Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 96(13): e0071422, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35730976

RESUMEN

Pseudorabies virus (PRV) is a porcine alphaherpesvirus that belongs to the Herpesviridae family. We showed earlier that infection of porcine epithelial cells with PRV triggers activation of the nuclear factor κB (NF-κB) pathway, a pivotal signaling axis in the early immune response. However, PRV-induced NF-κB activation does not lead to NF-κB-dependent gene expression. Here, using electrophoretic mobility shift assays (EMSAs), we show that PRV does not disrupt the ability of NF-κB to interact with its κB target sites. Assessing basal cellular transcriptional activity in PRV-infected cells by quantitation of prespliced transcripts of constitutively expressed genes uncovered a broad suppression of cellular transcription by PRV, which also affects the inducible expression of NF-κB target genes. Host cell transcription inhibition was rescued when viral genome replication was blocked using phosphonoacetic acid (PAA). Remarkably, we found that host gene expression shutoff in PRV-infected cells correlated with a substantial retention of the NF-κB subunit p65, the TATA box binding protein, and RNA polymerase II-essential factors required for (NF-κB-dependent) gene transcription-in expanding PRV replication centers in the nucleus and thereby away from the host chromatin. This study reveals a potent mechanism used by the alphaherpesvirus PRV to steer the protein production capacity of infected cells to viral proteins by preventing expression of host genes, including inducible genes involved in mounting antiviral responses. IMPORTANCE Herpesviruses are highly successful pathogens that cause lifelong persistent infections of their host. Modulation of the intracellular environment of infected cells is imperative for the success of virus infections. We reported earlier that a DNA damage response in epithelial cells infected with the alphaherpesvirus pseudorabies virus (PRV) results in activation of the hallmark proinflammatory NF-κB signaling axis but, remarkably, that this activation does not lead to NF-κB-induced (proinflammatory) gene expression. Here, we report that PRV-mediated inhibition of host gene expression stretches beyond NF-κB-dependent gene expression and in fact reflects a broad inhibition of host gene transcription, which correlates with a substantial recruitment of essential host transcription factors in viral replication compartments in the nucleus, away from the host chromatin. These data uncover a potent alphaherpesvirus mechanism to interfere with production of host proteins, including proteins involved in antiviral responses.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Transcripción Genética , Animales , Herpesvirus Suido 1/fisiología , Interacciones Microbiota-Huesped , FN-kappa B/genética , FN-kappa B/metabolismo , Seudorrabia/inmunología , Seudorrabia/fisiopatología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/fisiopatología
2.
Proc Natl Acad Sci U S A ; 115(40): E9419-E9428, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30154170

RESUMEN

Previous studies have shown changes in membrane properties of neurons in rat deep cerebellar nuclei (DCN) as a function of development, but due to technical difficulties in obtaining viable DCN slices from adult animals, it remains unclear whether there are learning-related alterations in the membrane properties of DCN neurons in adult rats. This study was designed to record from identified DCN cells in cerebellar slices from postnatal day 25-26 (P25-26) rats that had a relatively mature sensory nervous system and were able to acquire learning as a result of tone-shock eyeblink conditioning (EBC) and to document resulting changes in electrophysiological properties. After electromyographic electrode implantation at P21 and inoculation with a fluorescent pseudorabies virus (PRV-152) at P22-23, rats received either four sessions of paired delay EBC or unpaired stimulus presentations with a tone conditioned stimulus and a shock unconditioned stimulus or sat in the training chamber without stimulus presentations. Compared with rats given unpaired stimuli or no stimulus presentations, rats given paired EBC showed an increase in conditioned responses across sessions. Whole-cell recordings of both fluorescent and nonfluorescent DCN projection neurons showed that delay EBC induced significant changes in membrane properties of evoked DCN action potentials including a reduced after-hyperpolarization amplitude and shortened latency. Similar findings were obtained in hyperpolarization-induced rebound spikes of DCN neurons. In sum, delay EBC produced significant changes in the membrane properties of juvenile rat DCN projection neurons. These learning-specific changes in DCN excitability have not previously been reported in any species or task.


Asunto(s)
Parpadeo , Membrana Celular , Núcleos Cerebelosos/fisiopatología , Neuronas , Animales , Estimulación Eléctrica , Herpesvirus Suido 1 , Seudorrabia/fisiopatología , Ratas , Ratas Long-Evans
4.
Vet Res ; 50(1): 9, 2019 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-30717799

RESUMEN

As a key cellular transcription factor that plays a central role in cellular responses to a broad range of stress factors, p53 has generally been considered as a host cell restriction factor for various viral infections. However, the defined roles of p53 in pseudorabies virus (PRV) replication, pathogenesis, and host responses remain unclear. In the present study, we initially constructed a p53 overexpressing a porcine kidney epithelial cell line (PK-15) to detect the effect of p53 on PRV replication in vitro. The results show that viral glycoprotein B (gB) gene copies and the titers of virus were significantly higher in p53 overexpressing PK-15 cells than in PK-15 and p53 inhibitor treated p53 overexpressing PK-15 cells. A similar result was also found in the p53 inhibitor PFT-α-treated PK-15 cells. We then examined the effects of p53 on PRV infection in vivo by using p53-knockout (p53-/-) mice. The results show that p53 knockout not only led to significantly reduced rates of mortality but also to reduced viral replication and development of viral encephalitis in the brains of mice following intracranial inoculation. Furthermore, we examined the effect of p53 knockout on the expression of the reported host cell regulators of PRV replication in the brains of mice by using RNA sequencing. The results show that p53 knockout downregulated the interferon (IFN) regulator genes, chemokine genes, and antiviral genes after PRV infection. This finding suggests that p53 positively regulates viral replication and pathogenesis both in vitro and in vivo. These findings offer novel targets of intrinsic host cell immunity for PRV infection.


Asunto(s)
Herpesvirus Suido 1/fisiología , Herpesvirus Suido 1/patogenicidad , Inmunidad Innata , Seudorrabia/inmunología , Enfermedades de los Porcinos/inmunología , Proteína p53 Supresora de Tumor/genética , Replicación Viral , Animales , Línea Celular , Interacciones Huésped-Patógeno , Seudorrabia/fisiopatología , Seudorrabia/virología , Porcinos , Enfermedades de los Porcinos/fisiopatología , Enfermedades de los Porcinos/virología , Proteína p53 Supresora de Tumor/metabolismo , Virulencia
5.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R353-R365, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29118019

RESUMEN

Chronic pelvic pain causes significant patient morbidity and is a challenge to clinicians. Using a murine neurogenic cystitis model that recapitulates key aspects of interstitial cystitis/bladder pain syndrome (IC), we recently showed that pseudorabies virus (PRV) induces severe pelvic allodynia in BALB/c mice relative to C57BL/6 mice. Here, we report that a quantitative trait locus (QTL) analysis of PRV-induced allodynia in F2CxB progeny identified a polymorphism on chromosome 13, rs6314295 , significantly associated with allodynia (logarithm of odds = 3.11). The nearby gene encoding acyloxyacyl hydrolase ( Aoah) was induced in the sacral spinal cord of PRV-infected mice. AOAH-deficient mice exhibited increased vesicomotor reflex in response to bladder distension, consistent with spontaneous bladder hypersensitivity, and increased pelvic allodynia in neurogenic cystitis and postbacterial chronic pain models. AOAH deficiency resulted in greater bladder pathology and tumor necrosis factor production in PRV neurogenic cystitis, markers of increased bladder mast cell activation. AOAH immunoreactivity was detectable along the bladder-brain axis, including in brain sites previously correlated with human chronic pelvic pain. Finally, AOAH-deficient mice had significantly higher levels of bladder vascular endothelial growth factor, an emerging marker of chronic pelvic pain in humans. These findings indicate that AOAH modulates pelvic pain severity, suggesting that allelic variation in Aoah influences pelvic pain in IC.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Cistitis Intersticial/enzimología , Infecciones por Escherichia coli/enzimología , Hiperalgesia/enzimología , Dolor Pélvico/enzimología , Seudorrabia/enzimología , Vejiga Urinaria/inervación , Infecciones Urinarias/enzimología , Animales , Conducta Animal , Hidrolasas de Éster Carboxílico/deficiencia , Hidrolasas de Éster Carboxílico/genética , Cistitis Intersticial/genética , Cistitis Intersticial/fisiopatología , Cistitis Intersticial/psicología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/fisiopatología , Infecciones por Escherichia coli/psicología , Femenino , Predisposición Genética a la Enfermedad , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Percepción del Dolor , Umbral del Dolor , Dolor Pélvico/genética , Dolor Pélvico/fisiopatología , Fenotipo , Seudorrabia/genética , Seudorrabia/fisiopatología , Seudorrabia/psicología , Sitios de Carácter Cuantitativo , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/metabolismo , Vejiga Urinaria/metabolismo , Infecciones Urinarias/genética , Infecciones Urinarias/fisiopatología , Infecciones Urinarias/psicología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Virus Genes ; 53(4): 565-572, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28417300

RESUMEN

Emerging pseudorabies virus (PRV) variant has led to frequent outbreaks of PRV infection among Bartha-K61-vaccinated swine population in Chinese swine farms and caused high mortality in pigs of all age since late 2011. Here, we generated a gE/gI-deleted PRV (rPRVXJ-delgI/gE-EGFP) based on PRV variant strain (PRV-XJ) through homologous DNA recombination. Compared to parental strain, rPRVXJ-delgI/gE-EGFP showed similar growth kinetics in vitro. Its safety and immunogenicity were evaluated in weaned piglets. Our results showed that piglets immunized with rPRVXJ-delgI/gE-EGFP did not exhibit any clinical symptoms, and a high level of gB-specific antibody was detected. After lethal challenge with variant PRV (PRV-FJ strain), all vaccinated piglets survived without showing any clinical symptoms except slight fever within 7 days post-challenge. In unvaccinated piglets, typical clinical symptoms of pseudorabies were observed, and the piglets were all died at 5 days post-challenge. These results indicated that a live rPRVXJ-delgI/gE-EGFP vaccine could be a maker vaccine candidate to control the currently epidemic pseudorabies in China.


Asunto(s)
Eliminación de Gen , Herpesvirus Suido 1/genética , Seudorrabia/virología , Enfermedades de los Porcinos/virología , Proteínas del Envoltorio Viral/genética , Animales , Anticuerpos Antivirales/inmunología , Herpesvirus Suido 1/inmunología , Herpesvirus Suido 1/metabolismo , Seudorrabia/inmunología , Seudorrabia/fisiopatología , Seudorrabia/prevención & control , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/fisiopatología , Enfermedades de los Porcinos/prevención & control , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/inmunología , Destete
7.
J Neurosci ; 32(36): 12472-87, 2012 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-22956838

RESUMEN

Rabbit eyeblink conditioning is a well characterized model of associative learning. To identify specific neurons that are part of the eyeblink premotor pathway, a retrograde transsynaptic tracer (pseudorabies virus) was injected into the orbicularis oculi muscle. Four time points (3, 4, 4.5, and 5 d) were selected to identify sequential segments of the pathway and a map of labeled structures was generated. At 3 d, labeled first-order motor neurons were found in dorsolateral facial nucleus ipsilaterally. At 4 d, second-order premotor neurons were found in reticular nuclei, and sensory trigeminal, auditory, vestibular, and motor structures, including contralateral red nucleus. At 4.5 d, labeled third-order premotor neurons were found in the pons, midbrain, and cerebellum, including dorsolateral anterior interpositus nucleus and rostral fastigial nucleus. At 5 d, labeling revealed higher-order premotor structures. Labeled fourth-order Purkinje cells were found in ipsilateral cerebellar cortex in cerebellar lobule HVI and in lobule I. The former has been implicated in eyeblink conditioning and the latter in vestibular control. Labeled neurons in anterior interpositus were studied, using neurotransmitter immunoreactivity to classify individual cell types and delineate their interconnectivity. Labeled third-order premotor neurons were immunoreactive for glutamate and corresponded to large excitatory projection neurons. Labeled fourth-order premotor interneurons were immunoreactive for GABA (30%), glycine (18%), or both GABA and glycine (52%) and form a functional network within anterior interpositus involved in modulation of motor commands. These results identify a complete eyeblink premotor pathway, deep cerebellar interconnectivity, and specific neurons responsible for the generation of eyeblink responses.


Asunto(s)
Parpadeo , Cerebelo/anatomía & histología , Neuronas Motoras/citología , Red Nerviosa/anatomía & histología , Músculos Oculomotores/anatomía & histología , Seudorrabia , Animales , Parpadeo/fisiología , Corteza Cerebelosa/anatomía & histología , Corteza Cerebelosa/fisiología , Cerebelo/fisiología , Neuronas Motoras/fisiología , Red Nerviosa/fisiología , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Músculos Oculomotores/química , Músculos Oculomotores/fisiología , Seudorrabia/patología , Seudorrabia/fisiopatología , Conejos
8.
PLoS Pathog ; 5(10): e1000640, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19876391

RESUMEN

Alpha-herpesviruses, including human herpes simplex virus 1 & 2, varicella zoster virus and the swine pseudorabies virus (PRV), infect the peripheral nervous system of their hosts. Symptoms of infection often include itching, numbness, or pain indicative of altered neurological function. To determine if there is an in vitro electrophysiological correlate to these characteristic in vivo symptoms, we infected cultured rat sympathetic neurons with well-characterized strains of PRV known to produce virulent or attenuated symptoms in animals. Whole-cell patch clamp recordings were made at various times after infection. By 8 hours of infection with virulent PRV, action potential (AP) firing rates increased substantially and were accompanied by hyperpolarized resting membrane potentials and spikelet-like events. Coincident with the increase in AP firing rate, adjacent neurons exhibited coupled firing events, first with AP-spikelets and later with near identical resting membrane potentials and AP firing. Small fusion pores between adjacent cell bodies formed early after infection as demonstrated by transfer of the low molecular weight dye, Lucifer Yellow. Later, larger pores formed as demonstrated by transfer of high molecular weight Texas red-dextran conjugates between infected cells. Further evidence for viral-induced fusion pores was obtained by infecting neurons with a viral mutant defective for glycoprotein B, a component of the viral membrane fusion complex. These infected neurons were essentially identical to mock infected neurons: no increased AP firing, no spikelet-like events, and no electrical or dye transfer. Infection with PRV Bartha, an attenuated circuit-tracing strain delayed, but did not eliminate the increased neuronal activity and coupling events. We suggest that formation of fusion pores between infected neurons results in electrical coupling and elevated firing rates, and that these processes may contribute to the altered neural function seen in PRV-infected animals.


Asunto(s)
Potenciales de Acción/fisiología , Herpesvirus Suido 1/fisiología , Neuronas/fisiología , Seudorrabia/fisiopatología , Animales , Células Cultivadas , Electrofisiología , Colorantes Fluorescentes/metabolismo , Células Gigantes/virología , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Seudorrabia/virología , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/virología , Porcinos , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Replicación Viral
9.
PLoS Pathog ; 5(4): e1000387, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19381253

RESUMEN

The neuroinvasive property of several alpha-herpesviruses underlies an uncommon infectious process that includes the establishment of life-long latent infections in sensory neurons of the peripheral nervous system. Several herpesvirus proteins are required for replication and dissemination within the nervous system, indicating that exploiting the nervous system as a niche for productive infection requires a specialized set of functions encoded by the virus. Whether initial entry into the nervous system from peripheral tissues also requires specialized viral functions is not known. Here we show that a conserved deubiquitinase domain embedded within a pseudorabies virus structural protein, pUL36, is essential for initial neural invasion, but is subsequently dispensable for transmission within and between neurons of the mammalian nervous system. These findings indicate that the deubiquitinase contributes to neurovirulence by participating in a previously unrecognized initial step in neuroinvasion.


Asunto(s)
Endopeptidasas/fisiología , Herpesvirus Suido 1/enzimología , Seudorrabia/virología , Células Receptoras Sensoriales/virología , Ubiquitina/metabolismo , Proteínas Estructurales Virales/fisiología , Animales , Cámara Anterior/virología , Transporte Axonal/fisiología , Chlorocebus aethiops , Endopeptidasas/genética , Infecciones Virales del Ojo/virología , Herpesvirus Suido 1/genética , Masculino , Seudorrabia/fisiopatología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Proteasas Ubiquitina-Específicas , Células Vero , Proteínas Estructurales Virales/genética
10.
J Vet Sci ; 22(2): e20, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33774936

RESUMEN

BACKGROUND: Pseudorabies virus (PRV) infection leads to high mortality in swine. Despite extensive efforts, effective treatments against PRV infection are limited. Furthermore, the inflammatory response induced by PRV strain GXLB-2013 is unclear. OBJECTIVES: Our study aimed to investigate the inflammatory response induced by PRV strain GXLB-2013, establish an inflammation model to elucidate the pathogenesis of PRV infection further, and develop effective drugs against PRV infection. METHODS: Kunming mice were infected intramuscularly with medium, LPS, and different doses of PRV-GXLB-2013. Viral spread and histopathological damage to brain, spleen, and lung were determined at 7 days post-infection (dpi). Immune organ indices, levels of reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines, as well as levels of activity of COX-2 and iNOS were determined at 4, 7, and 14 dpi. RESULTS: At 105-106 TCID50 PRV produced obviously neurological symptoms and 100% mortality in mice. Viral antigens were detectable in kidney, heart, lung, liver, spleen, and brain. In addition, inflammatory injuries were apparent in brain, spleen, and lung of PRV-infected mice. Moreover, PRV induced increases in immune organ indices, ROS and NO levels, activity of COX-2 and iNOS, and the content of key pro-inflammatory cytokines, including interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, interferon-γ and MCP-1. Among the tested doses, 10² TCID50 of PRV produced a significant inflammatory mediator increase. CONCLUSIONS: An inflammatory model induced by PRV infection was established in mice, and 10² TCID50 PRV was considered as the best concentration for the establishment of the model.


Asunto(s)
Herpesvirus Suido 1/fisiología , Inflamación/veterinaria , Seudorrabia/inmunología , Animales , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/fisiopatología , Inflamación/virología , Seudorrabia/fisiopatología , Seudorrabia/virología , Sus scrofa , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/fisiopatología , Enfermedades de los Porcinos/virología
11.
BMC Microbiol ; 9: 246, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19948073

RESUMEN

BACKGROUND: Pseudorabies virus (PRV) is an alphaherpesviruses whose native host is pig. PRV infection mainly causes signs of central nervous system disorder in young pigs, and respiratory system diseases in the adult. RESULTS: In this report, we have analyzed native host (piglets) gene expression changes in response to acute pseudorabies virus infection of the brain and lung using a printed human oligonucleotide gene set from Illumina. A total of 210 and 1130 out of 23,000 transcript probes displayed differential expression respectively in the brain and lung in piglets after PRV infection (p-value < 0.01), with most genes displaying up-regulation. Biological process and pathways analysis showed that most of the up-regulated genes are involved in cell differentiation, neurodegenerative disorders, the nervous system and immune responses in the infected brain whereas apoptosis, cell cycle control, and the mTOR signaling pathway genes were prevalent in the infected lung. Additionally, a number of differentially expressed genes were found to map in or close to quantitative trait loci for resistance/susceptibility to pseudorabies virus in piglets. CONCLUSION: This is the first comprehensive analysis of the global transcriptional response of the native host to acute alphaherpesvirus infection. The differentially regulated genes reported here are likely to be of interest for the further study and understanding of host viral gene interactions.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Herpesvirus Suido 1/fisiología , Pulmón/metabolismo , Seudorrabia/metabolismo , Seudorrabia/fisiopatología , Animales , Biología Computacional/métodos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/fisiopatología
12.
Vet Microbiol ; 239: 108485, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31767094

RESUMEN

Pseudorabies virus (PRV) infection causes great economic losses in the pig industry. By disrupting the homeostasis of the endoplasmic reticulum (ER), many viral infections induce ER stress and trigger the unfolded protein response (UPR). However, the roles of ER stress and UPR in PRV infection remain unclear. In the present study, we demonstrate that the expression of the ER stress marker glucose-regulated protein 78 (GRP78) increased during the early stages of PRV infection, indicating that ER stress was induced. Examination of the three branches of the UPR revealed that the IRE1-XBP1 and eIF2α-ATF4 pathways were activated during PRV infection. In addition, PRV induced apoptosis in later stages of infection through the CHOP-Bcl2 axis. Overexpression of GRP78 or ER stress inducer treatment with thapsigargin could enhance PRV production. Conversely, ER stress inhibitor treatment with tauroursodeoxycholic acid reduced PRV replication. Taken together, our results reveal that PRV infection induces ER stress and activates the IRE1-XBP1 and eIF2α-ATF4 pathways.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Seudorrabia/fisiopatología , Respuesta de Proteína Desplegada/genética , Animales , Antivirales/farmacología , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Proteínas de Choque Térmico/genética , Transducción de Señal/efectos de los fármacos , Porcinos , Ácido Tauroquenodesoxicólico/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Replicación Viral/efectos de los fármacos
13.
Neuron ; 6(6): 957-69, 1991 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1711350

RESUMEN

Uptake and transneuronal passage of wild-type and attenuated strains of a swine alpha-herpesvirus (pseudorabies [PRV]) were examined in rat visual projections. Both strains of virus infected subpopulations of retinal ganglion cells and passed transneuronally to infect retino-recipient neurons in the forebrain. However, the location of infected forebrain neurons varied with the strain of virus. Intravitreal injection of wild-type virus produced two temporally separated waves of infection that eventually reached all known retino-recipient regions of the central neuraxis. By contrast, the attenuated strain of PRV selectively infected a functionally distinct subset of retinal ganglion cells with restricted central projections. The data indicate that projection-specific groups of ganglion cells are differentially susceptible to the two strains of virus and suggest that this sensitivity may be receptor mediated.


Asunto(s)
Encéfalo/microbiología , Herpesvirus Suido 1/fisiología , Neuronas/microbiología , Seudorrabia/fisiopatología , Retina/microbiología , Vías Visuales/microbiología , Animales , Transporte Axonal , Línea Celular , Herpesvirus Suido 1/patogenicidad , Masculino , Ratas , Ratas Endogámicas , Especificidad de la Especie , Porcinos , Factores de Tiempo , Virulencia
14.
J Neuroimmunol ; 180(1-2): 88-103, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16930726

RESUMEN

Ba-DupGreen (BDG) is a highly attenuated, Bartha-derived pseudorabies virus (PRV) expressing green fluorescent protein (GFP) with immediate-early kinetics. Innate immune mechanisms underlying the low infectivity of the virus and the disappearance of infected neurons from the brain were studied at cellular level following injection of BDG into the spleen. The temporal shift in the expression between GFP and viral structural proteins allowed us to discriminate three stages of viral infection in the compromised neurons in correlation with the ongoing local inflammatory response. Iba1/lectin/OX42-positive microglia were recruited to infected neurons within 4-6 h following the initiation of virus replication, incorporated BrdU, isolated the infected cells before the disintegration of their membranes and phagocytosed collapsed neurons. Ex vivo-labeled blood and bone marrow-derived leukocytes, including ED-1-positive macrophages were involved in the immune cell assembly around compromised neurons, which resulted in the complete clearance of infected neurons from the early-infected brain regions.


Asunto(s)
Encéfalo/inmunología , Encéfalo/virología , Herpesvirus Suido 1/inmunología , Inmunidad Innata/inmunología , Seudorrabia/inmunología , Animales , Transporte Axonal/inmunología , Muerte Celular/inmunología , Línea Celular Transformada , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Gliosis/inmunología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/patogenicidad , Macrófagos/inmunología , Masculino , Microglía/inmunología , Vías Nerviosas/inmunología , Vías Nerviosas/virología , Neuronas/inmunología , Neuronas/virología , Fagocitosis/inmunología , Seudorrabia/fisiopatología , Ratas , Ratas Wistar , Sus scrofa , Factores de Tiempo , Replicación Viral/inmunología
15.
Microsc Res Tech ; 66(4): 186-92, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15889426

RESUMEN

Increased density of catecholaminergic nerves in the human polycystic ovary has been observed. The aim of the present study was to investigate the distribution of transsynaptically virus-labeled neurons in the central nervous system from the rat polycystic ovary to see whether is it different or not from that of cycling control rats. To induce a polycystic ovary, a single injection of estradiol valerate was given to adult female rats and 30 days later a neurotropic virus was injected into the right ovary. Rats were sacrificed 72 or 96 hours after viral infection. Weight of the ovaries of the estradiol valerate-treated rats was significantly lower compared to controls, and the histology of the ovaries of the treated rats displayed severely atretic large antral follicles. There was almost no viral labeling in the central nervous system from the ovaries showing precystic morphology, in spite of the fact that such altered organs are rich in nerve fibres. It is assumed that presently unidentified factors in the precystic ovary, presumably related to the link between the immune and the nervous system, might be involved in the infectivity of the virus, and thus be responsible for the lack of viral labeling from such an ovary.


Asunto(s)
Encéfalo/virología , Herpesvirus Suido 1/aislamiento & purificación , Síndrome del Ovario Poliquístico/virología , Seudorrabia/virología , Médula Espinal/virología , Animales , Estradiol/análogos & derivados , Femenino , Inmunohistoquímica , Neuronas/virología , Tamaño de los Órganos , Ovario/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/patología , Seudorrabia/fisiopatología , Ratas , Ratas Sprague-Dawley , Sinapsis/virología
16.
World J Gastroenterol ; 11(38): 5997-6002, 2005 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-16273613

RESUMEN

AIM: To examine the effect of acute infection caused by herpesvirus (pseudorabies virus, PRV) on pancreatic ductal secretion. METHODS: The virulent Ba-DupGreen (BDG) and non-virulent Ka-RREp0lacgfp (KEG) genetically modified strains of PRV were used in this study and both of them contain the gene for green fluorescent protein (GFP). Small intra/interlobular ducts were infected with BDG virus (10(7) PFU/mL for 6 h) or with KEG virus (10(10) PFU/mL for 6 h), while non-infected ducts were incubated only with the culture media. The ducts were then cultured for a further 18 h. The rate of HCO(3)(-) secretion (base efflux -J(B-)) was determined from the buffering capacity of the cells and the initial rate of intracellular acidification (1) after sudden blockage of basolateral base loaders with dihydro-4,4-diisothiocyanatostilbene-2,2-disulfonic acid (500 micromol/L) and amiloride (200 micromol/L), and (2) after alkali loading the ducts by exposure to NH(4)Cl. All the experiments were performed in HCO(3)(-)-buffered Ringer solution at 37 degrees (n = 5 ducts for each experimental condition). Viral structural proteins were visualized by immunohistochemistry. Virally-encoded GFP and immunofluorescence signals were recorded by a confocal laser scanning microscope. RESULTS: The BDG virus infected the majority of accessible cells of the duct as judged by the appearance of GFP and viral antigens in the ductal cells. KEG virus caused a similarly high efficiency of infection. After blockage of basolateral base loaders, BDG infection significantly elevated -J(B-) 24 h after the infection, compared to the non-infected group. However, KEG infection did not modify -J(B-). After alkali loading the ducts, -J(B-) was significantly elevated in the BDG group compared to the control group 24 h after the infection. As we found with the inhibitor stop method, no change was observed in the group KEG compared to the non-infected group. CONCLUSION: Incubation with the BDG or KEG strains of PRV results in an effective infection of ductal epithelial cells. The BDG strain of PRV, which is able to initiate a lytic viral cycle, stimulates HCO(3)(-) secretion in guinea pig pancreatic duct by about four- to fivefold, 24 h after the infection. However, the KEG strain of PRV, which can infect, but fails to replicate, has no effect on HCO(3)(-) secretion. We suggest that this response of pancreatic ducts to virulent PRV infection may represent a defense mechanism against invasive pathogens to avoid pancreatic injury.


Asunto(s)
Conductos Pancreáticos/metabolismo , Seudorrabia/fisiopatología , Animales , Bicarbonatos/metabolismo , Proteínas Fluorescentes Verdes/genética , Cobayas , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/patogenicidad , Técnicas In Vitro , Conductos Pancreáticos/patología , Seudorrabia/patología
17.
J Biol Rhythms ; 15(6): 453-61, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11106062

RESUMEN

Transneuronal tracing of neuronal circuitry with neurotropic viruses has provided valuable insights in the way in which the nervous system imposes temporal organization on physiological processes and behavior. The swine alpha herpes virus known as pseudorabies virus, or PRV, has been particularly useful in this regard. Early studies identified attenuated mutants with selective tropism for visual circuitry involved in circadian regulation, and subsequent experiments employing this virus have provided considerable insight into the polysynaptic organization of the suprachiasmatic nuclei and associated circuitry. This literature, which has emerged during the past decade, is the subject of this mini review.


Asunto(s)
Encéfalo/virología , Ritmo Circadiano , Herpesvirus Suido 1/fisiología , Seudorrabia/fisiopatología , Animales , Herpesvirus Suido 1/genética , Neuronas/patología , Neuronas/fisiología , Seudorrabia/patología , Porcinos
18.
Neurosci Biobehav Rev ; 22(6): 685-94, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9809304

RESUMEN

The development of neurotrophic alpha herpesviruses for transneuronal analysis of neuronal circuitry has emerged from interdisciplinary characterizations of the viral life cycle and the defense response mounted by the nervous system to contain and eliminate the infection. Important findings from a number of fields have combined to provide compelling evidence that these viruses, when used appropriately, are powerful probes of multisynaptic circuits. These studies have also revealed that a number of variables can influence the outcome of infection and should be considered in designing and interpreting data derived from studies employing this experimental approach. The purpose of this paper is to review the literature that has established this experimental approach as a viable method for transynaptic analysis of neuronal circuitry and to define the factors that should be considered in applying this technology.


Asunto(s)
Herpesvirus Suido 1/fisiología , Red Nerviosa/virología , Neuronas/virología , Neurofisiología/métodos , Seudorrabia/fisiopatología , Animales , Herpesvirus Suido 1/ultraestructura , Humanos , Red Nerviosa/patología , Red Nerviosa/fisiología , Neuronas/patología , Neuronas/fisiología , Seudorrabia/patología , Seudorrabia/virología
19.
Endocrinology ; 141(10): 3832-41, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11014240

RESUMEN

Frequent blood sampling via intraatrial cannula revealed daily rhythms of TSH and thyroid hormones in both male and female Wistar rats. Thermic ablation of the biological clock, i.e. the suprachiasmatic nucleus (SCN), eliminated the diurnal peak in circulating TSH and thyroid hormones. In addition, SCN lesions produced a clear decrease of 24-h mean T4 concentrations. A more pronounced effect of SCN-lesions on thyroid hormones, as opposed to TSH, suggested routes of SCN control additional to the neuroendocrine hypothalamopituitary-thyroid axis. Retrograde, transneuronal virus tracing was used to identify the type and localization of neurons in the central nervous system that control the autonomic innervation of the thyroid gland. In the spinal cord and brain stem, both the sympathetic and parasympathetic motorneurons were labeled. By varying the postinoculation survival time, it was possible to follow the viral infection as it proceeded. Subsequently, the pseudorabies virus (PRV) infected neurons in several brain stem cell groups, the paraventricular nucleus of the hypothalamus (PVN) and the central nucleus of the amygdala (second order labeling). Among PRV-infected neurons in the PVN were TRH-containing cells. Third order neurons were found in several hypothalamic cell groups, among which was the SCN. Therefore, we propose that the SCN has a dual control mechanism for thyroid activity by affecting neuroendocrine control of TSH release on the one hand and the autonomic input to the thyroid gland on the other.


Asunto(s)
Núcleo Supraquiasmático/fisiología , Glándula Tiroides/fisiología , Amígdala del Cerebelo/fisiología , Animales , Sistema Nervioso Autónomo/fisiología , Tronco Encefálico/fisiología , Ritmo Circadiano/fisiología , Femenino , Herpesvirus Suido 1 , Masculino , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Neuronas/fisiología , Sistema Nervioso Parasimpático/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Seudorrabia/fisiopatología , Ratas , Ratas Wistar , Médula Espinal/fisiología , Sistema Nervioso Simpático/fisiología , Glándula Tiroides/inervación , Hormonas Tiroideas/sangre , Tirotropina/sangre , Hormona Liberadora de Tirotropina/metabolismo , Tiroxina/sangre
20.
J Comp Neurol ; 471(3): 298-313, 2004 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-14991563

RESUMEN

Intraocular injection of the Bartha strain of pseudorabies virus (PRV Bartha) results in transsynaptic infection of the hypothalamic suprachiasmatic nucleus (SCN), a retinorecipient circadian oscillator. PRV Bartha infection of a limited number of retinorecipient structures, including the SCN, was initially interpreted as the differential infection of a subpopulation of rat retinal ganglion cells, followed by replication and anterograde transport via the optic nerve. A recent report that used a recombinant strain of PRV Bartha (PRV152) expressing enhanced green fluorescent protein demonstrated that SCN infection actually results from retrograde transneuronal transport of the virus via the autonomic innervation of the eye in the golden hamster. In the present study using the rat, the pattern of infection after intravitreal inoculation with PRV152 was examined to determine if infection of the rat SCN is also restricted to retrograde transsynaptic transport. It was observed that infection in preganglionic autonomic nuclei (i.e., Edinger-Westphal nucleus, superior salivatory nucleus, and intermediolateral nucleus) precedes infection in the SCN. Sympathetic superior cervical ganglionectomy did not abolish label in the SCN after intraocular infection, nor did lesions of parasympathetic preganglionic neurons in the Edinger-Westphal nucleus. However, combined Edinger-Westphal nucleus ablation and superior cervical ganglionectomy eliminated infection of the SCN. This observation allowed a detailed examination of the SCN contribution to descending autonomic circuits afferent to the eye. The results indicate that in the rat, as in the hamster, SCN infection after intraocular PRV152 inoculation is by retrograde transsynaptic transport via autonomic pathways to the eye.


Asunto(s)
Ojo/metabolismo , Ojo/virología , Herpesvirus Suido 1/fisiología , Red Nerviosa/metabolismo , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/virología , Sinapsis/metabolismo , Animales , Transporte Biológico/fisiología , Ojo/química , Ojo/inervación , Masculino , Red Nerviosa/química , Red Nerviosa/virología , Seudorrabia/fisiopatología , Seudorrabia/virología , Ratas , Ratas Sprague-Dawley , Núcleo Supraquiasmático/química , Porcinos , Sinapsis/química , Sinapsis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA