Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796067

RESUMO

Neurotropic Alphaherpesvirinae subfamily members such as bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) establish and maintain lifelong latent infections in neurons. Following infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for latency. Certain external stressors can trigger reactivation from latency, in part because activation of the glucocorticoid receptor (GR) stimulates productive infection and promoters that drive expression of key viral transcriptional regulators. The Akt serine/threonine protein kinase family is linked to maintaining latency. For example, Akt3 is detected in more TG neurons during BoHV-1 latency than in reactivation and uninfected calves. Furthermore, Akt signaling correlates with maintaining HSV-1 latency in certain neuronal models of latency. Finally, an active Akt protein kinase is crucial for the ability of the HSV-1 latency-associated transcript (LAT) to inhibit apoptosis in neuronal cell lines. Consequently, we hypothesized that viral and/or cellular factors impair stress-induced transcription and reduce the incidence of reactivation triggered by low levels of stress. New studies demonstrate that Akt1 and Akt2, but not Akt3, significantly reduced GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, the HSV-1 infected cell protein 0 (ICP0) promoter, and the mouse mammary tumor virus long terminal repeat (MMTV-LTR). Akt3, but not Akt1 or Akt2, significantly enhanced neurite formation in mouse neuroblastoma cells, which correlates with repairing damaged neurons. These studies suggest that unique biological properties of the three Akt family members promote the maintenance of latency in differentiated neurons.IMPORTANCE External stressful stimuli are known to increase the incidence of reactivation of Alphaherpesvirinae subfamily members. Activation of the glucocorticoid receptor (GR) by the synthetic corticosteroid dexamethasone (DEX) stimulates bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) reactivation. Furthermore, GR and dexamethasone stimulate productive infection and promoters that drive expression of viral transcriptional regulators. These observations lead us to predict that stress-induced transcription is impaired by factors abundantly expressed during latency. Interestingly, activation of the Akt family of serine/threonine protein kinases is linked to maintenance of latency. New studies reveal that Akt1 and Ak2, but not Akt3, impaired GR- and dexamethasone-mediated transactivation of the BoHV-1 immediate early transcription unit 1 and HSV-1 ICP0 promoters. Strikingly, Akt3, but not Akt1 or Akt2, stimulated neurite formation in mouse neuroblastoma cells, a requirement for neurogenesis. These studies provide insight into how Akt family members may promote the maintenance of lifelong latency.


Assuntos
Herpes Simples/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Células Receptoras Sensoriais/virologia , Animais , Bovinos , Diferenciação Celular , Linhagem Celular Tumoral , Herpes Simples/genética , Herpes Simples/patologia , Herpes Simples/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Camundongos , Neuritos/imunologia , Neuritos/ultraestrutura , Neuritos/virologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/imunologia , Células Receptoras Sensoriais/imunologia , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Ativação Transcricional/imunologia , Gânglio Trigeminal/imunologia , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
2.
J Virol ; 94(20)2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32669337

RESUMO

During primary infection, herpes simplex virus 2 (HSV-2) replicates in epithelial cells and enters neurites to infect neurons of the peripheral nervous system. Growth factors and attractive and repulsive directional cues influence neurite outgrowth and neuronal survival. We hypothesized that HSV-2 modulates the activity of such cues to increase neurite outgrowth. To test this hypothesis, we exposed sensory neurons to nerve growth factor (NGF) and mock- or HSV-2-infected HEK-293T cells, since they express repellents of neurite outgrowth. We show that HEK-293T cells secrete factors that inhibit neurite outgrowth, while infection with HSV-2 strains MS and 333 reduces this repelling phenotype, increasing neurite numbers. The HSV-2-mediated restoration of neurite outgrowth required the activity of NGF. In the absence of infection, however, NGF did not overcome the repulsion mediated by HEK-293T cells. We previously showed that recombinant, soluble glycoprotein G of HSV-2 (rSgG2) binds and enhances NGF activity, increasing neurite outgrowth. However, the effect of gG2 during infection has not been investigated. Therefore, we addressed whether gG2 contributes to overcoming neurite outgrowth repulsion. To do so, we generated viruses lacking gG2 expression and complemented them by exogenous expression of gG2. Overall, our results suggest that HSV-2 infection of nonneuronal cells reduces their repelling effect on neurite outgrowth in an NGF-dependent manner. gG2 contributed to this phenotype, but it was not the only factor. The enhanced neurite outgrowth may facilitate HSV-2 spread from epithelial cells into neurons expressing NGF receptors and increase HSV-2-mediated pathogenesis.IMPORTANCE Herpes simplex virus 2 (HSV-2) is a prevalent human pathogen that establishes lifelong latency in neurons of the peripheral nervous system. Colonization of neurons is required for HSV-2 persistence and pathogenesis. The viral and cellular factors required for efficient infection of neurons are not fully understood. We show here that nonneuronal cells repel neurite outgrowth of sensory neurons, while HSV-2 infection overcomes this inhibition and, rather, stimulates neurite outgrowth. HSV-2 glycoprotein G and nerve growth factor contribute to this phenotype, which may attract neurites to sites of infection and facilitate virus spread to neurons. Understanding the mechanisms that modulate neurite outgrowth and facilitate HSV-2 infection of neurons might foster the development of therapeutics to reduce HSV-2 colonization of the nervous system and provide insights on neurite outgrowth and regeneration.


Assuntos
Herpes Genital/metabolismo , Herpesvirus Humano 2/metabolismo , Fator de Crescimento Neural/metabolismo , Neuritos , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Herpesvirus Humano 2/patogenicidade , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neuritos/metabolismo , Neuritos/virologia , Células Vero
3.
PLoS Pathog ; 16(2): e1008343, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32069324

RESUMO

Neurotropic viral infections continue to pose a serious threat to human and animal wellbeing. Host responses combatting the invading virus in these infections often cause irreversible damage to the nervous system, resulting in poor prognosis. Rabies is the most lethal neurotropic virus, which specifically infects neurons and spreads through the host nervous system by retrograde axonal transport. The key pathogenic mechanisms associated with rabies infection and axonal transmission in neurons remains unclear. Here we studied the pathogenesis of different field isolates of lyssavirus including rabies using ex-vivo model systems generated with mouse primary neurons derived from the peripheral and central nervous systems. In this study, we show that neurons activate selective and compartmentalized degeneration of their axons and dendrites in response to infection with different field strains of lyssavirus. We further show that this axonal degeneration is mediated by the loss of NAD and calpain-mediated digestion of key structural proteins such as MAP2 and neurofilament. We then analysed the role of SARM1 gene in rabies infection, which has been shown to mediate axonal self-destruction during injury. We show that SARM1 is required for the accelerated execution of rabies induced axonal degeneration and the deletion of SARM1 gene significantly delayed axonal degeneration in rabies infected neurons. Using a microfluidic-based ex-vivo neuronal model, we show that SARM1-mediated axonal degeneration impedes the spread of rabies virus among interconnected neurons. However, this neuronal defense mechanism also results in the pathological loss of axons and dendrites. This study therefore identifies a potential host-directed mechanism behind neurological dysfunction in rabies infection. This study also implicates a novel role of SARM1 mediated axonal degeneration in neurotropic viral infection.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Axônios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Raiva/fisiopatologia , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/fisiologia , Transporte Axonal/fisiologia , Axônios/fisiologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/fisiologia , Modelos Animais de Doenças , Gânglios Espinais/virologia , Lyssavirus/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuritos/metabolismo , Neuritos/virologia , Neurônios/metabolismo , Neurônios/virologia , Raiva/metabolismo , Vírus da Raiva/metabolismo , Vírus da Raiva/patogenicidade
4.
J Neurovirol ; 25(3): 384-396, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30758811

RESUMO

Despite the fact that human immunodeficiency virus type 1 (HIV-1) does not enter or replicate in neurons, its infection of a subset of resident brain glia cells (microglia and astrocytes) induces via disparate mechanisms, dysregulation of glutamate metabolism, neurotoxicity, and inflammation. Antiretroviral therapies suppress viral load, but cellular activation and release of proinflammatory factors, some of which is likely related to viral reservoirs, continue to promote a microenvironment that is injurious to neurons. However, the molecular mechanisms remain to be identified. Osteopontin (OPN) is a proinflammatory cytokine-like, extracellular matrix protein that is elevated within the brain and CSF in several neurodegenerative disorders, including HIV-associated cognitive disorder. However, the impact of elevated OPN on neuronal integrity and function in HIV-infected individuals who exhibit cognitive dysfunction remains unknown. In this study, using a neuronal cell line and primary cultures of cortical rat neurons, we identify the mammalian target of rapamycin pathway involvement in a signaling interaction between OPN-ß1-integrins and the HIV-1 envelope glycoprotein, which stimulates neurite growth. These findings link for the first time HIV X4-envelope receptor engagement and osteopontin-mediated signaling through ß1-integrin receptors to the mTOR pathway and alterations in the cytoskeleton of cortical neurons.


Assuntos
Complexo AIDS Demência/metabolismo , Cadeias beta de Integrinas/metabolismo , Neuritos/metabolismo , Osteopontina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Células Cultivadas , HIV-1 , Humanos , Neuritos/virologia , Ratos , Transdução de Sinais/fisiologia , Produtos do Gene env do Vírus da Imunodeficiência Humana
5.
Uirusu ; 68(1): 78-88, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-31105138

RESUMO

Many tick-borne flaviviruses causes fatal encephalitis in humans and animals with severe sequelae. However, it remains unclear how viral replication and pathogenicity contribute to the neurologic manifestations. In this paper, I summarized the specific replication mechanism of tick-borne flaviviruses in neurons and their effect on the pathogenicity of neurological disease. Our findings of the unique virus-host interaction in central nerve system will improve further understanding of the molecular mechanisms of viral replication and the pathogenicity of neurotropic viruses.


Assuntos
Sistema Nervoso Central/virologia , Infecções por Flavivirus/virologia , Flavivirus/fisiologia , Flavivirus/patogenicidade , Carrapatos/virologia , Proteínas não Estruturais Virais/fisiologia , Replicação Viral , Aminoácidos , Animais , Antígenos Virais/metabolismo , Modelos Animais de Doenças , Encefalite Viral/virologia , Flavivirus/genética , Flavivirus/imunologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Neuritos/virologia , Neurônios/virologia , Ratos , Proteínas não Estruturais Virais/química
6.
Microbes Infect ; 19(9-10): 476-484, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28602914

RESUMO

In the field of live viral vaccines production, there is an unmet need for in vitro tests complying a 3R approach (Refine, Replace and Reduce the use of animal experimentation) to replace the post-licensing safety tests currently assayed in animals. Here, we performed a pilot study evaluating whether virulence of rabies virus, RABV, can be forecast by an in vitro test of neurite outgrowth. The rationale to use neurite outgrowth as a read-out for this test is based on the salient property of the cytoplasmic domain of the G-protein (Cyto-G) of virulent RABV strains - not of attenuated RABV strains - to stimulate neurite outgrowth in vitro. We observed that neurite elongation triggered by the Cyto-Gs encoded by different RABV field isolates correlate with the distinct virulence scores obtained in a mouse model of experimental rabies. Our results cast the idea that it could be feasible to predict RABV virulence by testing the in vitro property of a RABV strain to promote neurite outgrowth without the use of animal experimentation.


Assuntos
Alternativas aos Testes com Animais , Glicoproteínas/metabolismo , Neuritos/virologia , Fragmentos de Peptídeos/metabolismo , Vírus da Raiva , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica/fisiologia , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Neuritos/fisiologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Vacina Antirrábica/imunologia , Ratos , Proteínas Recombinantes de Fusão , Proteínas Virais/química , Proteínas Virais/genética
7.
PLoS One ; 9(6): e100196, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24949623

RESUMO

HIV-1 enters the CNS soon after initial systemic infection; within the CNS parenchyma infected and/or activated perivascular macrophages, microglia and astrocytes release viral and cellular toxins that drive secondary toxicity in neurons and other cell types. Our previous work has largely modeled HIV-neuropathology using the individual viral proteins Tat or gp120, with murine striatal neurons as targets. To model disease processes more closely, the current study uses supernatant from HIV-1-infected cells. Supernatant from HIV-1SF162-infected differentiated-U937 cells (HIV+sup) was collected and p24 level was measured by ELISA to assess the infection. Injection drug abuse is a significant risk factor for HIV-infection, and opiate drug abusers show increased HIV-neuropathology, even with anti-retroviral treatments. We therefore assessed HIV+sup effects on neuronal survival and neurite growth/pruning with or without concurrent exposure to morphine, an opiate that preferentially acts through µ-opioid receptors. Effects of HIV+sup ± morphine were assessed on neuronal populations, and also by time-lapse imaging of individual cells. HIV+sup caused dose-dependent toxicity over a range of p24 levels (10-500 pg/ml). Significant interactions occurred with morphine at lower p24 levels (10 and 25 pg/ml), and GSK3ß was implicated as a point of convergence. In the presence of glia, selective neurotoxic measures were significantly enhanced and interactions with morphine were also augmented, perhaps related to a decreased level of BDNF. Importantly, the arrest of neurite growth that occurred with exposure to HIV+sup was reversible unless neurons were continuously exposed to morphine. Thus, while reducing HIV-infection levels may be protective, ongoing exposure to opiates may limit recovery. Opiate interactions observed in this HIV-infective environment were similar, though not entirely concordant, with Tat/gp120 interactions reported previously, suggesting unique interactions with virions or other viral or cellular proteins released by infected and/or activated cells.


Assuntos
HIV-1/fisiologia , Morfina/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/virologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Neuritos/metabolismo , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/virologia , Transdução de Sinais/efeitos dos fármacos , Carga Viral/efeitos dos fármacos
8.
J Virol ; 87(2): 1183-92, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23152506

RESUMO

Bovine herpesvirus 1 (BHV-1) infection induces clinical symptoms in the upper respiratory tract, inhibits immune responses, and can lead to life-threatening secondary bacterial infections. Following acute infection, BHV-1 establishes latency in sensory neurons within trigeminal ganglia, but stress can induce reactivation from latency. The latency-related (LR) RNA is the only viral transcript abundantly expressed in latently infected sensory neurons. An LR mutant virus with stop codons at the amino terminus of the first open reading frame (ORF) in the LR gene (ORF2) is not reactivated from latency, in part because it induces higher levels of apoptosis in infected neurons. ORF2 inhibits apoptosis in transiently transfected cells, suggesting that it plays a crucial role in the latency-reactivation cycle. ORF2 also interacts with Notch1 or Notch3 and inhibits its ability to trans activate certain viral promoters. Notch3 RNA and protein levels are increased during reactivation from latency, suggesting that Notch may promote reactivation. Activated Notch signaling interferes with neuronal differentiation, in part because neurite and axon generation is blocked. In this study, we demonstrated that ORF2 promotes neurite formation in mouse neuroblastoma cells overexpressing Notch1 or Notch3. ORF2 also interfered with Notch-mediated trans activation of the promoter that regulates the expression of Hairy Enhancer of Split 5, an inhibitor of neurite formation. Additional studies provided evidence that ORF2 promotes the degradation of Notch3, but not that of Notch1, in a proteasome-dependent manner. In summary, these studies suggest that ORF2 promotes a mature neuronal phenotype that enhances the survival of infected neurons and consequently increases the pool of latently infected neurons.


Assuntos
Herpesvirus Bovino 1/fisiologia , Neuritos/virologia , Neurônios/virologia , Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Proteínas Virais/metabolismo , Latência Viral , Animais , Linhagem Celular Tumoral , Camundongos , Receptor Notch3
9.
J Virol ; 83(10): 4791-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19279108

RESUMO

In animal models of infection, glycoprotein E (gE) is required for efficient herpes simplex virus type 1 (HSV-1) spread from the inoculation site to the cell bodies of innervating neurons (retrograde direction). Retrograde spread in vivo is a multistep process, in that HSV-1 first spreads between epithelial cells at the inoculation site, then infects neurites, and finally travels by retrograde axonal transport to the neuron cell body. To better understand the role of gE in retrograde spread, we used a compartmentalized neuron culture system, in which neurons were infected in the presence or absence of epithelial cells. We found that gE-deleted HSV-1 (NS-gEnull) retained retrograde axonal transport activity when added directly to neurites, in contrast to the retrograde spread defect of this virus in animals. To better mimic the in vivo milieu, we overlaid neurites with epithelial cells prior to infection. In this modified system, virus infects epithelial cells and then spreads to neurites, revealing a 100-fold retrograde spread defect for NS-gEnull. We measured the retrograde spread defect of NS-gEnull from a variety of epithelial cell lines and found that the magnitude of the spread defect from epithelial cells to neurons correlated with epithelial cell plaque size defect, indicating that gE plays a similar role in both types of spread. Therefore, gE-mediated spread between epithelial cells and neurites likely explains the retrograde spread defect of gE-deleted HSV-1 in vivo.


Assuntos
Células Epiteliais/virologia , Herpes Simples/virologia , Herpesvirus Humano 1/crescimento & desenvolvimento , Neuritos/virologia , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Camundongos , Ratos , Ratos Sprague-Dawley , Células Vero , Ensaio de Placa Viral
10.
J Neurosci ; 25(21): 5207-16, 2005 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-15917460

RESUMO

Invasion of the nervous system and neuronal spread of infection are critical, but poorly understood, steps in the pathogenesis of transmissible spongiform encephalopathies or prion diseases. To characterize pathways for the uptake and intraneuronal trafficking of infectious, protease-resistant prion protein (PrP-res), fluorescent-labeled PrP-res was used to infect a neuronally derived murine cell line (SN56) and adult hamster cortical neurons in primary culture. Concurrent with the establishment of persistent scrapie infection, SN56 cells internalized PrP-res aggregates into vesicles positive for markers for late endosomes and/or lysosomes but not synaptic, early endocytic, or raft-derived vesicles. Internalized PrP-res was then transported along neurites to points of contact with other cells. Similar trafficking was observed with dextran, Alzheimer's Abeta1-42 fibrils and noninfectious recombinant PrP fibrils, suggesting that PrP-res is internalized by a relatively nonspecific pinocytosis or transcytosis mechanism. Hamster cortical neurons were also capable of internalizing and disseminating exogenous PrP-res. Similar trafficking of exogenous PrP-res by cortical neurons cultured from the brains of PrP knock-out mice showed that uptake and neuritic transport did not require the presence of endogenous cellular PrP. These experiments visualize and characterize the initial steps associated with prion infection and transport within neuronal cells.


Assuntos
Doenças do Sistema Nervoso/metabolismo , Neuritos/fisiologia , Neurônios/metabolismo , Proteínas PrPSc/metabolismo , Animais , Transporte Biológico , Western Blotting/métodos , Células Cultivadas , Córtex Cerebral/citologia , Toxina da Cólera/metabolismo , Cricetinae , Diagnóstico por Imagem/métodos , Endopeptidase K/farmacologia , Imunofluorescência/métodos , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Mutagênese/fisiologia , Neuritos/virologia , Neurônios/virologia , Transporte Proteico/fisiologia , Fatores de Tempo , Transfecção/métodos , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
11.
J Virol ; 77(22): 12222-31, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14581559

RESUMO

Borna disease virus (BDV) is a nonsegmented negative-strand RNA virus with a tropism for neurons. Infection with BDV causes neurological diseases in a wide variety of animal species. Although it is known that the virus spreads from neuron to neuron, assembled viral particles have never been visualized in the brains of infected animals. This has led to the hypothesis that BDV spreads as nonenveloped ribonucleoproteins (RNP) rather than as enveloped viral particles. We assessed whether the viral envelope glycoprotein (GP) is required for neuronal dissemination of BDV by using primary cultures of rat hippocampal neurons. We show that upon in vitro infection, BDV replicated and spread efficiently in this system. Despite rapid virus dissemination, very few infectious viral particles were detectable in the culture. However, neutralizing antibodies directed against BDV-GP inhibited BDV spread. In addition, interference with BDV-GP processing by inhibiting furin-mediated cleavage of the glycoprotein blocked virus spread. Finally, antisense treatment with peptide nucleic acids directed against BDV-GP mRNA inhibited BDV dissemination, marking BDV-GP as an attractive target for antiviral therapy against BDV. Together, our results demonstrate that the expression and correct processing of BDV-GP are necessary for BDV dissemination in primary cultures of rat hippocampal neurons, arguing against the hypothesis that the virus spreads from neuron to neuron in the form of nonenveloped RNP.


Assuntos
Vírus da Doença de Borna/fisiologia , Glicoproteínas/fisiologia , Neurônios/virologia , Proteínas do Envelope Viral/fisiologia , Animais , Chlorocebus aethiops , Hipocampo/virologia , Neuritos/virologia , Ratos , Ratos Sprague-Dawley , Células Vero , Proteínas do Envelope Viral/antagonistas & inibidores , Vírion/isolamento & purificação
12.
Brain Pathol ; 13(3): 364-72, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12946025

RESUMO

Two mutations in alpha-synuclein, the main constituent of Lewy bodies, have been identified in familial Parkinson's disease. We have stereotactically injected lentiviral vectors encoding wild-type and A30P mutant human alpha-synuclein in different brain regions (striatum, substantia nigra, amygdala) of mice. Overexpression of alpha-synuclein induced time-dependent neuropathological changes reminiscent of Lewy pathology: abnormal accumulation of alpha-synuclein in cell bodies and neurites, alpha-synuclein-positive neuritic varicosities and cytoplasmic inclusions that stained with ubiquitin antibodies and became larger and more frequent with time. After one year, alpha-synuclein- and ubiquitin-positive neurons displayed a degenerative morphology and a significant loss of alpha-synuclein-positive cells was observed. Similar findings were observed with both the wild-type and the A30P mutant form of alpha-synuclein and this in different brain regions. This indicates that overexpression of alpha-synuclein is sufficient to induce Lewy-like pathology and neurodegeneration and that this effect is not restricted to dopaminergic cells. Our data also demonstrate the use of lentiviral vectors to create animal models for neurodegenerative diseases.


Assuntos
Infecções por Lentivirus/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/patologia , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Tonsila do Cerebelo/virologia , Animais , Western Blotting , Contagem de Células , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Corpo Estriado/virologia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Corpos de Inclusão Viral/metabolismo , Corpos de Inclusão Viral/patologia , Doença por Corpos de Lewy , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Mutação , Proteínas do Tecido Nervoso/genética , Neuritos/metabolismo , Neuritos/patologia , Neuritos/virologia , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/virologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Substância Negra/metabolismo , Substância Negra/patologia , Substância Negra/virologia , Sinucleínas , Fatores de Tempo , Transdução Genética/métodos , Células Tumorais Cultivadas , Tirosina 3-Mono-Oxigenase/metabolismo , Ubiquitina/metabolismo , alfa-Sinucleína
13.
Neuroreport ; 14(8): 1159-62, 2003 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-12821800

RESUMO

Mouse retinal explants were transfected with recombinant adenovirus vector carrying the green fluorescent protein (GFP) gene and the rat bcl-x(L) gene (Adeno-Bcl-xL) to determine its ability to protect retinal ganglion cells against apoptotic cell death and to promote retinal ganglion cell neurite regeneration. Adeno-Bcl-xL-incubated retinas had reduced apoptosis compared with controls. However, neurite regeneration in adeno-treated retinas was less than that of vector-free retina. These results suggest that the usefulness of adenovirus vectors for gene therapy for retinal ganglion cells may be limited.


Assuntos
Regeneração Nervosa/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Retina/citologia , Adenoviridae/genética , Animais , Morte Celular , DNA Recombinante/genética , DNA Recombinante/metabolismo , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Indóis/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NG-Nitroarginina Metil Éster/farmacologia , Regeneração Nervosa/fisiologia , Neuritos/fisiologia , Neuritos/virologia , Neurônios , Técnicas de Cultura de Órgãos , Ratos , Retina/virologia , Antígenos Thy-1/metabolismo , Transfecção/métodos , Transfecção/veterinária , Proteína bcl-X
14.
J Neurovirol ; 8(4): 306-17, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12161815

RESUMO

Cultures of purified rat embryonic spinal cord motoneurons were used to investigate the capacity of the neurons to survive rabies virus infection in vitro. In crude primary spinal cord cultures, neurons did not survive more than 2 days after rabies virus infection with the fixed strain Challenge Virus Standard. In contrast, virus-infected purified motoneurons resisted cytolysis for at least 7 days, as also did infected motoneurons treated with conditioned medium sampled from rabies virus-infected crude spinal cord cultures. This survival rate was also observed when motoneurons were grown in the presence of astrocytes or fibroblasts and it was not dependent on the presence of growth factors in the culture medium. Moreover, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling experiments showed that only 30% of infected motoneurons were apoptotic after 7 days of infection. In vivo, despite the massive infection of the spinal cord in infected rat neonates, the moderate number of apoptotic cells in the ventral horn suggests that only a few motoneurons were affected by this mechanism of cell death. Morphometric analyses showed that motoneurons' axon elongated at a comparable rate in virus-infected and noninfected cultures, a sign of high metabolic activity maintained in rabies virus-infected motoneurons. In contrast, hippocampus neurons were susceptible to rabies virus infection, because 70% of infected neurons were destroyed within 3 days, a large proportion of them being apoptotic. These experiments suggest that spinal cord motoneurons consist in a neuronal population that survive rabies virus infection because the viral induction of apoptosis is delayed in these neurons. They suggest also that paralyses frequently observed in rabid animals could be the consequence of dysfunctions of the locomotor network or of the spinal cord motoneurons themselves, whose parameters could be studied in vitro.


Assuntos
Neurônios Motores/virologia , Raiva/patologia , Medula Espinal/citologia , Animais , Apoptose , Técnicas de Cultura de Células/métodos , Sobrevivência Celular , Células Cultivadas , Suscetibilidade a Doenças , Feto/citologia , Hipocampo/citologia , Marcação In Situ das Extremidades Cortadas , Neurônios Motores/ultraestrutura , Neuritos/virologia , Ratos , Ratos Wistar
15.
FASEB J ; 16(8): 878-80, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11967226

RESUMO

Reciprocal transmission between epithelia and sensory neurons of the peripheral nervous system is a crucial step in the life cycle of herpes simplex virus type 1 (HSV-1) and related alphaherpesviruses. In searching for an easy-to-perform and generally applicable experimental approach that enables the direct analysis of virus transfer between primary epithelial cells and sensory neurites, we investigated the spread of HSV-1 in a dual-chamber organ model comprising chick embryonic corneal epithelia and trigeminal sensory neurons. Embryonic chick corneal and trigeminal tissues were found to be permissive for productive infection with HSV-1. Our data show that HSV-1 efficiently enters neurites re-innervating the cornea and reaches the ganglion explant by retrograde axonal transport, with the first antigen-positive cells being detectable approximately 12 h postinfection. After direct infection of trigeminal tissues, the virus is transported by anterograde axonal transport to the corneal epithelium, causing a visible cytopathic effect approximately 48 h postinfection. These results suggest that the organ model presented in this study holds particular promise for the direct observation and molecular analysis of herpes simplex virus spread between primary epithelia and sensory neurons and that it may be an attractive alternative to current experimental approaches based on laboratory animals or human fetal tissues.


Assuntos
Epitélio Corneano/virologia , Herpesvirus Humano 1/crescimento & desenvolvimento , Neuritos/virologia , Gânglio Trigeminal/virologia , Animais , Transporte Axonal , Células Cultivadas , Embrião de Galinha , Chlorocebus aethiops , Epitélio Corneano/citologia , Técnicas de Cultura de Órgãos , Gânglio Trigeminal/citologia , Células Vero , Replicação Viral
16.
Am J Dermatopathol ; 19(2): 133-7, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9129697

RESUMO

Cutaneous herpesvirus infection is a common viral disorder manifest by epidermal and/or mucosal vesicle formation. Though it is believed that the virus most likely resides in regional sensory ganglia following primary infection and that cutaneous involvement represents reactivation of a latent infection, the histopathology of cutaneous nerves in sites of disease has not been well characterized. In order to assess and characterize the pathologic changes of these nerves, we retrospectively examined 54 cases of cutaneous and mucosal herpesvirus infection as defined by the presence of diagnostic multinucleate epithelial giant cells that demonstrated viral cytopathic effect. Dermal nerves were evaluable in 48 of 54 cases. All cases showed perineural inflammation that consisted of a dense mixed lymphocyte-polymorphonuclear cell infiltrate. Twenty-six cases exhibited intraneural infiltrations accompanied by Schwann cell hypertrophy with nuclear eosinophilia and pyknosis. Frank neuronal necrosis was present in 21 cases, with viral cytopathic effect evident within neurons of four cases. The degree of peri- and intraneural inflammation correlated with the severity of the inflammatory response within the dermis in most cases; however, in eight cases there was inflammatory involvement of neurovascular structures distant from and out of proportion to dermal and epidermal changes. Immunoperoxidase staining using a polyvalent antibody to human herpesvirus was performed in two cases and demonstrated viral antigen within nerve twigs. This pattern of peripheral nerve twig inflammation, along with the occurrence of more distant neural involvement, may prove to have diagnostic implications and serve as a clue in the recognition of cutaneous herpesvirus infection, particularly in cases with subtle or absent epidermal alteration. Furthermore, the presence of inflammation within and around nerves as well as degenerative changes suggest that nerve twigs are not passive conduits for viral spread but may be directly involved in infection.


Assuntos
Herpes Simples/patologia , Doenças do Sistema Nervoso Periférico/virologia , Pele/inervação , Antígenos Virais/análise , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Epiderme/patologia , Epiderme/virologia , Epitélio/patologia , Epitélio/virologia , Gânglios Sensitivos/virologia , Células Gigantes/patologia , Células Gigantes/virologia , Herpes Zoster/patologia , Herpesvirus Humano 3/crescimento & desenvolvimento , Herpesvirus Humano 3/imunologia , Humanos , Hipertrofia , Técnicas Imunoenzimáticas , Linfócitos/patologia , Linfócitos/virologia , Necrose , Neuritos/imunologia , Neuritos/ultraestrutura , Neuritos/virologia , Neurite (Inflamação)/patologia , Neurite (Inflamação)/virologia , Neurônios/patologia , Neurônios/virologia , Neutrófilos/patologia , Neutrófilos/virologia , Doenças do Sistema Nervoso Periférico/patologia , Estudos Retrospectivos , Células de Schwann/patologia , Células de Schwann/virologia , Simplexvirus/crescimento & desenvolvimento , Simplexvirus/imunologia , Pele/patologia , Pele/virologia , Ativação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...