Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 152(4): 743-54, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23415224

RESUMO

Long noncoding RNAs (lncRNAs) are increasingly appreciated as regulators of cell-specific gene expression. Here, an enhancer-like lncRNA termed NeST (nettoie Salmonella pas Theiler's [cleanup Salmonella not Theiler's]) is shown to be causal for all phenotypes conferred by murine viral susceptibility locus Tmevp3. This locus was defined by crosses between SJL/J and B10.S mice and contains several candidate genes, including NeST. The SJL/J-derived locus confers higher lncRNA expression, increased interferon-γ (IFN-γ) abundance in activated CD8(+) T cells, increased Theiler's virus persistence, and decreased Salmonella enterica pathogenesis. Transgenic expression of NeST lncRNA alone was sufficient to confer all phenotypes of the SJL/J locus. NeST RNA was found to bind WDR5, a component of the histone H3 lysine 4 methyltransferase complex, and to alter histone 3 methylation at the IFN-γ locus. Thus, this lncRNA regulates epigenetic marking of IFN-γ-encoding chromatin, expression of IFN-γ, and susceptibility to a viral and a bacterial pathogen.


Assuntos
Suscetibilidade a Doenças , Epigênese Genética , Interferon gama/genética , RNA Longo não Codificante/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Infecções por Cardiovirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Theilovirus/imunologia
2.
Acta Neuropathol ; 137(6): 961-980, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30927072

RESUMO

Progressive aggregation of the protein alpha-synuclein (α-syn) and loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) are key histopathological hallmarks of Parkinson's disease (PD). Accruing evidence suggests that α-syn pathology can propagate through neuronal circuits in the brain, contributing to the progressive nature of the disease. Thus, it is therapeutically pertinent to identify modifiers of α-syn transmission and aggregation as potential targets to slow down disease progression. A growing number of genetic mutations and risk factors has been identified in studies of familial and sporadic forms of PD. However, how these genes affect α-syn aggregation and pathological transmission, and whether they can be targeted for therapeutic interventions, remains unclear. We performed a targeted genetic screen of risk genes associated with PD and parkinsonism for modifiers of α-syn aggregation, using an α-syn preformed-fibril (PFF) induction assay. We found that decreased expression of Lrrk2 and Gba modulated α-syn aggregation in mouse primary neurons. Conversely, α-syn aggregation increased in primary neurons from mice expressing the PD-linked LRRK2 G2019S mutation. In vivo, using LRRK2 G2019S transgenic mice, we observed acceleration of α-syn aggregation and degeneration of dopaminergic neurons in the SNpc, exacerbated degeneration-associated neuroinflammation and behavioral deficits. To validate our findings in a human context, we established a novel human α-syn transmission model using induced pluripotent stem cell (iPS)-derived neurons (iNs), where human α-syn PFFs triggered aggregation of endogenous α-syn in a time-dependent manner. In PD subject-derived iNs, the G2019S mutation enhanced α-syn aggregation, whereas loss of LRRK2 decreased aggregation. Collectively, these findings establish a strong interaction between the PD risk gene LRRK2 and α-syn transmission across mouse and human models. Since clinical trials of LRRK2 inhibitors in PD are currently underway, our findings raise the possibility that these may be effective in PD broadly, beyond cases caused by LRRK2 mutations.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/fisiologia , Mutação de Sentido Incorreto , Neurônios/metabolismo , Doença de Parkinson/genética , Agregação Patológica de Proteínas/etiologia , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Comportamento Exploratório , Glucosilceramidase/genética , Hipocampo/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/deficiência , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Doença de Parkinson/patologia , Parte Compacta da Substância Negra/patologia , Cultura Primária de Células , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Interferência de RNA , Proteínas Recombinantes/metabolismo , Teste de Desempenho do Rota-Rod
3.
Neurobiol Dis ; 109(Pt B): 219-225, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28323023

RESUMO

Intra-neuronal protein aggregates made of fibrillar alpha-synuclein (α-syn) are the hallmark of Parkinson's disease (PD). With time, these aggregates spread through the brain following axonal projections. Understanding the mechanism of this spread is central to the study of the progressive nature of PD. Here we review data relevant to the uptake, transport and release of α-syn fibrils. We summarize several cell surface receptors that regulate the uptake of α-syn fibrils by neurons. The aggregates are then transported along axons, both in the anterograde and retrograde direction. The kinetics of transport suggests that they are part of the slow component b of axonal transport. Recent findings indicate that aggregated α-syn is secreted by neurons by non-canonical pathways that may implicate various molecular chaperones including USP19 and the DnaJ/Hsc70 complex. Additionally, α-syn fibrils may also be released and transmitted from neuron-to-neuron via exosomes and tunneling nanotubes. Understanding these different mechanisms and molecular players underlying α-syn spread is crucial for the development of therapies that could halt the progression of α-syn-related degenerative diseases.


Assuntos
Transporte Axonal/fisiologia , Neurônios/metabolismo , alfa-Sinucleína/metabolismo , Animais , Humanos , Doenças Neurodegenerativas/metabolismo
4.
Acta Neuropathol ; 131(4): 539-48, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26820848

RESUMO

Accruing evidence suggests that prion-like behavior of fibrillar forms of α-synuclein, ß-amyloid peptide and mutant huntingtin are responsible for the spread of the lesions that characterize Parkinson disease, Alzheimer disease and Huntington disease, respectively. It is unknown whether these distinct protein assemblies are transported within and between neurons by similar or distinct mechanisms. It is also unclear if neuronal death or injury is required for neuron-to-neuron transfer. To address these questions, we used mouse primary cortical neurons grown in microfluidic devices to measure the amounts of α-synuclein, Aß42 and HTTExon1 fibrils transported by axons in both directions (anterograde and retrograde), as well as to examine the mechanism of their release from axons after anterograde transport. We observed that the three fibrils were transported in both anterograde and retrograde directions but with strikingly different efficiencies. The amount of Aß42 fibrils transported was ten times higher than that of the other two fibrils. HTTExon1 was efficiently transported in the retrograde direction but only marginally in the anterograde direction. Finally, using neurons from two distinct mutant mouse strains whose axons are highly resistant to neurodegeneration (Wld(S) and Sarm1(-/-)), we found that the three different fibrils were secreted by axons after anterograde transport, in the absence of axonal lysis, indicating that trans-neuronal spread can occur in intact healthy neurons. In summary, fibrils of α-synuclein, Aß42 and HTTExon1 are all transported in axons but in directions and amounts that are specific of each fibril. After anterograde transport, the three fibrils were secreted in the medium in the absence of axon lysis. Continuous secretion could play an important role in the spread of pathology between neurons but may be amenable to pharmacological intervention.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Transporte Axonal/fisiologia , Proteína Huntingtina/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Ácido Poliglutâmico/metabolismo , alfa-Sinucleína/metabolismo , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Encéfalo/patologia , Toxina da Cólera/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Embrião de Mamíferos , Glutationa Peroxidase/metabolismo , Humanos , Proteína Huntingtina/genética , Dispositivos Lab-On-A-Chip , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Ácido Poliglutâmico/genética , Príons/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
Ann Neurol ; 72(4): 517-24, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23109146

RESUMO

OBJECTIVE: The lesions of Parkinson disease spread through the brain in a characteristic pattern that corresponds to axonal projections. Previous observations suggest that misfolded α-synuclein could behave as a prion, moving from neuron to neuron and causing endogenous α-synuclein to misfold. Here, we characterized and quantified the axonal transport of α-synuclein fibrils and showed that fibrils could be transferred from axons to second-order neurons following anterograde transport. METHODS: We grew primary cortical mouse neurons in microfluidic devices to separate somata from axonal projections in fluidically isolated microenvironments. We used live-cell imaging and immunofluorescence to characterize the transport of fluorescent α-synuclein fibrils and their transfer to second-order neurons. RESULTS: Fibrillar α-synuclein was internalized by primary neurons and transported in axons with kinetics consistent with slow component-b of axonal transport (fast axonal transport with saltatory movement). Fibrillar α-synuclein was readily observed in the cell bodies of second-order neurons following anterograde axonal transport. Axon-to-soma transfer appeared not to require synaptic contacts. INTERPRETATION: These results support the hypothesis that the progression of Parkinson disease can be caused by neuron-to-neuron spread of α-synuclein aggregates and that the anatomical pattern of progression of lesions between axonally connected areas results from the axonal transport of such aggregates. That the transfer did not appear to be trans-synaptic gives hope that α-synuclein fibrils could be intercepted by drugs during the extracellular phase of their journey.


Assuntos
Transporte Axonal/fisiologia , Neurofibrilas/fisiologia , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , alfa-Sinucleína/fisiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Corantes Fluorescentes , Imuno-Histoquímica , Maleimidas , Camundongos , Técnicas Analíticas Microfluídicas , Microscopia Confocal , Microscopia de Fluorescência , Neuroglia/fisiologia , Fragmentos de Peptídeos/metabolismo
6.
Ann Neurol ; 68(1): 6-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20582990

RESUMO

Discussing the problem of multiple sclerosis and viruses should not be limited to reviewing the epidemiological evidence in favor, or against, a particular candidate, such as Epstein-Barr virus or human herpes virus 6. In this text, I discuss the difficulty of going from association to causation in human epidemiology; the fact that viruses can trigger or prevent autoimmunity; the problem of our very limited knowledge of the viruses that we harbor as part of our metagenome; and the role of such viral flora in multifactorial diseases and also, possibly, in health.


Assuntos
Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Animais , Autoimunidade , Humanos , Metagenoma , Esclerose Múltipla/epidemiologia , Esclerose Múltipla/genética
7.
PLoS Pathog ; 3(2): e23, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17305428

RESUMO

Theiler's virus, a picornavirus, persists for life in the central nervous system of mouse and causes a demyelinating disease that is a model for multiple sclerosis. The virus infects neurons first but persists in white matter glial cells, mainly oligodendrocytes and macrophages. The mechanism, by which the virus traffics from neurons to glial cells, and the respective roles of oligodendrocytes and macrophages in persistence are poorly understood. We took advantage of our previous finding that the shiverer mouse, a mutant with a deletion in the myelin basic protein gene (Mbp), is resistant to persistent infection to examine the role of myelin in persistence. Using immune chimeras, we show that resistance is not mediated by immune responses or by an efficient recruitment of inflammatory cells into the central nervous system. With both in vivo and in vitro experiments, we show that the mutation does not impair the permissiveness of neurons, oligodendrocytes, and macrophages to the virus. We demonstrate that viral antigens are present in cytoplasmic channels of myelin during persistent infection of wild-type mice. Using the optic nerve as a model, we show that the virus traffics from the axons of retinal ganglion cells to the cytoplasmic channels of myelin, and that this traffic is impaired by the shiverer mutation. These results uncover an unsuspected axon to myelin traffic of Theiler's virus and the essential role played by the infection of myelin/oligodendrocyte in persistence.


Assuntos
Encéfalo/virologia , Bainha de Mielina/fisiologia , Theilovirus/crescimento & desenvolvimento , Animais , Antígenos Virais/análise , Células da Medula Óssea/fisiologia , Infecções por Cardiovirus/imunologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C3H , Mutação , Bainha de Mielina/genética , Oligodendroglia/virologia , Theilovirus/patogenicidade
9.
Virologie (Montrouge) ; 15(1): 3-5, 2011 Feb 01.
Artigo em Francês | MEDLINE | ID: mdl-36151648
10.
FASEB J ; 18(7): 863-5, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15033926

RESUMO

Infection of the central nervous system by Borna disease virus (BDV) provides a unique model to study the mechanisms whereby a persistent viral infection can impair neuronal function and cause behavioral diseases reminiscent of mood disorders, schizophrenia, or autism in humans. In the present work, we studied the effect of BDV infection on the response of hippocampal neurons, the main target for this virus, to the neurotrophin BDNF. We showed that persistent infection did not affect neuronal survival or morphology. However, it blocked BDNF-induced ERK 1/2 phosphorylation, despite normal expression of the TrkB BDNF receptor. In addition, BDNF-induced expression of synaptic vesicle proteins was abrogated, which resulted in severely impaired synaptogenesis and defects in synaptic organization. Thus, we provide the first evidence that a virus can interfere specifically with neurotrophin-regulated neuroplasticity, thereby hampering proper neuronal connectivity. These results may help to understand the behavioral disorders associated with BDV infection.


Assuntos
Vírus da Doença de Borna/fisiologia , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neurônios/virologia , Sinapses/ultraestrutura , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Células Cultivadas/ultraestrutura , Células Cultivadas/virologia , Efeito Citopatogênico Viral , Hipocampo/citologia , Hipocampo/virologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transtornos Mentais/virologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas do Tecido Nervoso/biossíntese , Plasticidade Neuronal , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/ultraestrutura , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo
11.
Brain Pathol ; 13(4): 519-33, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14655757

RESUMO

Theiler virus persists and induces immune-mediated demyelination in susceptible mice and serves as a model of multiple sclerosis. Previously, we identified 4 markers--D14Mit54, D14Mit60, D14Mit61, and D14Mit90--in a 40-cM region of chromosome 14 that are associated with demyelination in a cross between susceptible DBA/2 and resistant B10.D2 mice. We generated congenic-inbred mice to examine the contribution of this 40-cM region to disease. DBA Chr.14B10 mice, containing the chromosomal segment marked by the microsatellite polymorphisms, developed less spinal cord demyelination than did DBA/2 mice. More demyelination was found in the reciprocal congenic mouse B10.D2 Chr.14D2 than in the B10.D2 strain. Introduction of the DBA/2 chromosomal region onto the B10.D2 genetic background resulted in more severe disease in the striatum and cortex relative to B10.D2 mice. The importance of the marked region of chromosome 14 is indicated by the decrease in neurological performance using the Rotarod test during chronic disease in B10.D2 Chr.14D2 mice in comparison to B10.D2 mice. Viral replication was increased in B10.D2 Chr.14D2 mice as determined by quantitative real-time RT-PCR. These results indicate that the 40-cM region on chromosome 14 of DBA/2 mice contributes to viral persistence, subsequent demyelination, and loss of neurological function.


Assuntos
Encefalopatias/virologia , Cromossomos , Doenças Desmielinizantes , Esclerose Múltipla/genética , Poliomielite/genética , Animais , Comportamento Animal , Encefalopatias/veterinária , Doenças Desmielinizantes/veterinária , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Imuno-Histoquímica , Meningite/patologia , Meningite/virologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos DBA , Repetições de Microssatélites , Atividade Motora/fisiologia , Esclerose Múltipla/veterinária , Esclerose Múltipla/virologia , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Teste de Desempenho do Rota-Rod/métodos , Teste de Desempenho do Rota-Rod/veterinária , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/virologia , Theilovirus/metabolismo , Theilovirus/patogenicidade , Fatores de Tempo , Vírion/metabolismo
12.
PLoS One ; 2(12): e1331, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18159229

RESUMO

We showed previously that Theiler's virus, a neurotropic non-enveloped picornavirus of mouse, traffics from the axon of infected neurons into the surrounding myelin. When this traffic is interrupted, as in the shiverer mouse which bears a mutation in the myelin basic protein gene, the virus is unable to persist in the central nervous system. In the present work, we used the Wld(s) mutant mouse, a strain in which axonal degeneration is considerably slowed down, to show that axon to myelin traffic takes place in the absence of axon degeneration. Our results suggest the existence of a mechanism of transfer of axonal cytoplasm into the myelin which Theiler's virus might exploit to ensure its persistence.


Assuntos
Axônios , Bainha de Mielina/fisiologia , Theilovirus/fisiologia , Animais , Feminino , Camundongos , Camundongos Mutantes , Nervo Óptico/virologia , Retina/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Annu Rev Microbiol ; 59: 279-98, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16153171

RESUMO

Theiler's virus causes a persistent and demyelinating infection of the central nervous system of the mouse, which is one of the best animal models to study multiple sclerosis. This review focuses on the mechanism of persistence. The virus infects neurons for a few weeks and then shifts to white matter, where it persists in glial cells and macrophages. Oligodendrocytes are crucial host cells, as shown by the resistance to persistent infection of mice bearing myelin mutations. Two viral proteins, L and L*, contribute to persistence by interfering with host defenses. L, a small zinc-finger protein, restricts the production of interferon. L*, a unique example of a picornaviral protein translated from an overlapping open reading frame, facilitates the infection of macrophages. Susceptibility to persistent infection, which varies among inbred mouse strains, is multigenic. H2 class I genes have a major effect on susceptibility. Among several non-H2 susceptibility loci, Tmevp3 appears to regulate the expression of important cytokines.


Assuntos
Infecções por Cardiovirus/complicações , Doenças do Sistema Nervoso Central/genética , Doenças Desmielinizantes/genética , Theilovirus/fisiologia , Theilovirus/patogenicidade , Sequência de Aminoácidos , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Doenças do Sistema Nervoso Central/etiologia , Doenças do Sistema Nervoso Central/virologia , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Locos de Características Quantitativas , Theilovirus/genética , Proteínas Virais/química , Proteínas Virais/genética
14.
Vaccine ; 23(36): 4463-72, 2005 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-15993518

RESUMO

Live attenuated measles vaccine (MV) could provide a safe and efficient pediatric vaccination vector to immunize children simultaneously against measles and human immunodeficiency virus type 1 (HIV-1). To evaluate the capacity of a vector derived from the certified Schwarz measles vaccine (MVSchw) to prime effective cytotoxic T cells (CTL) and broad neutralizing antibodies against HIV-1 conserved epitopes, we generated recombinant MVSchw viruses expressing HIV-1 antigens. We demonstrate that a recombinant MVSchw virus expressing an HIV-1-derived CTL polyepitope primes effective HLA-A0201-restricted CTLs against multiple conserved HIV-1 epitopes in mice susceptible to measles and humanized for the major histocompatibility complex (MHC) class-I molecule HLA-A0201. Additionally, we show that a recombinant MVSchw virus expressing an HIV-1(89.6) gp140 glycoprotein whose hyper variable V1, V2 and V3 loops were deleted (DeltaV1V2V3gp140), induces broadly neutralizing antibodies against HIV-1 primary isolates. These results show that the MVSchw pediatric vaccination vector induces efficient cellular and humoral HIV-specific immune responses.


Assuntos
Vacinas contra a AIDS/imunologia , Anticorpos Anti-HIV/sangue , HIV-1/imunologia , Antígenos HLA-A/imunologia , Vacina contra Sarampo/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas Sintéticas/imunologia , Animais , Antígenos CD/análise , Contagem de Linfócito CD4 , Epitopos , Produtos do Gene env/imunologia , Vetores Genéticos , Imunização , Interferon-alfa/análise , Vacina contra Sarampo/genética , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/análise , Camundongos , Vacinas Atenuadas/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana
15.
J Infect Dis ; 191(2): 207-14, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15609230

RESUMO

The Schwarz strain of measles virus (MV), a live attenuated RNA virus, is one of the safest and most effective human vaccines available. Immunization with MV vaccine expressing heterologous antigen is an attractive strategy to prevent emerging viral diseases. West Nile virus (WNV), which recently emerged in North America, is an important mosquito-borne flavivirus that causes numerous cases of human encephalitis, thus urging the development of a vaccine. To evaluate the efficacy of recombinant MV for the prevention of WNV encephalitis, we constructed a live attenuated Schwarz MV (MVSchw-sE(WNV)) expressing the secreted form of the envelope glycoprotein from the virulent IS-98-ST1 strain of WNV. Inoculation of MV-susceptible mice with MVSchw-sE(WNV) induced both high levels of specific anti-WNV neutralizing antibodies and protection from a lethal challenge with WNV. Passive administration with antisera to MVSchw-sE(WNV) prevented WNV encephalitis in BALB/c mice challenged with a high dose of WNV. The present study is the first to report that a recombinant live attenuated vector based on an approved and widely used MV vaccine can protect against a heterologous, medically important pathogen.


Assuntos
Encefalite/prevenção & controle , Vacina contra Sarampo/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/uso terapêutico , Vírus do Nilo Ocidental/imunologia , Animais , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Clonagem Molecular , Encefalite/virologia , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Camundongos , Camundongos Endogâmicos BALB C , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/patogenicidade
16.
J Infect Dis ; 191(2): 255-63, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15609236

RESUMO

Human T cell lymphotropic virus (HTLV) type I is the agent of adult T cell leukemia and HTLV-I-associated myelopathy. Because its pathogenesis is not well understood, a mouse model of HTLV-I infection would be valuable. We report the infection of adult BALB/c, C3H/He, 129Sv, and 129Sv IFNAR(-/-) mice with an infectious chimeric HTLV-I provirus bearing the Moloney-murine leukemia virus (Mo-MuLV) envelope glycoprotein truncated for the C-terminal R peptide. Mice were persistently infected (500-800 proviral DNA copies/10(5) splenocytes) for at least 20 weeks after inoculation. The chimeric virus disseminated to several organs, such as spleen, thymus, lung, brain, and spinal cord. The amplification of proviral integration sites showed an oligoclonal integration resembling that reported in HTLV-I-infected humans. Infected mice developed lasting humoral and cellular immune responses. This DeltaR HTLV-I/Mo-MuLV chimeric virus, with the Mo-MuLV Env tropism and HTLV-I replication characteristics, could provide a mouse model of HTLV-I infection.


Assuntos
Quimera/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Vírus da Leucemia Murina de Moloney/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Formação de Anticorpos/imunologia , Linhagem Celular , Modelos Animais de Doenças , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Imunidade Celular/imunologia , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Provírus , Proteínas do Envelope Viral/genética
17.
J Virol ; 76(24): 12823-33, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438607

RESUMO

Theiler's virus, a murine picornavirus, causes a persistent infection of macrophage/microglial cells in the central nervous systems of SJL/J mice. Viral replication is restricted in the majority of infected cells, whereas a minority of them contain large amounts of viral RNA and antigens. For the present work, we infected primary cultures of bone marrow monocytes/macrophages from SJL/J mice with Theiler's virus. During the first 10 h postinfection (p.i.), infected monocytes/macrophages were round and covered with filopodia and contained large amounts of viral antigens throughout their cytoplasm. Later on, they were large, flat, and devoid of filopodia and they contained only small amounts of viral antigens distributed in discrete inclusions. These two types of infected cells were very reminiscent of the two types of infected macrophages found in the spinal cords of SJL/J mice. At the peak of virus production, the viral yield per cell was approximately 200 times lower than that for BHK-21 cells. Cell death occurred in the culture during the first 24 h p.i. but not thereafter. No infected cells could be detected after 4 days p.i., and the infection never spread to 100% of the cells. This restriction was unchanged by treating the medium at pH 2 but was abolished by treating it with a neutralizing alpha/beta interferon antiserum, indicating a role for this cytokine in limiting virus expression in monocyte/macrophage cultures. The role of alpha/beta interferon was confirmed by the observation that monocytes/macrophages from IFNA/BR(-/-) mice were fully permissive.


Assuntos
Células da Medula Óssea/virologia , Macrófagos/virologia , Monócitos/virologia , Theilovirus/fisiologia , Animais , Células Cultivadas , Feminino , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Camundongos
18.
J Virol ; 78(23): 13356-61, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542687

RESUMO

We compared the infection of bone marrow macrophages by the DA and GDVII strains of Theiler's virus and by two viruses constructed by exchanging the DA and GDVII capsids. The replication of the GDVII strain and of both chimeric viruses was restricted in macrophages. Therefore, the infection of macrophages requires both capsid and noncapsid viral determinants.


Assuntos
Capsídeo/fisiologia , Macrófagos/virologia , Theilovirus/fisiologia , Animais , Células Cultivadas , Feminino , Citometria de Fluxo , Camundongos , Replicação Viral
19.
J Virol ; 76(11): 5807-12, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11992010

RESUMO

The DA strain of Theiler's murine encephalomyelitis virus persists in the white matter of the spinal cords of susceptible mice. Previous results showed that the difference in susceptibility to viral persistence between the susceptible SJL/J strain and the resistant B10.S strain was due to multiple non-H-2 loci. The respective roles of hematopoietic and nonhematopoietic cells in this difference have been evaluated with bone marrow chimeras. The results show that non-H-2 loci with a major effect on susceptibility are expressed in hematopoietic cells. However, the study of the SJL.B10-D10Mit180-D10Mit74 congenic line suggests that other loci expressed in nonhematopoietic cells also play a role.


Assuntos
Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Antígenos H-2/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Poliomielite/imunologia , Theilovirus/imunologia , Quimeras de Transplante/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Genótipo , Antígenos H-2/genética , Hematopoese/genética , Hematopoese/imunologia , Antígeno de Histocompatibilidade H-2D , Complexo Principal de Histocompatibilidade/genética , Camundongos , Poliomielite/genética , Poliomielite/virologia , RNA Viral , Carga Viral , Latência Viral
20.
J Virol ; 77(10): 5632-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12719555

RESUMO

The Tmevp3 locus controls the load of Theiler's virus RNA during persistent infection of the mouse central nervous system (CNS). We identified a candidate gene at this locus, Tmevpg1, by using a positional cloning approach. Tmevpg1 and its human ortholog, TMEVPG1, are expressed in the immune system and encode what appears to be a noncoding RNA. They are located in a cluster of cytokine genes that includes the genes for gamma interferon and one or two homolog of interleukin-10. We now report that Tmevpg1 is expressed in CNS-infiltrating immune cells of resistant B10.S mice, but not in those of susceptible SJL/J mice, following inoculation with Theiler's virus. The pattern of expression of Tmevpg1 is the same in B10.S mice and in SJL/J mice congenic for the resistant B10.S haplotype of Tmevp3. Nineteen polymorphisms were identified when the Tmevpg1 genes of B10.S and SJL/J mice were compared. Interestingly, Tmevpg1 is down regulated after in vitro stimulation of murine CD4(+) or CD8(+) splenocytes, whereas Ifng is up regulated. Similar patterns of expression of TMEVPG1 and IFNG were observed in human NK cells and CD4(+) and CD8(+) T lymphocytes. Therefore, Tmevpg1 is a strong candidate gene for the Tmevp3 locus and may be involved in the control of Ifng gene expression.


Assuntos
Regulação da Expressão Gênica , Predisposição Genética para Doença , Interferon gama/metabolismo , RNA/genética , Telômero/genética , Theilovirus/patogenicidade , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Sistema Nervoso Central/metabolismo , Cromossomos/genética , Humanos , Interferon gama/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Congênicos , Dados de Sequência Molecular , Análise de Sequência de DNA , Baço/metabolismo , Theilovirus/fisiologia , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA