Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Neuron ; 7(3): 365-79, 1991 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1654946

RESUMO

We have examined the pathological lesions and sites of infection in mice inoculated with a highly neurovirulent recombinant wild mouse ecotropic retrovirus (FrCasE). The spongiform lesions appeared initially as swollen postsynaptic neuronal processes, progressing to swelling in neuronal cell bodies, all in the absence of detectable gliosis. Infection of neurons in regions of vacuolation was not detected. However, high level infection of cerebellar granule neurons was observed in the absence of cytopathology, wherein viral protein was found associated with both axons and dendrites. Infection of ramified and amoeboid microglial cells was associated with cytopathology in the brain stem, and endothelial cell-pericyte infection was found throughout the CNS. No evidence of defective retroviral expression was observed. These results are consistent with an indirect mechanism of retrovirus-induced neuropathology.


Assuntos
Doenças do Sistema Nervoso Central/microbiologia , Infecções por Retroviridae/patologia , Retroviridae/patogenicidade , Animais , Vasos Sanguíneos/microbiologia , Doenças do Sistema Nervoso Central/patologia , Córtex Cerebelar/microbiologia , Córtex Cerebelar/patologia , Produtos do Gene env/metabolismo , Imuno-Histoquímica , Camundongos , Neurônios/microbiologia , Neurônios/patologia , Hibridização de Ácido Nucleico , Vírus de RNA/patogenicidade , Vírus Reordenados/patogenicidade , Infecções por Retroviridae/microbiologia , Replicação Viral
2.
J Virol ; 67(5): 2601-10, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8386268

RESUMO

In this report, we have examined the role of central nervous system (CNS) development in the pathogenesis of neurodegenerative disease induced by murine retroviruses. This was accomplished by comparing the effect of delivering viruses, with either severe or marginal neurovirulence (J. L. Portis, S. Czub, C. F. Garon, and F. J. McAtee, J. Virol. 64:1648-1656, 1990), during the midgestational development of the mouse (gestation days 9 to 10). Midgestation inoculation of the marginally neurovirulent virus, 15-1, resulted in high level CNS infection, as determined by viral DNA and protein analysis. The high-level infection resulted in rapid, severe disease with 100% incidence and an average clinical onset on postnatal day 17 (P17). The disease onset was comparable to that observed for the highly neurovirulent virus, FrCasE, when inoculated neonatally (onset ca. P16). To evaluate whether disease could be induced even earlier in CNS development, FrCasE was inoculated during midgestation. Surprisingly, neither clinical nor histological manifestations of CNS disease were accelerated but rather appeared at the same developmental time as seen for neonatally inoculated animals (onset of neuropathology at ca. P10; onset of clinical disease at ca. P15). CNS infection, on the other hand, occurred at earlier times (< P0), at higher levels, and with a broader distribution than in neonatally inoculated animals. No infection of the neurons which ultimately degenerate was observed in any regimen of virus inoculation. It was observed, however, that the gp70 viral envelope protein from the CNS showed an increase mobility on sodium dodecyl sulfate-polyacrylamide gel electrophoresis compared with the envelope protein from infected spleens or purified virions. These results indicate that a postnatal developmental event must occur to allow the presence of a neurovirulent virus to precipitate spongiform degeneration and that an altered envelope protein may be participating in the process.


Assuntos
Sistema Nervoso Central/microbiologia , Infecções por Retroviridae/patologia , Retroviridae/patogenicidade , Doenças dos Roedores/microbiologia , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Encéfalo/microbiologia , Encéfalo/patologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/patologia , Produtos do Gene env/isolamento & purificação , Imuno-Histoquímica , Camundongos/embriologia , Camundongos/microbiologia , Neurônios/patologia , Retroviridae/crescimento & desenvolvimento , Infecções por Retroviridae/etiologia , Doenças dos Roedores/etiologia , Doenças dos Roedores/patologia , Fatores de Tempo , Distribuição Tecidual , Virulência , Replicação Viral
3.
J Virol ; 74(3): 1558-65, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10627570

RESUMO

The wild mouse ecotropic retrovirus, Cas-Br-E, induces progressive, noninflammatory spongiform neurodegenerative disease in susceptible mice. Functional genetic analysis of the Cas-Br-E genome indicates that neurovirulence maps to the env gene, which encodes the surface glycoprotein responsible for binding and fusion of virus to host cells. To understand how the envelope protein might be involved in the induction of disease, we examined the regional and temporal expression of Cas-Br-E Env protein in the central nervous systems (CNS) of mice infected with the highly neurovirulent chimeric virus FrCas(E). We observed that multiple isoforms of Cas-Br-E Env were expressed in the CNS, with different brain regions exhibiting unique patterns of processed Env glycoprotein. Specifically, the expression of gp70 correlated with regions showing microglial infection and spongiform neurodegeneration. In contrast, regions high in neuronal infection and without neurodegenerative changes (the cerebellum and olfactory bulb) were characterized by a gp65 Env protein isoform. Sedimentation analysis of brain region extracts indicated that gp65 rather than gp70 was incorporated into virions. Biochemical analysis of the Cas-Br-E Env isoforms indicated that they result from differential processing of N-linked sugars. Taken together, these results indicate that differential posttranslational modification of the Cas-Br-E Env is associated with a failure to incorporate certain Env isoforms into virions in vivo, suggesting that defective viral assembly may be associated with the induction of spongiform neurodegeneration.


Assuntos
Encéfalo/patologia , Encéfalo/virologia , Gammaretrovirus/patogenicidade , Infecções por Retroviridae/virologia , Proteínas do Envelope Viral/metabolismo , Animais , Encéfalo/metabolismo , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Glicosilação , Camundongos , Isoformas de Proteínas , Infecções por Retroviridae/patologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
4.
Virology ; 271(2): 227-33, 2000 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-10860875

RESUMO

The discovery within the past decade that neural stem cells (NSCs) from the developing and adult mammalian brain can be propagated, cloned, and genetically manipulated ex vivo for ultimate transfer back into the CNS has opened the door to a novel means for modifying the CNS environment for experimental and therapeutic purposes. While a great deal of interest has been focused on the properties and promise of this new technology, especially in regard to cellular replacement and gene therapy, this minireview will focus on the recent use of NSCs to study the neuropathogenesis of the murine oncornaviruses. In brief, the use of this NSC-based approach has provided a means for selective reconstitution within the brain, of specific retroviral life cycle events, in order to consider their contribution to the induction of neurodegeneration. Furthermore, by virtue of their ability to disseminate virus within the brain, NSCs have provided a reliable means for assessing the true neurovirulence potential of murine oncornaviruses by directly circumventing a restriction to virus entry into the CNS. Importantly, these experiments have demonstrated that the neurovirulence of oncornaviruses requires late virus life cycle events occurring specifically within microglia, the resident macrophages of the brain. This initial application of NSC biology to the analysis of oncornavirus-CNS interactions may serve as an example for how other questions in viral neuropathogenesis might be addressed in the future.


Assuntos
Neurônios/virologia , Retroviridae/patogenicidade , Células-Tronco/virologia , Animais , Humanos , Fusão de Membrana/fisiologia , Microglia/virologia , Infecções por Retroviridae/virologia , Proteínas dos Retroviridae/metabolismo , Proteínas do Envelope Viral/metabolismo , Virulência
5.
J Virol ; 69(11): 7300-3, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474158

RESUMO

The neurovirulent chimeric mouse ecotropic retrovirus FrCasE causes a rapid neurodegenerative disease of the central nervous system (CNS) characterized by the appearance of spongiform lesions in motor areas 10 days after neonatal inoculation. To study the details of the pathogenic process, we examined the ability of an ex vivo spinal cord model to recapitulate disease. Organotypic spinal cord slice cultures were established from IRW mice 7 days after neonatal inoculation. This corresponds to a time when virus expression in the CNS is first detectable but spongiform changes have yet to evolve. Infectivity associated with these cultures peaked at 7 days in vitro and persisted at this level for 6 weeks. FrCasE infection of the spinal cord slices was primarily found associated with microglial cells. Infection of neurons, astrocytes, oligodendroglia, and endothelial cells was not observed; however, significant astrogliosis was found. Despite the presence of extensive microglial infection in close association with spinal motor neurons in organotypic cultures, no virus-specific spongiform degenerative changes were observed. These results suggest that removal of motor neurons from the developing CNS, despite maintaining the local cytoarchitectural relationships, prevents the virus from eliciting its pathological effects. Possible reasons for the interruption of lesion development are discussed.


Assuntos
Doenças do Sistema Nervoso Central/virologia , Retroviridae/patogenicidade , Medula Espinal/patologia , Medula Espinal/virologia , Animais , Anticorpos Monoclonais , Doenças do Sistema Nervoso Central/patologia , Quimera , Imunofluorescência , Proteína Glial Fibrilar Ácida/análise , Camundongos , Técnicas de Cultura de Órgãos , Retroviridae/isolamento & purificação , Retroviridae/fisiologia , Proteínas do Envelope Viral/análise
6.
J Virol ; 69(3): 1408-19, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7853473

RESUMO

FrCasE is a highly neurovirulent murine leukemia virus which causes a noninflammatory spongiform neurodegenerative disease after neonatal inoculation. The central nervous system (CNS) infection is wide-spread, involving several different cell types, whereas the lesions are localized to motor areas of the brain and spinal cord. Inoculation of FrCasE at 10 days of age (P10) results in viremia, but infection of the CNS is restricted and neurological disease is not observed (M. Czub, S. Czub, F. McAtee, and J. Portis, J. Virol. 65:2539-2544, 1991). In this study, we used this developmental resistance to restrict the extent and the distribution of FrCasE in the brain to examine whether the spongiform degeneration is a consequence of infection of cells in proximity to the lesions. Two approaches were used to infect the brain on or after P10. First, mice were inoculated with FrCasE at P10 to induce viremia and then at P17 were subjected to focal CNS injury within brain regions known to be susceptible to virus-induced spongiform degeneration. The injury resulted in local inflammation, glial activation, migration of inflammatory cells into the wound site, and high-level parenchymal infection about the wound site. However, no evidence of spongiform neurodegeneration was observed over a period of 3 months. The second approach involved the implantation of FrCasE-infected microglia into the CNS at > or = P10. This resulted in microglial engraftment and focal CNS infection unilaterally at the implantation sites and bilaterally along white matter tracts of the corpus callosum and pons and in cells of the subventricular layers of the lateral cerebral ventricles. Strikingly, focal spongiform degeneration colocalized with the sites of infection. In contrast to the wounding experiments, the implantation model was not associated with an inflammatory response or significant glial activation. Results of these studies suggest that (i) the developmental resistance of the CNS to infection lies at the blood-brain barrier and can be bypassed by direct introduction into the brain of virus-infected cells, (ii) the neuropathology induced by this virus is a consequence of local effects of the infection and does not appear to require endothelial or neuronal infection, and (iii) elements of the inflammatory response and/or glial activation may modulate the expression of neuropathology induced by neurovirulent retroviruses.


Assuntos
Vírus da Leucemia Murina/patogenicidade , Microglia/microbiologia , Degeneração Neural , Doenças do Sistema Nervoso/microbiologia , Fatores Etários , Animais , Barreira Hematoencefálica , Camundongos , Camundongos Endogâmicos , Microglia/transplante , Doenças do Sistema Nervoso/patologia
7.
J Virol ; 73(8): 6841-51, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400782

RESUMO

The induction of spongiform myeloencephalopathy by murine leukemia viruses is mediated primarily by infection of central nervous system (CNS) microglia. In this regard, we have previously shown that CasBrE-induced disease requires late, rather than early, virus replication events in microglial cells (W. P. Lynch et al., J. Virol. 70:8896-8907, 1996). Furthermore, neurodegeneration requires the presence of unique sequences within the viral env gene. Thus, the neurodegeneration-inducing events could result from microglial expression of retroviral envelope protein alone or from the interaction of envelope protein with other viral structural proteins in the virus assembly and maturation process. To distinguish between these possible mechanisms of disease induction, we engineered the engraftable neural stem cell line C17-2 into packaging/producer cells in order to deliver the neurovirulent CasBrE env gene to endogenous CNS cells. This strategy resulted in significant CasBrE env expression within CNS microglia without the appearance of replication competent virus. CasBrE envelope expression within microglia was accompanied by increased expression of activation markers F4/80 and Mac-1 (CD11b) but failed to induce spongiform neurodegenerative changes. These results suggest that envelope expression alone within microglia is not sufficient to induce neurodegeneration. Rather, microglia-mediated disease appears to require neurovirulent Env protein interaction with other viral proteins during assembly or maturation. More broadly, the results presented here prove the efficacy of a novel method by which neural stem cell biology may be harnessed for genetically manipulating the CNS, not only for studying neurodegeneration but also as a paradigm for the disseminated distribution of retroviral vector-transduced genes.


Assuntos
Técnicas de Transferência de Genes , Genes env , Microglia , Infecções por Retroviridae/virologia , Retroviridae/fisiologia , Células-Tronco , Animais , Linhagem Celular , Transplante de Células-Tronco Hematopoéticas , Camundongos , Microglia/citologia , Degeneração Neural , Retroviridae/genética , Infecções por Retroviridae/patologia , Células-Tronco/citologia
8.
J Biol Chem ; 262(15): 7429-37, 1987 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-3584120

RESUMO

Caldesmon is a major F-actin binding protein of smooth muscle that has been implicated as a component of a thin filament regulatory system. Chicken gizzard caldesmon consists of polypeptides of Mr-135,000 and 140,000 which are closely related as determined by analysis of cyanogen bromide cleavage fragments. It is a highly extended flexible protein having a contour length of about 146 nm and a secondary structure composed primarily of random coil. Physical and chemical cross-linking data suggest that caldesmon exists as a monomer in solution. The cysteine content of caldesmon was determined to be 2 residues/polypeptide. Remarkably, in solution it readily undergoes sulfhydryl oxidation to form either an internal disulfide bridge in the protein or cross-links between individual polypeptides to form dimers, trimers, tetramers, etc. The internally cross-linked species have a smaller Stokes radius than the reduced molecules, indicating that the cross-link "trapped" the molecule in a compact conformation. Oxidized protein containing caldesmon oligomers is a potent F-actin bundling protein. Complete reduction of caldesmon abolishes the F-actin bundling activity. Since a vast excess of reducing agent is required to convert caldesmon from an oxidized to reduced state, it may exist in either state in vivo. Thus, the ability of caldesmon to undergo reversible sulfhydryl cross-linking, and thereby reversible F-actin cross-linking, may be of physiological significance.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Músculo Liso/análise , Compostos de Sulfidrila/metabolismo , Animais , Galinhas , Reagentes de Ligações Cruzadas , Brometo de Cianogênio , Cisteína/análise , Dissulfetos/metabolismo , Moela das Aves/análise , Peso Molecular , Oxirredução , Fragmentos de Peptídeos , Conformação Proteica , Coelhos , Soluções
9.
J Virol ; 68(5): 3401-9, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8151801

RESUMO

The observation of murine retrovirus infection of microglial cells in brain regions expressing spongiform neurodegenerative changes suggests that these cells may play an important role in pathogenesis. To evaluate this potential in vitro, murine microglial cells were infected in mixed glial cultures with the highly neurovirulent murine retrovirus, FrCasE. The microglia were then isolated from the mixed cultures on the basis of their differential adherence and shown to be approximately 98% pure. The infected microglia expressed viral envelope protein at the plasma membrane, while viral budding was primarily intracellular. Evaluation of the viral envelope protein by immunoblotting indicated that the immunoreactive species produced was exclusively a 90-kDa precursor protein. Very little of the envelope protein was associated with particles released from these cells, and viral titers in the culture supernatant were low. Interestingly, these cells were still capable of infecting permissive target cells when seeded as infectious centers. This partially defective infection of microglial cells suggests a potential cellular means by which a neurovirulent retrovirus could disrupt normal microglia and in turn central nervous system motor system functioning.


Assuntos
Gammaretrovirus/crescimento & desenvolvimento , Microglia/microbiologia , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/metabolismo , Animais , Gammaretrovirus/patogenicidade , Gammaretrovirus/ultraestrutura , Camundongos , Microglia/patologia , Microglia/ultraestrutura , Virulência
10.
Lab Invest ; 70(5): 711-23, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8196367

RESUMO

BACKGROUND: A chimeric murine retrovirus, FrCasE, causes a rapid noninflammatory spongiform neurodegenerative disease of the motor system with an incubation period of 15 to 16 days after neonatal inoculation. Neurovirulence is determined by the viral envelope gene, but the neurodegeneration is an indirect consequence of virus infection, because the neurons that degenerate appear not to be infected. EXPERIMENTAL DESIGN: The current study was undertaken to compare the kinetics of lesion development and the expression of viral envelope protein in the central nervous system (CNS). Neonatal mice were inoculated with FrCasE intraperitoneally and were killed at various times for determination of the kinetics of the CNS infection, the distribution of lesion in the CNS, and the distribution of viral envelope protein. In addition, qualitative features of both viral envelope and gag proteins were followed by immunoblot analysis. RESULTS: The lesions induced by FrCasE consisted of vacuolar degeneration but without associated astrocytosis, the lack of an astroglial response being a consequence of the rapidity of the disease process. Vacuoles were observed primarily in the neuropil of the motor centers of spinal cord, brain stem, and cerebral cortex. Lesions appeared in all of these areas during a narrow window of time (< or = 3 days). Cells in which viral envelope protein was detected by immunohistochemistry before the appearance of spongiform degeneration included premigratory cerebellar cortical granule neurons as well as vascular elements in the regions that would ultimately exhibit spongiform degeneration. Two forms of viral envelope protein were detected in the CNS. A 70-kilodalton species appeared first, followed by an approximately 64-kilodalton species, which was detected coincident with the first appearance of spongiform lesions. CONCLUSIONS: Astrocytosis is a secondary reaction to the neuronal cytopathology induced by FrCasE and appears to be dependent on the developmental state of the CNS. The abrupt, diffuse nature of lesion development in this disease suggests a global effect of the virus infection. Cells of the CNS vasculature (either endothelial cells, perivascular microglial cells, or both) as well as cerebellar granule neurons appear to be seminally involved in the pathogenesis of the spongiform degeneration. The two species of viral envelope protein appear to be expressed by different cell types in the CNS.


Assuntos
Astrócitos/patologia , Encéfalo/patologia , Doenças Priônicas/patologia , Retroviridae/patogenicidade , Medula Espinal/patologia , Animais , Anticorpos Monoclonais , Antígenos Virais/análise , Encéfalo/microbiologia , Tronco Encefálico/patologia , Cerebelo/patologia , Córtex Cerebral/patologia , Quimera , DNA Viral/análise , Suscetibilidade a Doenças , Proteína Glial Fibrilar Ácida/análise , Immunoblotting , Imuno-Histoquímica , Cinética , Camundongos , Camundongos Endogâmicos , Doenças Priônicas/microbiologia , Retroviridae/genética , Fatores de Tempo , Vacúolos/patologia , Proteínas do Envelope Viral/análise , Proteínas Virais/análise
11.
Arch Virol ; 142(5): 1011-9, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9191865

RESUMO

Recent studies have reported the detection of rabies viral antigens and virions in astrocytes and microglia of rabies-infected animals. As a first step toward understanding whether these glial cells may be involved in rabies virus replication, persistence, and/or pathogenesis, we explored their potential to be infected in vitro. Primary cultures of murine, feline, and human microglia and astrocytes were infected with several different rabies viruses: two unpassaged street virus isolates, a cell culture-adapted strain, and a mouse brain-passaged strain. Infection, as determined by immunofluorescence, was detected in 15 of the 16 (94%) virus-glial cell combinations. Replication of infectious virus, determined by infectivity assay, was detected in 7 of the 8 (88%) virus-cell combinations. These results show that astrocytes and microglia can be infected by rabies viruses, suggesting that they may have a potential role in disease, perhaps contributing to viral spread, persistence and/or neuronal dysfunction.


Assuntos
Astrócitos/virologia , Microglia/virologia , Vírus da Raiva/crescimento & desenvolvimento , Animais , Gatos , Células Cultivadas , Humanos , Camundongos , Replicação Viral
12.
Virology ; 206(1): 372-80, 1995 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-7831792

RESUMO

Perinatal infection of susceptible mice with the neurotropic retrovirus CasBrE leads to a noninflammatory spongiform degeneration of the central nervous system with a long incubation period of up to 1 year. Virus replication in infected animals can be suppressed by administration of antiviral antibodies, cytotoxic T cells, or by AZT treatment, which results in partial to complete protection from neurological disease. A highly neuropathogenic chimeric retrovirus, FrCasE, which contains the envelope gene of CasBrE, induces rapid neurodegeneration within only 16 days. Here we report that this fatal disease could be prevented if a nonneuropathogenic Friend murine leukemia virus was administered to mice prior to their infection with FrCasE. This double inoculation led to a substantial reduction of the replication level of FrCasE in spleen and CNS. Only live but not heat-inactivated nonneuropathogenic virus was able to protect from FrCasE-induced neurological disease. The extent of protection was influenced by the viral envelope gene and the kinetics of replication of the nonneuropathogenic virus. These observations in addition to the rapidity of the effect make it likely that competition for replication sites through the mechanism of viral interference is responsible for the protection. Resistance was demonstrable in vivo even when the "protecting" and "challenge" virus belonged to different in vitro interference groups. However, the protection was considerably weaker than that seen between viruses belonging to the same interference group.


Assuntos
Doenças do Sistema Nervoso Central/prevenção & controle , Vírus da Leucemia Murina de Friend/fisiologia , Infecções por Retroviridae/prevenção & controle , Retroviridae/fisiologia , Animais , Encéfalo/virologia , Doenças do Sistema Nervoso Central/virologia , Cinética , Camundongos , Infecções por Retroviridae/fisiopatologia , Baço/virologia , Interferência Viral , Replicação Viral
13.
J Virol ; 70(12): 8896-907, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8971019

RESUMO

CasBrE is a neurovirulent murine retrovirus which induces a spongiform myeloencephalopathy in susceptible mice. Genetic mapping studies have indicated that sequences responsible for neurovirulence reside within the env gene. To address the question of direct envelope protein neuroxicity in the central nervous system (CNS), we have generated chimeric mice expressing the CasBrE envelope protein in cells of neuroectodermal origin. Specifically, the multipotent neural progenitor cell line C17.2 was engineered to express the CasBrE env gene as either gp70/p15E (CasE) or gp70 alone (CasES). CasE expression in these cells resulted in complete (>10(5)) interference of superinfection with Friend murine leukemia virus clone FB29, whereas CasES expression resulted in a 1.8-log-unit decrease in FB29 titer. Introduction of these envelope-expressing C17.2 cells into the brains of highly susceptible IRW mice resulted in significant engraftment as integral cytoarchitecturally correct components of the CNS. Despite high-level envelope protein expression from the engrafted cells, no evidence of spongiform neurodegeneration was observed. To examine whether early virus replication events were necessary for pathogenesis, C17.2 cells expressing whole virus were transplanted into mice in which virus replication in the host was specifically restricted by Fv-1 to preintegration events. Again, significant C17.2 cell engraftment and infectious virus expression failed to precipitate spongiform lesions. In contrast, transplantation of virus-expressing C17.2 progenitor cells in the absence of the Fv-1 restriction resulted in extensive spongiform neurodegeneration by 2 weeks postengraftment. Cytological examination indicated that infection had spread beyond the engrafted cells, and in particular to host microglia. Spongiform neuropathology in these animals was directly correlated with CasBrE env expression in microglia rather than expression from neural progenitor cells. These results suggest that the envelope protein of CasBrE is not itself neurotoxic but that virus infectious events beyond binding and fusion in microglia are necessary for the induction of CNS disease.


Assuntos
Encéfalo/metabolismo , Produtos do Gene gag/metabolismo , Microglia/metabolismo , Doenças Priônicas/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Retroviridae/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Sequência de Bases , Encéfalo/virologia , Linhagem Celular , Sistema Nervoso Central/virologia , DNA Viral , Expressão Gênica , Produtos do Gene gag/genética , Camundongos , Dados de Sequência Molecular , Doenças Priônicas/virologia , Retroviridae/genética , Retroviridae/patogenicidade , Retroviridae/fisiologia , Proteínas Oncogênicas de Retroviridae/genética , Proteínas do Envelope Viral/genética , Virulência , Replicação Viral
14.
J Biol Chem ; 264(5): 2869-75, 1989 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-2914935

RESUMO

Caldesmons are major Ca2+-calmodulin regulated F-actin binding proteins of smooth and non-muscle cells that have been implicated as components of a thin filament regulatory system. Chicken gizzard caldesmons are monomeric proteins of Mr 140,000 and 135,000. We have employed enzymatic and chemical cleavage methods in order to dissect the protein to locate the Ca2+-calmodulin and F-actin binding domain and the site of molecular weight heterogeneity. Using a novel mapping procedure that employs partial chemical cleavage at cysteine residues, we show that both caldesmon polypeptides contain 2 cysteine residues located approximately 28,000 from the protein's amino terminus and the second approximately 25,000 from the carboxyl terminus. Identification of the composition of partial cleavage products with region-specific antibodies is consistent with this derived map. The apparent molecular weight heterogeneity was found to lie in the approximately 80,000 region between the 2 cysteine residues and therefore is not due to proteolytic processing. Digestion with alpha-chymotrypsin yields a relatively stable basic Mr 40,000 Ca2+-calmodulin and F-actin binding fragment that we have purified and characterized. The chymotryptic 40,000 fragment contains the 25,000 carboxyl-terminal fragment and therefore is derived from the carboxyl-terminal region of caldesmon. The 25,000 fragment obtained after chemical cleavage at cysteine under native conditions has also been purified and shown to bind F-actin and Ca2+-calmodulin. Surprisingly, the purified carboxyl 25,000 fragment, unlike the reduced intact monomer, cross-links F-actin into tightly ordered bundles in which the filaments are in register.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , Músculo Liso/metabolismo , Aminoácidos/análise , Animais , Anticorpos , Galinhas , Eletroforese em Gel de Poliacrilamida , Moela das Aves/metabolismo , Peso Molecular , Fragmentos de Peptídeos/análise , Mapeamento de Peptídeos
15.
Immunity ; 3(5): 541-7, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7584144

RESUMO

The B7-CD28/CTLA-4 costimulatory pathway can provide a signal pivotal for T cell activation. Signaling through this pathway is complex due to the presence of two B7 family members, B7-1 and B7-2, and two counterreceptors, CD28 and CTLA-4. Studies with anti-CTLA-4 monoclonal antibodies have suggested both positive and negative roles for CTLA-4 in T cell activation. To elucidate the in vivo function of CTLA-4, we generated CTLA-4-deficient mice. These mice rapidly develop lymphoproliferative disease with multiorgan lymphocytic infiltration and tissue destruction, with particularly severe myocarditis and pancreatitis, and die by 3-4 weeks of age. The phenotype of the CTLA-4-deficient mouse strain is supported by studies that have suggested a negative role for CTLA-4 in T cell activation. The severe phenotype of mice lacking CTLA-4 implies a critical role for CTLA-4 in down-regulating T cell activation and maintaining immunologic homeostasis. In the absence of CTLA-4, peripheral T cells are activated, can spontaneously proliferate, and may mediate lethal tissue injury.


Assuntos
Antígenos de Diferenciação/fisiologia , Imunoconjugados , Tecido Linfoide/patologia , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/patologia , Vísceras/patologia , Abatacepte , Animais , Antígenos CD , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Sequência de Bases , Antígeno CTLA-4 , Células Cultivadas , Citocinas/biossíntese , Primers do DNA/química , Regulação para Baixo/genética , Imuno-Histoquímica , Ativação Linfocitária/genética , Transtornos Linfoproliferativos/mortalidade , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Dados de Sequência Molecular , Linfócitos T/imunologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA