraiva/terapia , Resveratrol/farmacologia , Doenças dos Suínos/terapia , Animais , Antivirais/uso terapêutico , Citocinas/sangue , Citocinas/imunologia , Herpesvirus Suídeo 1/imunologia , Herpesvirus Suídeo 1/patogenicidade , Humanos , Inflamação/imunologia , Inflamação/terapia , Pseudorraiva/sangue , Pseudorraiva/mortalidade , Pseudorraiva/patologia , Resveratrol/uso terapêutico , Suínos , Doenças dos Suínos/sangue , Doenças dos Suínos/mortalidade , Doenças dos Suínos/patologia , Carga Viral/efeitos dos fármacos , Virulência/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
6.
BMC Vet Res ; 12(1): 277, 2016 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-27923365

RESUMO

BACKGROUND: Since the end of 2011 an outbreak of pseudorabies affected Chinese pig herds that had been vaccinated with the commercial vaccine made of Bartha K61 strain. It is now clear that the outbreak was caused by an emergent PRV variant. Even though vaccines made of PRV Bartha K61 strain can confer certain cross protection against PRV variants based on experimental data, less than optimal clinical protection and virus shedding reduction were observed, making the control or eradication of this disease difficult. RESULTS: An infectious clone of PRV AH02LA strain was constructed to generate a gE deletion mutant PRV(LA-AB) strain. PRV(LA-AB) strain can reach a titer of 108.43 TCID50 /mL (50% tissue culture infectious dose) on BHK-21 cells. To evaluate the efficiency of the inactivated vaccine made of PRV(LA-AB) strain, thirty 3-week-old PRV-negative piglets were divided randomly into six groups for vaccination and challenge test. All five piglets in the challenge control showed typical clinical symptoms of pseudorabies post challenge. Sneezing and nasal discharge were observed in four and three piglets in groups C(vaccinated with inactivated PRV Bartha K61 strain vaccine) and D(vaccinated with live PRV Bartha K61 strain vaccine) respectively. In contrast, piglets in both groups A(vaccinated with inactivated PRV LA-AB strain vaccine) and B(vaccinated with inactivated PRV LA-AB strain vaccine with adjuvant) presented mild or no clinical symptoms. Moreover, viral titers detected via nasal swabs were approximately 100 times lower in group B than in the challenge control, and the duration of virus shedding (3-4 days) was shorter than in either the challenge control (5-10 days) or groups C and D (5-6 days). CONCLUSIONS: The infectious clone constructed in this study harbors the whole genome of the PRV variant AH02LA strain. The gE deletion mutant PRV(LA-AB)strain generated from PRV AH02LA strain can reach a high titer on BHK-21 cells. An inactivated vaccine of PRV LA-AB provides clinical protection and significantly reduces virus shedding post challenge, especially if accompanied by the adjuvant CVC1302. While Inactivated or live vaccines made of PRV Barth K61 strain can provide only partial protection in this test.


Assuntos
Vacinas contra Pseudorraiva/normas , Pseudorraiva/prevenção & controle , Doenças dos Suínos/prevenção & controle , Vacinas Virais/normas , Eliminação de Partículas Virais/imunologia , Animais , China , Deleção de Genes , Herpesviridae/genética , Herpesviridae/imunologia , Nariz/virologia , Pseudorraiva/patologia , Pseudorraiva/virologia , Vacinas contra Pseudorraiva/imunologia , Distribuição Aleatória , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/normas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia
7.
Arch Virol ; 161(2): 445-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26563317

RESUMO

Pseudorabies (PR, Aujeszky's disease) is an acute, highly contagious viral disease resulting in major economic losses to the swine industry. PR is endemic in wild and domestic animals, although its natural host is the pig. Here, we report an outbreak of PR in foxes on a fur-producing farm in Yuncheng county, Shandong, China, that were fed pig offal. The diagnosis of PR was based on nervous signs and standard PCR methods and by isolation of PRV from fox brain tissue in Vero cells. The diagnosis was confirmed by an indirect immunofluorescence assay and electron microscopy. Phylogenetic analysis of a partial (804 nt) viral glycoprotein gC gene sequence indicated that it was likely to be a field strain closely related to a cluster of PRV previously identified in China.


Assuntos
Surtos de Doenças , Raposas/virologia , Pseudorraiva/epidemiologia , Ração Animal , Animais , Composição de Bases , China/epidemiologia , Chlorocebus aethiops , Análise por Conglomerados , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/isolamento & purificação , Herpesvirus Suídeo 1/ultraestrutura , Microscopia Eletrônica de Transmissão , Filogenia , Reação em Cadeia da Polimerase , Pseudorraiva/patologia , Pseudorraiva/virologia , Análise de Sequência de DNA , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Cultura de Vírus
8.
J Feline Med Surg ; 15(7): 555-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23813813

RESUMO

OVERVIEW: Although pseudorabies in swine - Aujeszky's disease - has been eradicated from many pork-producing countries, the virus may still lurk in other vertebrate species and cause feline cases. Infection occurs through the ingestion of uncooked meat and organ material and presents as an acute encephalitis with a short incubation period and a rapidly fatal outcome. The ABCD considers this reason enough to include a review of this, now very rare, condition in this Special Issue.


Assuntos
Doenças do Gato/virologia , Pseudorraiva/prevenção & controle , Animais , Doenças do Gato/patologia , Gatos , Pseudorraiva/patologia
9.
Cell Host Microbe ; 13(2): 193-203, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23414759

RESUMO

Microtubule transport of herpesvirus capsids from the cell periphery to the nucleus is imperative for viral replication and, in the case of many alphaherpesviruses, transmission into the nervous system. Using the neuroinvasive herpesvirus, pseudorabies virus (PRV), we show that the viral protein 1/2 (VP1/2) tegument protein associates with the dynein/dynactin microtubule motor complex and promotes retrograde microtubule transport of PRV capsids. Functional activation of VP1/2 requires binding to the capsid protein pUL25 or removal of the capsid-binding domain. A proline-rich sequence within VP1/2 is required for the efficient interaction with the dynein/dynactin microtubule motor complex as well as for PRV virulence and retrograde axon transport in vivo. Additionally, in the absence of infection, functionally active VP1/2 is sufficient to move large surrogate cargoes via the dynein/dynactin microtubule motor complex. Thus, VP1/2 tethers PRV capsids to dynein/dynactin to enhance microtubule transport, neuroinvasion, and pathogenesis.


Assuntos
Dineínas/metabolismo , Herpesvirus Suídeo 1/patogenicidade , Células Receptoras Sensoriais/virologia , Proteínas Estruturais Virais/metabolismo , Animais , Axônios/metabolismo , Chlorocebus aethiops , Coinfecção/metabolismo , Coinfecção/virologia , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Herpesvirus Suídeo 1/metabolismo , Humanos , Imunoprecipitação , Masculino , Camundongos , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Membrana Nuclear/virologia , Prolina/metabolismo , Mapeamento de Interação de Proteínas , Transporte Proteico , Pseudorraiva/metabolismo , Pseudorraiva/patologia , Pseudorraiva/virologia , Ratos , Ratos Long-Evans , Células Receptoras Sensoriais/metabolismo , Células Vero , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética
10.
PLoS One ; 7(1): e30988, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22292087

RESUMO

Pseudorabies virus (PRV) belongs to Alphaherpesvirinae subfamily that causes huge economic loss in pig industry worldwide. It has been recently demonstrated that many herpesviruses encode microRNAs (miRNAs), which play crucial roles in viral life cycle. However, the knowledge about PRV-encoded miRNAs is still limited. Here, we report a comprehensive analysis of both viral and host miRNA expression profiles in PRV-infected porcine epithelial cell line (PK-15). Deep sequencing data showed that the ∼4.6 kb intron of the large latency transcript (LLT) functions as a primary microRNA precursor (pri-miRNA) that encodes a cluster of 11 distinct miRNAs in the PRV genome, and 209 known and 39 novel porcine miRNAs were detected. Viral miRNAs were further confirmed by stem-loop RT-PCR and northern blot analysis. Intriguingly, all of these viral miRNAs exhibited terminal heterogeneity both at the 5' and 3' ends. Seven miRNA genes produced mature miRNAs from both arms and two of the viral miRNA genes showed partially overlapped in their precursor regions. Unexpectedly, a terminal loop-derived small RNA with high abundance and one special miRNA offset RNA (moRNA) were processed from a same viral miRNA precursor. The polymorphisms of viral miRNAs shed light on the complexity of host miRNA-processing machinery and viral miRNA-regulatory mechanism. The swine genes and PRV genes were collected for target prediction of the viral miRNAs, revealing a complex network formed by both host and viral genes. GO enrichment analysis of host target genes suggests that PRV miRNAs are involved in complex cellular pathways including cell death, immune system process, metabolic pathway, indicating that these miRNAs play significant roles in virus-cells interaction of PRV and its hosts. Collectively, these data suggest that PRV infected epithelial cell line generates a diverse set of host miRNAs and a special cluster of viral miRNAs, which might facilitate PRV replication in cells.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/virologia , Herpesvirus Suídeo 1/fisiologia , MicroRNAs/genética , Pseudorraiva/genética , Animais , Sequência de Bases , Linhagem Celular , Perfilação da Expressão Gênica , Regulação Viral da Expressão Gênica , Herpesvirus Suídeo 1/genética , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/genética , MicroRNAs/metabolismo , Modelos Teóricos , Dados de Sequência Molecular , Pseudorraiva/patologia , RNA Viral/genética , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/patologia
11.
J Neurotrauma ; 27(8): 1369-78, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20528165

RESUMO

The retrograde transsynaptic tracer pseudorabies virus (PRV) has been widely used as a marker for synaptic connectivity in the spinal cord. Notably, the PRV-152 construct expresses enhanced green fluorescent protein (EGFP). We recently reported a significant attenuation of PRV-152 labeling of the intermediolateral cell column (IML) and celiac ganglia after complete T4 spinal cord transection versus sham injury in rats at 96 h after PRV-152 inoculation of the left kidney. Here we found a significant increase in noxious colorectal distention (CRD)-evoked c-Fos expression in spinal cords of injured versus sham rats without PRV infection. In order to assess whether enhancing neuronal activity in spinalized rats might increase PRV-152 labeling, we subjected awake spinalized rats to 1.5 h of intermittent noxious CRD either: (1) just prior to inoculation, or (2) 96 h after inoculation (n = 3/group). Equal numbers of spinalized rats in both groups received PRV-152 inoculations without CRD (non-stimulated; n = 3/group). At 96 h post-inoculation fixed spinal cords and left celiac ganglionic tissues were assessed for the distribution and quantification of EGFP-labeled cells. The injured cohort that received CRD just prior to PRV injection showed a significant reduction in EGFP-labeled cells in both the IML and left celiac ganglion compared to non-stimulated injured rats. In contrast, the injured cohort that received CRD 96 h after PRV-152 inoculation showed no differences in EGFP-labeled cell numbers in the IML or celiac ganglia versus non-stimulated injured rats. Interestingly, microglia near c-Fos-positive cells after acute CRD appeared more reactive compared to non-stimulated spinalized rats, and activated microglial cells markedly reduce viral transduction and progression following PRV inoculation of the CNS. Hence our results imply that increased CRD-induced c-Fos expression in the injured paradigm, prior to but not after PRV injection, further attenuates PRV-152 uptake, perhaps through changes in neuronal activity and/or innate neuro-immune responses.


Assuntos
Colo/fisiologia , Estado de Descerebração/fisiopatologia , Neurônios/patologia , Pseudorraiva/patologia , Reto/fisiologia , Sistema Nervoso Simpático/patologia , Animais , Fibras Autônomas Pré-Ganglionares/patologia , Contagem de Células , Feminino , Gânglios Simpáticos/patologia , Herpesvirus Suídeo 1/metabolismo , Imuno-Histoquímica , Microglia/patologia , Estimulação Física , Proteínas Proto-Oncogênicas c-fos/biossíntese , Ratos , Ratos Wistar , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia
12.
Vaccine ; 25(3): 547-60, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-16920232

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) infection still remains today as the most significant health threat to swine and poses a challenge to current vaccination strategies. To develop a new generation of vaccine against PRRSV, a live attenuated pseudorabies virus (PRV) was used as vaccine vector to express the two major membrane-associated proteins (GP5 or M) of PRRSV in various forms. Four PRV recombinants, rPRV-GP5 (expressing native GP5), rPRV-GP5m (expressing GP5m, a modified GP5), rPRV-GP5-M (co-expressing GP5 and M proteins), rPRV-GP5m-M (co-expressing GP5m and M proteins) were generated. Mouse immunized with all these recombinants developed comparable PRV-specific humoral immune responses and provided complete protection against a lethal PRV challenge. However, the highest level of PRRSV-specific neutralizing antibodies and lymphocyte proliferative responses was observed in mice immunized with rPRV-GP5m-M. The immunogenicity and protective efficiency of rPRV-GP5m-M were further evaluated in the piglets. Compared to commercial PRRSV killed vaccine, detectable PRRSV-specific neutralizing antibody and higher lymphocyte proliferative responses could be developed in piglets immunized with rPRV-GP5m-M before virus challenge. Furthermore, more efficient protection against a PRRSV challenge was obtained in piglets immunized with rPRV-GP5m-M, as showed by the balanced body-temperature fluctuation, shorter-term viremia, lower proportion of virus load in nasal and oropharyngeal scrapings and tissues, and milder lung lesions. These data indicate that the recombinant rPRV-GP5m-M is a promising candidate bivalent vaccine against both PRV and PRRSV infection.


Assuntos
Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/imunologia , Pseudorraiva/prevenção & controle , Animais , Temperatura Corporal/fisiologia , Proliferação de Células , Feminino , Linfócitos/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Síndrome Respiratória e Reprodutiva Suína/patologia , Pseudorraiva/patologia , RNA Viral/biossíntese , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Vacinas Atenuadas , Proteínas do Envelope Viral/imunologia , Viremia/virologia , Eliminação de Partículas Virais
13.
J Virol ; 80(13): 6387-98, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775327

RESUMO

The pseudorabies virus (PRV) Us3 gene is conserved among the alphaherpesviruses and encodes a serine/threonine protein kinase that is not required for growth in standard cell lines. In this report, we used a compartmented culture system to investigate the role of PRV Us3 in viral replication in neurons, in spread from neurons to PK15 cells, and in axon-mediated spread of infection. We also examined the role of Us3 in neuroinvasion and virulence in rodents. Us3 null mutants produce about 10-fold less infectious virus from neurons than wild-type virus and have no discernible phenotypes for axonal targeting of viral components in cultured peripheral nervous system neurons. After eye infection in rodents, Us3 null mutants were slightly attenuated for virulence, with a delayed onset of symptoms compared to the wild type or a Us3 null revertant. While initially delayed, the symptoms increased in severity until they approximated those of the wild-type virus. Us3 null mutants were neuroinvasive, spreading in both efferent and afferent circuits innervating eye tissues.


Assuntos
Axônios/metabolismo , Infecções Oculares Virais/metabolismo , Herpesvirus Suídeo 1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Pseudorraiva/metabolismo , Replicação Viral , Animais , Axônios/patologia , Axônios/virologia , Técnicas de Cocultura , Olho/inervação , Olho/metabolismo , Olho/patologia , Olho/virologia , Infecções Oculares Virais/genética , Infecções Oculares Virais/patologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Mutação , Células PC12 , Proteínas Serina-Treonina Quinases/deficiência , Pseudorraiva/genética , Pseudorraiva/patologia , Ratos , Replicação Viral/genética
14.
J Virol ; 79(6): 3831-40, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731276

RESUMO

Homologs of the UL51 protein of herpes simplex virus have been identified in all herpesvirus subfamilies, but until now, no function has been assigned to any of them. To investigate function of the UL51 gene product of the alphaherpesvirus pseudorabies virus (PrV), we isolated and analyzed a mutant lacking the major part of the open reading frame, PrV-DeltaUL51F, and a rescuant. One-step growth analysis of PrV-DeltaUL51F revealed only slightly reduced titers, but plaque size was notably diminished and reached only approximately 30% the plaque size of wild-type PrV. Ultrastructurally, intracytoplasmic capsids were found in large numbers either without envelope or in different stages of envelopment, indicating that secondary envelopment in the cytoplasm was less efficient. However, neuroinvasion in the mouse trigeminal pathway after intranasal infection was only slightly delayed. A PrV UL11 mutant also showed a defect in secondary envelopment (M. Kopp, H. Granzow, W. Fuchs, B. G. Klupp, E. Mundt, A. Karger, and T. C. Mettenleiter, J. Virol. 77:5339-5351, 2003). Since both proteins are part of the viral tegument and are predicted to be membrane associated, they may serve similar, possibly redundant functions during viral morphogenesis. Therefore, we also isolated a mutant simultaneously lacking UL51 and UL11. This mutant exhibited further reduced plaque size compared to the single-deletion mutants, but viral titers were comparable to those for the UL11 mutant. In electron microscopic analyses, the observed defect in secondary envelopment was similar to that found in the UL11 single-deletion mutant. In conclusion, both conserved tegument proteins, either singly or in combination, are involved in virion morphogenesis in the cytoplasm but are not essential for viral replication in vitro and in vivo.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Animais , Capsídeo/ultraestrutura , Citoplasma/virologia , Modelos Animais de Doenças , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Camundongos , Microscopia Eletrônica , Morfogênese , Pseudorraiva/patologia , Pseudorraiva/virologia , Coelhos , Deleção de Sequência , Ensaio de Placa Viral , Proteínas Virais , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/fisiologia , Vírion/ultraestrutura
15.
Vet Microbiol ; 86(1-2): 5-16, 2002 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-11888685

RESUMO

Pseudorabies virus (PRV), an alpha-herpesvirus, is capable of spreading between synaptically connected neurons in diverse hosts. In this report, two lines of experimentation are summarized that provide insight into the mechanism of virus spread in neurons. First, techniques were developed to measure the transport dynamics of capsids in infected neurons. Individual viral capsids labeled with green fluorescent protein (GFP) were visualized and tracked as they moved in axons away from infected neuronal cell bodies in culture during egress. Second, the effects of three viral membrane proteins (gE, gI and Us9) on the localization of envelope, tegument, and capsid proteins in infected, cultured sympathetic neurons were determined. These three proteins are necessary for spread of infection from pre-synaptic neurons to post-synaptic neurons in vivo (anterograde spread). Us9 mutants apparently are defective in anterograde spread in neural circuits because essential viral membrane proteins such as gB are not transported to axon terminals to facilitate spread to the connected neuron. By contrast, gE and gI mutants manifest their phenotype because these proteins most likely function at the axon terminal of the infected neuron to promote spread. These two sets of experiments are consistent with a model for herpesvirus spread in neurons first suggested by Cunningham and colleagues where capsids and envelope proteins, but not whole virions, are transported separately into the axon.


Assuntos
Herpesvirus Suídeo 1/crescimento & desenvolvimento , Doenças do Sistema Nervoso Periférico/virologia , Pseudorraiva/virologia , Proteínas Virais , Animais , Axônios/virologia , Embrião de Galinha , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas/análise , Lipoproteínas/fisiologia , Microscopia Confocal , Microscopia de Fluorescência , Neurônios Aferentes/patologia , Neurônios Aferentes/virologia , Doenças do Sistema Nervoso Periférico/patologia , Fosfoproteínas/análise , Fosfoproteínas/fisiologia , Terminações Pré-Sinápticas/virologia , Pseudorraiva/patologia , Ratos , Gravação de Videoteipe , Proteínas do Envelope Viral/análise , Proteínas do Envelope Viral/fisiologia
16.
Vet Microbiol ; 86(1-2): 89-94, 2002 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-11888692

RESUMO

Rounding and loosening of cells is a consequence of infection with pseudorabies virus (PrV), both in vitro and in vivo. These changes in the normal structure of the cell may be the result of cytoskeletal changes. Immunofluorescence staining of actin filaments and microtubule bundles was performed to examine whether PrV induces a reorganization of these cytoskeletal components in infected swine kidney (SK) cells. Every 2h until 12h post-inoculation (p.i.), cells were washed in cytoskeleton stabilizing buffer (CSB), fixed with paraformaldehyde and washed again with CSB. Cells were permeabilized with a 1/1000 dilution of Triton X-100 and actin filaments were stained by incubating cells with phalloidin-Texas Red. Staining of microtubules was done by incubating the cells subsequently with mouse monoclonal anti-alpha-tubulin and goat anti-mouse IgG-FITC. During the course of infection, actin fibers of SK cells were rearranged in the following sequence: (1) disappearance of thick actin stress fibers between 4 and 6h p.i., (2) complete loss of stress fibers between 6 and 8h p.i., and (3) reappearance of thin stress fibers starting from 10h p.i. In contrast to herpes simplex virus 1 (HSV1) or equine herpesvirus 1 (EHV1), PrV infection did not induce changes in the cellular microtubule network. PrV infection induces a temporary disassembly of actin stress fibers.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Citoesqueleto de Actina/virologia , Herpesvirus Suídeo 1/ultraestrutura , Rim/ultraestrutura , Rim/virologia , Pseudorraiva/patologia , Doenças dos Suínos/virologia , Citoesqueleto de Actina/fisiologia , Animais , Células Cultivadas , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Microscopia Confocal , Microtúbulos/fisiologia , Microtúbulos/ultraestrutura , Pseudorraiva/virologia , Suínos , Doenças dos Suínos/patologia
17.
J Neurosci ; 22(7): 2701-10, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11923435

RESUMO

Intravitreal injection of the attenuated strain of pseudorabies virus (PRV Bartha) results in transneuronal spread of virus to a restricted set of central nuclei in the rat and mouse. We examined the pattern of central infection in the golden hamster after intravitreal inoculation with a recombinant strain of PRV Bartha constructed to express enhanced green fluorescent protein (PRV 152). Neurons in a subset of retinorecipient nuclei [i.e., suprachiasmatic nucleus (SCN), intergeniculate leaflet, olivary pretectal nucleus (OPN), and lateral terminal nucleus] and autonomic nuclei [i.e., paraventricular hypothalamic nucleus and Edinger-Westphal nucleus (EW)] are labeled by late stages of infection. Infection of the EW precedes infection in retinorecipient structures, raising the possibility that the SCN becomes infected by retrograde transsynaptic infection via autonomic (i.e., EW) circuits. We tested this hypothesis in two ways: (1) by removing the infected eye 24 hr after PRV 152 inoculation, well before viral infection first appears in the SCN; and (2) by examining central infection after intravitreal PRV 152 injection in animals with ablation of the EW. The pattern and time course of central infection were unchanged after enucleation, whereas EW ablation before intravitreal inoculation eliminated viral infection in the SCN. The results of EW lesions along with known connections between EW, OPN, and SCN indicate that intravitreal injection of PRV Bartha produces a retrograde infection of the autonomic innervation of the eye, which subsequently labels a restricted set of retinorecipient nuclei via retrograde trans-synaptic infection. These results, taken together with other genetic data, indicate that the mutations in PRV Bartha render the virus incapable of anterograde transport. PRV Bartha is thus a retrograde transsynaptic marker in the CNS.


Assuntos
Sistema Nervoso Autônomo/virologia , Transporte Axonal , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Pseudorraiva/virologia , Núcleo Supraquiasmático/virologia , Corpo Vítreo/virologia , Animais , Sistema Nervoso Autônomo/patologia , Transporte Axonal/fisiologia , Transporte Biológico , Cricetinae , Progressão da Doença , Enucleação Ocular , Genes Reporter , Herpesvirus Suídeo 1/genética , Proteínas Luminescentes/genética , Mesocricetus , Neurônios/patologia , Neurônios/virologia , Pseudorraiva/patologia , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/virologia , Núcleo Supraquiasmático/patologia , Sinapses/patologia , Sinapses/virologia , Vias Visuais/patologia , Vias Visuais/virologia
18.
Physiol Behav ; 77(4-5): 557-63, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12526999

RESUMO

In addition to its role as a neurotransmitter, studies have postulated both neuroprotective and neurotoxic roles for nitric oxide (NO) generated in response to infections with neurotropic viruses. This study examined the expression of neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) isoforms of NOS induced by neuronal infection with virulent and attenuated strains of pseudorabies virus (PRV). Caudal brainstem neurons infected by peripheral inoculation of the viscera served as the model system. Neuronal infection induced the expression of nNOS and iNOS, but the timing and the apparent magnitude of NOS expression varied according to the virulence of the infecting strain of virus. Expression of nNOS was observed in infected neurons that did not express this enzyme in control animals, and the onset of expression was earlier in animals infected with virulent PRV. Expression of iNOS was largely restricted to monocytes and macrophages that invaded the brain in response to PRV infection. These iNOS-expressing cells were observed earlier in animals infected with the virulent virus, and were differentially concentrated in areas exhibiting virus-induced neuropathology. Collectively, these data suggest functionally diverse roles for NO in the brain response to PRV neuronal infection.


Assuntos
Vetores Genéticos/genética , Herpesvirus Suídeo 1/genética , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/genética , Animais , Isoenzimas/biossíntese , Isoenzimas/genética , Macrófagos/enzimologia , Masculino , Monócitos/enzimologia , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Pseudorraiva/patologia , Ratos , Ratos Sprague-Dawley , Replicação Viral/fisiologia
19.
Vaccine ; 19(27): 3752-8, 2001 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-11395210

RESUMO

Five week old, commercially available large white pigs were vaccinated with either a single dose or two doses of a recombinant porcine adenovirus expressing the glycoprotein D gene from pseudorabies virus (PRV). Pigs were monitored for the development of serum neutralizing antibodies to PRV and challenged 3 weeks after final vaccination. Prior to challenge, pigs given 2 doses of the vaccine demonstrated boosted levels of antibody compared with those given a single dose, and all surviving pigs had increased neutralization titres over pre-challenge levels. Following challenge, pigs were monitored for clinical signs of disease, with blood and nasal swabs collected for virus isolation. All control animals became sick with elevated temperatures for 6 days post challenge, whereas; vaccinated animals displayed an increase in body temperature for only 2-3 days. Control pigs and those given a single dose all lost condition, but the group given 2 doses remained healthy. At postmortem, gross lesions of pneumonia only occurred in control animals and those given a single dose of vaccine. Histology carried out on the brains of all animals demonstrated a difference in severity of infection and frequency of immunohistochemical antigen detection between test animals, with control and single dose groups being most severely affected and pigs given 2 doses the least. Virus isolation studies demonstrated that no viraemia could be detected, but virus was found in nasal swabs from some animals in both groups of vaccinates following challenge.


Assuntos
Adenoviridae/genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/uso terapêutico , Pseudorraiva/prevenção & controle , Vacinas de DNA/uso terapêutico , Proteínas do Envelope Viral/genética , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Herpesvirus Suídeo 1/isolamento & purificação , Esquemas de Imunização , Testes de Neutralização , Pseudorraiva/mortalidade , Pseudorraiva/patologia , Vacinas contra Pseudorraiva/genética , Suínos , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/imunologia
20.
J Comp Pathol ; 123(2-3): 152-63, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11032669

RESUMO

In the course of two experiments, an examination was made of the virulence and neuroinvasiveness for pigs of two pseudorabies virus mutants (strain 6C2TK(-), with a defect in thymidine kinase (TK) function; and strain 6C2TK(-), gI(-)/gE(-), with defects in TK and glycoproteins I and E) and of the wild-type parent strain (86/27V). At various times after intranasal inoculation, pigs were killed and samples of tonsil, lung and different levels of the trigeminal and olfactory nervous pathways were examined by methods that included viral isolation, polymerase chain reaction assay and immunohistochemistry. Both mutant viruses were of reduced virulence, as indicated by no more than moderate clinical signs and lesions, and only sporadic isolation of virus; moreover, unlike the wild-type parent strain, the mutant viruses were not reactivated from the latent state by corticosteroid treatment. In addition, migration of the mutant strains to the central nervous system (olfactory and trigeminal nervous pathways) was reduced as compared with that of the wild-type strain. Thus, mutations in the genes encoding the TK enzyme and the gI/gE complex were associated with reduced virulence, reduced replication in peripheral target tissues, and reduced migration to the olfactory and trigeminal pathways.


Assuntos
Herpesvirus Suídeo 1/patogenicidade , Pseudorraiva/patologia , Timidina Quinase/genética , Proteínas do Envelope Viral/genética , Ativação Viral , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/efeitos dos fármacos , Antígenos Virais/análise , Linhagem Celular , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , DNA Viral/genética , Dexametasona/uso terapêutico , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/imunologia , Imuno-Histoquímica , Mutação , Bulbo Olfatório/patologia , Bulbo Olfatório/virologia , Reação em Cadeia da Polimerase , Pseudorraiva/prevenção & controle , Pseudorraiva/virologia , Suínos , Fatores de Tempo , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia , Virulência/genética , Ativação Viral/genética , Latência Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA