Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Adv Mater ; 35(1): e2202670, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36208089

RESUMO

Herein, a tolerogenic nanovaccine is developed and tested on an animal model of multiple sclerosis. The nanovaccine is constructed to deliver the self-antigen, myelin oligodendrocyte glycoprotein (MOG) peptide, and dexamethasone on an abatacept-modified polydopamine core nanoparticle (AbaLDPN-MOG). AbaLDPN-MOG can target dendritic cells and undergo endocytosis followed by trafficking to lysosomes. AbaLDPN-MOG blocks the interaction between CD80/CD86 and CD28 in antigen-presenting cells and T cells, leading to decreased interferon gamma secretion. The subcutaneous administration of AbaLDPN-MOG to mice yields significant biodistribution to lymph nodes and, in experimental-autoimmune encephalomyelitis (EAE) model mice, increases the integrity of the myelin basic sheath and minimizes the infiltration of immune cells. EAE mice are treated with AbaLDPN-MOG before or after injection of the autoantigen, MOG. Preimmunization of AbaLDPN-MOG before the injection of MOG completely blocks the development of clinical symptoms. Early treatment with AbaLDPN-MOG at three days after injection of MOG also completely blocks the development of symptoms. Notably, treatment of EAE symptom-developed mice with AbaLDPN-MOG significantly alleviates the symptoms, indicating that the nanovaccine has therapeutic effects. Although AbaLDPN is used for MOG peptide delivery in the EAE model, the concept of AbaLDPN can be widely applied for the prevention and alleviation of other autoimmune diseases.


Assuntos
Encefalomielite Autoimune Experimental , Encefalomielite , Glicoproteína Mielina-Oligodendrócito , Animais , Camundongos , Encefalomielite/imunologia , Encefalomielite/prevenção & controle , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , Glicoproteína Mielina-Oligodendrócito/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Distribuição Tecidual , Vacinas , Nanopartículas/uso terapêutico , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia
2.
J Virol ; 85(14): 6822-31, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21593179

RESUMO

The absence of interleukin-10 (IL-10), a potent anti-inflammatory cytokine results in increased immune-mediated demyelination in mice infected with a neurotropic coronavirus (recombinant J2.2-V-1 [rJ2.2]). Here, we examined the therapeutic effects of increased levels of IL-10 at early times after infection by engineering a recombinant J2.2 virus to produce IL-10. We demonstrate that viral expression of IL-10, which occurs during the peak of virus replication and at the site of disease, enhanced survival and diminished morbidity in rJ2.2-infected wild-type B6 and IL-10(-/-) mice. The protective effects of increased IL-10 levels were associated with reductions in microglial activation, inflammatory cell infiltration into the brain, and proinflammatory cytokine and chemokine production. Additionally, IL-10 increased both the frequency and number of Foxp3(+) regulatory CD4 T cells in the infected central nervous system. Most strikingly, the ameliorating effects of IL-10 produced during the first 5 days after infection were long acting, resulting in decreased demyelination during the resolution phase of the infection. Collectively, these results suggest that the pathogenic processes that result in demyelination are initiated early during infection and that they can be diminished by exogenous IL-10 delivered soon after disease onset. IL-10 functions by dampening the innate or very early T cell immune response. Further, they suggest that early treatment with IL-10 may be useful adjunct therapy in some types of viral encephalitis.


Assuntos
Coronavirus/genética , Doenças Desmielinizantes/prevenção & controle , Encefalomielite/prevenção & controle , Interleucina-10/fisiologia , Doença Aguda , Animais , Sequência de Bases , Doença Crônica , Primers do DNA , Doenças Desmielinizantes/imunologia , Encefalomielite/imunologia , Citometria de Fluxo , Interleucina-10/genética , Camundongos , Proteínas Recombinantes/metabolismo , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
J Virol ; 84(1): 661-5, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864378

RESUMO

Enterovirus 71 (EV71) causes childhood hand, foot, and mouth disease and neurological complications, and no vaccines or therapeutic drugs are currently available. Formaldehyde-inactivated whole-virus vaccines derived from EV71 clinical isolates and a mouse-adapted virus (MAV) were tested in a mouse model of EV71 encephalomyelitis. After only two immunizations, given to mice at 1 and 7 days of age, the MAV vaccine protected mice at 14 days of age from disease. Tissues from immunized mice were negative for virus by viral culture, reverse transcriptase PCR, immunohistochemistry analysis, and in situ hybridization. Cross-neutralizing EV71 antibodies to strains with genotypes B3, B4, and C1 to C5 generated in immunized adult mice were able to passively protect 14-day-old mice from disease.


Assuntos
Encefalomielite/prevenção & controle , Infecções por Enterovirus/prevenção & controle , Enterovirus , Vacinas de Produtos Inativados/farmacologia , Animais , Anticorpos Neutralizantes/imunologia , Proteção Cruzada , Modelos Animais de Doenças , Formaldeído/farmacologia , Genótipo , Humanos , Imunização , Camundongos , Vacinas de Produtos Inativados/uso terapêutico
4.
J Virol ; 84(6): 3111-5, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20042510

RESUMO

Neurotropic coronavirus induces acute encephalomyelitis and demyelination in mice. Infection of BALB/c (H-2(d)) mice expressing a dominant negative gamma interferon (IFN-gamma) receptor specifically in oligodendrocytes was examined to determine the influence of IFN-gamma signaling on pathogenesis. Inhibition of IFN-gamma signaling in oligodendrocytes increased viral load, infection of oligodendrocytes, oligodendrocyte loss, demyelination, and axonal damage resulting in increased mortality. IFN-gamma levels and the inflammatory response were not altered, although the level of tumor necrosis factor (TNF) mRNA was increased. These data indicate that IFN-gamma signaling by oligodendroglia reduces viral replication but affects both demyelination and tissue destruction in a host-specific manner.


Assuntos
Infecções por Coronavirus/prevenção & controle , Encefalomielite/prevenção & controle , Interferon gama/imunologia , Oligodendroglia/metabolismo , Oligodendroglia/virologia , Transdução de Sinais/imunologia , Animais , Infecções por Coronavirus/imunologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/prevenção & controle , Doenças Desmielinizantes/virologia , Encefalomielite/imunologia , Encefalomielite/patologia , Encefalomielite/virologia , Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/patogenicidade , Oligodendroglia/patologia , Receptores de Interferon/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Carga Viral , Replicação Viral/imunologia
5.
Proc Natl Acad Sci U S A ; 105(9): 3575-80, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18296635

RESUMO

Neuronal cell death during fatal acute viral encephalomyelitis can result from damage caused by virus replication, glutamate excitotoxicity, and the immune response. A neurovirulent strain of the alphavirus Sindbis virus (NSV) causes fatal encephalomyelitis associated with motor neuron death in adult C57BL/6 mice that can be prevented by treatment with the prototypic noncompetitive alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) glutamate receptor antagonist GYKI 52466 [Nargi-Aizenman J, et al. (2004) Ann Neurol 55:541-549]. To determine the mechanism of protection, NSV-infected mice were treated with 7-acetyl-5-(4-aminophenyl)-8(R)-methyl-8,9-dihydro-7H-1,3-dioxolo-(4,5-h)-benzodiazepine (talampanel), a potent, orally available member of the 2,3 benzodiazepine class of noncompetitive AMPA glutamate receptor antagonists. Talampanel-treated mice were protected from NSV-induced paralysis and death. Examination of the brain during infection showed significantly less mononuclear cell infiltration and no increase in astrocyte expression of glial fibrillary acidic protein in treated mice compared with untreated mice. Lack of CNS inflammation was attributable to failure of treated mice to induce activation and proliferation of lymphocytes in secondary lymphoid tissue in response to infection. Antibody responses to NSV were also suppressed by talampanel treatment, and virus clearance was delayed. These studies reveal a previously unrecognized effect of AMPA receptor antagonists on the immune response and suggest that prevention of immune-mediated damage, in addition to inhibition of excitotoxicity, is a mechanism by which these drugs protect from death of motor neurons caused by viral infection.


Assuntos
Benzodiazepinas/farmacologia , Encefalomielite/prevenção & controle , Inflamação/tratamento farmacológico , Receptores de AMPA/antagonistas & inibidores , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/patologia , Animais , Benzodiazepinas/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/virologia , Encefalomielite/tratamento farmacológico , Encefalomielite/patologia , Encefalomielite/virologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Sindbis virus , Viroses/tratamento farmacológico , Viroses/patologia
6.
J Virol ; 82(7): 3369-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18234804

RESUMO

We evaluated the participatory role of human HLA-DR molecules in control of virus from the central nervous system and in the development of subsequent spinal cord demyelination. The experiments utilized intracranial infection with Theiler's murine encephalomyelitis virus (TMEV), a picornavirus that, in some strains of mice, results in primary demyelination. We studied DR2 and DR3 transgenic mice that were bred onto a combined class I-deficient mouse (beta-2 microglobulin deficient; beta2m(0)) and class II-deficient mouse (Abeta(0)) of the H-2(b) background. Abeta(0).beta2m(0) mice infected with TMEV died within 18 days of infection. These mice showed severe encephalomyelitis due to rapid replication of virus genome. In contrast, transgenic mice with insertion of a single human class II major histocompatibility complex (MHC) gene (DR2 or DR3) survived the acute infection. DR2 and DR3 mice controlled virus infection by 45 days and did not develop spinal cord demyelination. Levels of virus RNA were reduced in HLA-DR transgenic mice compared to Abeta(0).beta2m(0) mice. Virus-neutralizing antibody responses did not explain why DR mice survived the infection and controlled virus replication. However, DR mice showed an increase in gamma interferon and interleukin-2 transcripts in the brain, which were associated with protection. The findings support the hypothesis that the expression of a single human class II MHC molecule can, by itself, influence the control of an intracerebral pathogen in a host without a competent class I MHC immune response. The mechanism of protection appears to be the result of cytokines released by CD4(+) T cells.


Assuntos
Doenças Desmielinizantes/prevenção & controle , Encefalomielite/prevenção & controle , Antígenos HLA-DR/imunologia , Theilovirus/imunologia , Animais , Anticorpos Antivirais/sangue , Encéfalo/imunologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/imunologia , Doenças Desmielinizantes/patologia , Encefalomielite/patologia , Expressão Gênica , Humanos , Interferon gama/biossíntese , Interleucina-2/biossíntese , Camundongos , Camundongos Transgênicos , Testes de Neutralização , Análise de Sobrevida
7.
J Virol ; 81(24): 13291-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913802

RESUMO

Mutation within virus-derived CD8 T-cell epitopes can effectively abrogate cytotoxic T-lymphocyte (CTL) recognition and impede virus clearance in infected hosts. These so-called "CTL escape variant viruses" are commonly selected during persistent infections and are associated with rapid disease progression and increased disease severity. Herein, we tested whether antiviral antibody-mediated suppression of virus replication and subsequent virus clearance were necessary for preventing CTL escape in coronavirus-infected mice. We found that compared to wild-type mice, B-cell-deficient mice did not efficiently clear infectious virus, uniformly developed clinical disease, and harbored CTL escape variant viruses. These data directly demonstrate a critical role for antiviral antibody in protecting from the selective outgrowth of CTL escape variant viruses.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Coronavirus/mortalidade , Encefalomielite/imunologia , Vírus da Hepatite Murina/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Animais Lactentes , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Encefalomielite/mortalidade , Encefalomielite/prevenção & controle , Epitopos de Linfócito T/genética , Feminino , Humanos , Epitopos Imunodominantes/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Mutação
8.
Am J Vet Res ; 69(3): 396-402, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18312139

RESUMO

OBJECTIVE: To evaluate the effect of intermittent oral administration of ponazuril on immunoconversion against Sarcocystis neurona in horses inoculated intragastrically with S neurona sporocysts. ANIMALS: 20 healthy horses that were seronegative for S neurona-specific IgG. PROCEDURES: 5 control horses were neither inoculated with sporocysts nor treated. Other horses (5 horses/group) each received 612,500 S neurona sporocysts via nasogastric tube (day 0) and were not treated or were administered ponazuril (20 mg/kg, PO) every 7 days (beginning on day 5) or every 14 days (beginning on day 12) for 12 weeks. Blood and CSF samples were collected on day - 1 and then every 14 days after challenge for western blot assessment of immunoconversion. Clinical signs of equine protozoal myeloencephalitis (EPM) were monitored, and tissues were examined histologically after euthanasia. RESULTS: Sera from all challenged horses yielded positive western blot results within 56 days. Immunoconversion in CSF was detected in only 2 of 5 horses that were treated weekly; all other challenged horses immunoconverted within 84 days. Weekly administration of ponazuril significantly reduced the antibody response against the S neurona 17-kd antigen in CSF. Neurologic signs consistent with EPM did not develop in any group; likewise, histologic examination of CNS tissue did not reveal protozoa or consistent degenerative or inflammatory changes. CONCLUSIONS AND CLINICAL RELEVANCE: Administration of ponazuril every 7 days, but not every 14 days, significantly decreased intrathecal anti-S neurona antibody responses in horses inoculated with S neurona sporocysts. Protocols involving intermittent administration of ponazuril may have application in prevention of EPM.


Assuntos
Coccidiostáticos/administração & dosagem , Encefalomielite/veterinária , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/parasitologia , Sarcocystis/imunologia , Sarcocistose/veterinária , Triazinas/administração & dosagem , Administração Oral , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/líquido cefalorraquidiano , Western Blotting/veterinária , Encéfalo/parasitologia , Encefalomielite/imunologia , Encefalomielite/parasitologia , Encefalomielite/prevenção & controle , Feminino , Histocitoquímica/veterinária , Doenças dos Cavalos/líquido cefalorraquidiano , Doenças dos Cavalos/imunologia , Cavalos , Masculino , Sarcocistose/imunologia , Sarcocistose/parasitologia , Sarcocistose/prevenção & controle
9.
Vet Parasitol ; 247: 37-41, 2017 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-29080762

RESUMO

Equine protozoal myeloencephalitis (EPM) is an important equine neurologic disorder, and treatments for the disease are often unrewarding. Prevention of the disease is the most important aspect for EPM, and a killed vaccine was previously developed for just that purpose. Evaluation of the vaccine had been hampered by lack of post vaccination challenge. The purpose of this study was to determine if the vaccine could prevent development of clinical signs after challenge with Sarcocystis neurona sporocysts in an equine challenge model. Seventy horses that were negative for antibodies to S. neurona and were neurologically normal were randomly assigned to vaccine or placebo groups and divided into short-term duration of immunity (study #1) and long-term duration of immunity (study #2) studies. S. neurona sporocysts used for the challenge were generated in the opossum/raccoon cycle isolate SN 37-R. Study #1 horses received an initial vaccination and a booster, and were challenged 34days post second vaccination. Study #2 horses received a vaccination and two boosters and were challenged 139days post third vaccination. All horses in study #1 developed neurologic signs (n=30) and there was no difference between the vaccinates and controls (P=0.7683). All but four horses in study #2 developed detectable neurologic deficits. The neurologic signs, although not statistically significant, were worse in the vaccinated horses (P=0.1559). In these two studies, vaccination with the S. neurona vaccine failed to prevent development of clinical neurologic deficits.


Assuntos
Encefalomielite/veterinária , Doenças dos Cavalos/prevenção & controle , Vacinas Protozoárias/imunologia , Sarcocystis/imunologia , Sarcocistose/veterinária , Vacinação/veterinária , Animais , Encefalomielite/parasitologia , Encefalomielite/prevenção & controle , Doenças dos Cavalos/parasitologia , Cavalos , Gambás , Guaxinins , Distribuição Aleatória , Sarcocistose/parasitologia , Sarcocistose/prevenção & controle
10.
J Parasitol ; 92(3): 637-43, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16884012

RESUMO

The ability of ponazuril to prevent or limit clinical signs of equine protozoal myeloencephalitis (EPM) after infection with Sarcocystis neurona was evaluated. Eighteen horses were assigned to 1 of 3 groups: no treatment, 2.5 mg/kg ponazuril, or 5.0 mg/kg ponazuril. Horses were administered ponazuril, once per day, beginning 7 days before infection (study day 0) and continuing for 28 days postinfection. On day 0, horses were stressed by transport and challenged with 1 million S. neurona sporocysts per horse. Sequential neurologic examinations were performed, and serum and cerebrospinal fluid were collected and assayed for antibodies to S. neurona. All horses in the control group developed neurologic signs, whereas only 71 and 40% of horses in the 2.5 and 5.0 mg/kg ponazuril groups, respectively, developed neurologic abnormalities. This was significant at P = 0.034 by using Fisher exact test. In addition, seroconversion was decreased in the 5.0 mg/kg group compared with the control horses (100 vs. 40%; P = 0.028). Horses with neurologic signs were killed, and a post-mortem examination was performed. Mild-to-moderate, multifocal signs of neuroinflammation were observed. These results confirm that treatment with ponazuril at 5.0 mg/kg minimizes, but does not eliminate, infection and clinical signs of EPM in horses.


Assuntos
Antiprotozoários/uso terapêutico , Encefalomielite/veterinária , Doenças dos Cavalos/prevenção & controle , Sarcocistose/veterinária , Triazinas/uso terapêutico , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/líquido cefalorraquidiano , Western Blotting/veterinária , Contagem de Células/veterinária , Sistema Nervoso Central/patologia , Líquido Cefalorraquidiano/citologia , Encefalomielite/parasitologia , Encefalomielite/prevenção & controle , Contagem de Eritrócitos/veterinária , Feminino , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/parasitologia , Cavalos , Masculino , Exame Neurológico/veterinária , Distribuição Aleatória , Sarcocystis/imunologia , Sarcocistose/imunologia , Sarcocistose/prevenção & controle , Índice de Gravidade de Doença
11.
Neurotherapeutics ; 13(3): 455-60, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27114366

RESUMO

Mosquito-borne viruses are important causes of death and long-term neurologic disability due to encephalomyelitis. Studies of mice infected with the alphavirus Sindbis virus have shown that outcome is dependent on the age and genetic background of the mouse and virulence of the infecting virus. Age-dependent susceptibility reflects the acquisition by neurons of resistance to virus replication and virus-induced cell death with maturation. In mature mice, the populations of neurons most susceptible to infection are in the hippocampus and anterior horn of the spinal cord. Hippocampal infection leads to long-term memory deficits in mice that survive, while motor neuron infection can lead to paralysis and death. Neuronal death is immune-mediated, rather than a direct consequence of virus infection, and associated with entry and differentiation of pathogenic T helper 17 cells in the nervous system. To modulate glutamate excitotoxicity, mice were treated with an N-methyl-D-aspartate receptor antagonist, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor antagonists or a glutamine antagonist. The N-methyl-D-aspartate receptor antagonist MK-801 protected hippocampal neurons but not motor neurons, and mice still became paralyzed and died. α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor antagonists GYKI-52466 and talampanel protected both hippocampal and motor neurons and prevented paralysis and death. Glutamine antagonist 6-diazo-5-l-norleucine protected hippocampal neurons and improved memory generation in mice surviving infection with an avirulent virus. Surprisingly, in all cases protection was associated with inhibition of the antiviral immune response, reduced entry of inflammatory cells into the central nervous system, and delayed virus clearance, emphasizing the importance of treatment approaches that include prevention of immunopathologic damage.


Assuntos
Infecções por Alphavirus/tratamento farmacológico , Encefalomielite/tratamento farmacológico , Encefalomielite/virologia , Neurônios/virologia , Sindbis virus/fisiologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/prevenção & controle , Infecções por Alphavirus/virologia , Animais , Benzodiazepinas/administração & dosagem , Morte Celular , Diazo-Oxo-Norleucina/administração & dosagem , Maleato de Dizocilpina/administração & dosagem , Encefalomielite/imunologia , Encefalomielite/prevenção & controle , Hipocampo/efeitos dos fármacos , Hipocampo/virologia , Humanos , Inflamação/metabolismo , Memória/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/fisiologia , Sindbis virus/patogenicidade , Análise de Sobrevida , Células Th17/metabolismo
12.
Acta Trop ; 141(Pt A): 46-53, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312338

RESUMO

Over 20 species of Angiostrongylus have been described from around the world, but only Angiostrongylus cantonensis has been confirmed to cause central nervous system disease in humans. A neurotropic parasite that matures in the pulmonary arteries of rats, A. cantonensis is the most common cause of eosinophilic meningitis in southern Asia and the Pacific and Caribbean islands. The parasite can also cause encephalitis/encephalomyelitis and rarely ocular angiostrongyliasis. The present paper reviews the life cycle, epidemiology, pathogenesis, clinical features, diagnosis, treatment, prevention and prognosis of A. cantonesis infection. Emphasis is given on the spectrum of central nervous system manifestations and disease pathogenesis.


Assuntos
Angiostrongylus cantonensis/fisiologia , Encefalomielite/epidemiologia , Eosinofilia/epidemiologia , Encefalite Infecciosa/epidemiologia , Estágios do Ciclo de Vida , Meningite/epidemiologia , Infecções por Strongylida/epidemiologia , Animais , Ásia/epidemiologia , Encefalomielite/prevenção & controle , Encefalomielite/terapia , Eosinofilia/prevenção & controle , Eosinofilia/terapia , Humanos , Encefalite Infecciosa/prevenção & controle , Encefalite Infecciosa/terapia , Larva/fisiologia , Meningite/prevenção & controle , Meningite/terapia , Infecções por Strongylida/prevenção & controle , Infecções por Strongylida/terapia
13.
Sci Rep ; 5: 14935, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26447351

RESUMO

Ganciclovir is effective in the treatment of human infections with viruses of the Herpesviridae family. Beside antiviral properties, recently ganciclovir was described to inhibit microglial proliferation and disease severity of experimental autoimmune encephalomyelitis, an inflammatory model of multiple sclerosis. Microglial activation and proliferation are main characteristics of neuroinflammatory CNS diseases and inhibition of microglial functions might be beneficial in autoimmune diseases, or detrimental in infectious diseases. The objective of this study was to determine potential inhibitory effects of ganciclovir in three different murine animal models of CNS neuroinflammation in which microglia play an important role: Theiler´s murine encephalomyelitis, the cuprizone model of de- and remyelination, and the vesicular stomatitis virus encephalitis model. In addition, in vitro experiments with microglial cultures were performed to test the hypothesis that ganciclovir inhibits microglial proliferation. In all three animal models, neither microglial proliferation or recruitment nor disease activity was changed by ganciclovir. In vitro experiments confirmed that microglial proliferation was not affected by ganciclovir. In conclusion, our results show that the antiviral drug ganciclovir does not inhibit microglial activation and proliferation in the murine CNS.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ganciclovir/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Antivirais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/virologia , Células Cultivadas , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/prevenção & controle , Modelos Animais de Doenças , Encefalomielite/prevenção & controle , Encefalomielite/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Ativação de Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Theilovirus/fisiologia , Estomatite Vesicular/prevenção & controle , Estomatite Vesicular/virologia , Vesiculovirus/fisiologia
14.
Hum Gene Ther ; 12(8): 905-20, 2001 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-11387056

RESUMO

Systemic administration of antiinflammatory molecules to patients affected by immune-mediated inflammatory demyelinating diseases of the central nervous system (CNS) has limited therapeutic efficacy due to the presence of the blood-brain barrier (BBB). We found that three of five rhesus monkeys injected intrathecally with a replication-defective herpes simplex virus (HSV) type 1-derived vector engineered with the human interleukin 4 (IL-4) gene were protected from an hyperacute and lethal form of experimental autoimmune encephalomyelitis induced by whole myelin. The intrathecally injected vector consistently diffused within the CNS via the cerebrospinal fluid and infected ependymal cells, which in turn sustained in situ production of IL-4 without overt immunological or toxic side effects. In EAE-protected monkeys, IL-4-gene therapy significantly decreased the number of brain as well as spinal cord inflammatory perivenular infiltrates and the extent of demyelination, necrosis, and axonal loss. The protective effect was associated with in situ downregulation of inflammatory mediators such as tumor necrosis factor alpha (TNF-alpha) and monocyte chemoattractant protein 1 (MCP-1), upregulation of transforming growth factor beta (TGF-beta), and preservation of BBB integrity. Our results indicate that intrathecal delivery of HSV-1-derived vectors containing antiinflammatory cytokine genes may play a major role in the future therapeutic armamentarium of inflammatory CNS-confined demyelinating diseases and, in particular, in the most fulminant forms where conventional therapeutic approaches have, so far, failed to achieve a satisfactory control of the disease evolution.


Assuntos
Doenças Autoimunes/genética , Sistema Nervoso Central/metabolismo , Encefalomielite/genética , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Interleucina-4/genética , Animais , Doenças Autoimunes/prevenção & controle , Barreira Hematoencefálica , Encéfalo/patologia , Divisão Celular , Quimiocina CCL2/biossíntese , Citocinas/biossíntese , Regulação para Baixo , Encefalomielite/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-4/biossíntese , Macaca mulatta , Imageamento por Ressonância Magnética , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/imunologia , Linfócitos T/citologia , Fatores de Tempo , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima
15.
Clin Infect Dis ; 35(10): 1191-203, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12410479

RESUMO

B virus (Cercopithecine herpesvirus 1) is a zoonotic agent that can cause fatal encephalomyelitis in humans. The virus naturally infects macaque monkeys, resulting in disease that is similar to herpes simplex virus infection in humans. Although B virus infection generally is asymptomatic or mild in macaques, it can be fatal in humans. Previously reported cases of B virus disease in humans usually have been attributed to animal bites, scratches, or percutaneous inoculation with infected materials; however, the first fatal case of B virus infection due to mucosal splash exposure was reported in 1998. This case prompted the Centers for Disease Control and Prevention (Atlanta, Georgia) to convene a working group in 1999 to reconsider the prior recommendations for prevention and treatment of B virus exposure. The present report updates previous recommendations for the prevention, evaluation, and treatment of B virus infection in humans and considers the role of newer antiviral agents in postexposure prophylaxis.


Assuntos
Aciclovir/análogos & derivados , Antivirais/uso terapêutico , Encefalomielite/prevenção & controle , Diretrizes para o Planejamento em Saúde , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Cercopitecino 1 , Valina/análogos & derivados , Aciclovir/efeitos adversos , Aciclovir/uso terapêutico , Animais , Antivirais/efeitos adversos , Quimioprevenção/normas , Tratamento Farmacológico/normas , Encefalomielite/tratamento farmacológico , Encefalomielite/etiologia , Seguimentos , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/fisiopatologia , Infecções por Herpesviridae/transmissão , Herpesvirus Cercopitecino 1/efeitos dos fármacos , Herpesvirus Cercopitecino 1/imunologia , Humanos , Primatas/virologia , Valaciclovir , Valina/efeitos adversos , Valina/uso terapêutico
16.
Semin Oncol ; 28(2 Suppl 8): 66-70, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11395856

RESUMO

Erythropoietin (EPO) is a glycoprotein that has been shown to mediate response to hypoxia, and is most notably recognized for its central role in erythropoiesis. In a series of experiments using rodent models, the ability of systemically administered recombinant human erythropoietin (r-HuEPO, epoetin alfa) to cross the blood-brain barrier and affect the outcome of neuronal injury or cognitive function was evaluated. It was shown that EPO and EPO receptors are expressed at capillaries of the brain-periphery interface, and that systemically administered epoetin alfa crossed the blood-brain barrier. Compared with control animals, epoetin alfa significantly reduced tissue damage in an ischemic stroke model when administered 24 hours before inducing stroke, with significant protection still evident when epoetin alfa was administered 6 hours poststroke. Epoetin alfa reduced injury by blunt trauma when administered 24 hours before trauma, with a significantly smaller volume of tissue necrosis noted when compared with controls. The observation that epoetin alfa may reduce nervous system inflammation was confirmed when an experimental autoimmune encephalomyelitis model in which rats were shown to have significantly delayed onset and reduced severity of experimental autoimmune encephalomyelitis symptoms after treatment with epoetin alfa. Epoetin alfa also was shown to ameliorate the latency and severity of seizures, and significantly increase survival versus controls when exposed to kainate. These findings suggest future potential therapeutic uses for epoetin alfa beyond its current use to increase erythropoiesis.


Assuntos
Encéfalo/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Eritropoetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores da Eritropoetina/metabolismo , Animais , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Isquemia Encefálica/prevenção & controle , Encefalomielite/prevenção & controle , Epoetina alfa , Eritropoetina/metabolismo , Humanos , Ácido Caínico , Camundongos , Modelos Animais , Fármacos Neuroprotetores/metabolismo , Proteínas Recombinantes , Convulsões/induzido quimicamente , Convulsões/prevenção & controle
17.
J Neuroimmunol ; 22(1): 77-9, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2918076

RESUMO

In view of recent interest in the potential role of vasoactive amines in the expression of experimental autoimmune encephalomyelitis (EAE) and neuritis (EAN), we set out to determine the effect of slow-release methysergide, a serotonin antagonist, on the effector phase of EAE/EAN in rabbits immunized with homologous spinal cord in Freund's adjuvant. On day 6 post-immunization (p.i.), slow-release pellets of methysergide maleate were implanted subcutaneously in graded doses 0-400 mg. At the highest dose, blood concentrations of methysergide were approximately 90 ng/ml on day 8 p.i. falling to 20 ng/ml by day 16 p.i. However, even at the highest dose of methysergide, rabbits developed typical clinical and histological signs of EAE/EAN. It is concluded that serotonergic mechanisms do not play a critical role in the effector phase of EAE/EAN in the rabbit.


Assuntos
Doenças Autoimunes/prevenção & controle , Encefalomielite/prevenção & controle , Metisergida/uso terapêutico , Animais , Doenças Autoimunes/patologia , Doenças Autoimunes/fisiopatologia , Encefalomielite/patologia , Encefalomielite/fisiopatologia , Feminino , Neurite (Inflamação)/patologia , Neurite (Inflamação)/fisiopatologia , Neurite (Inflamação)/prevenção & controle , Coelhos , Antagonistas da Serotonina/uso terapêutico
18.
Vet Parasitol ; 114(2): 123-30, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12781474

RESUMO

Interferon gamma-knockout mice were challenged with 5000 Sarcocystis neurona sporocysts acquired from a naturally infected opossum. Ponazuril was administered once, by gavage, at day 1, 3, 7, 10, or 14 post-infection (pi). Ponazuril was given at either 20 or 200mg/kg. Mice that survived to day 30 pi were euthanized. Severity of CNS infection was quantified as schizont density in the cerebellum. Unchallenged mice in treatment and non-treatment groups remained free of disease and gained weight throughout the experiment. All challenged mice, regardless of treatment, developed histologic evidence of CNS infection even though clinical signs were prevented in some groups. The greatest treatment benefits were seen in mice given 200mg/kg ponazuril between days 4 and 14 pi. Weight gain over the course of the experiment occurred only in mice that were given 200mg/kg ponazuril on day 7 or 10 pi. With the exception of groups given 200mg/kg ponazuril on day 7 or 14 pi, mice in groups that got sporocysts developed abnormal neurologic signs. No deaths before day 30 pi occurred in mice given ponazuril at 20mg/kg on day 7 pi or 200mg/kg on day 1, 7, 10, or 14 pi. This effect was not significant. Mice given 200mg/kg on day 7 pi had significantly fewer cerebellar schizonts than did those of the control group that was not given ponazuril. These results indicate that single-dose administration of ponazuril for prevention of CNS infection is partially protective when given between days 4 and 14 pi.


Assuntos
Encefalomielite/veterinária , Sarcocistose/veterinária , Triazinas/administração & dosagem , Animais , Peso Corporal , Cerebelo/parasitologia , Modelos Animais de Doenças , Encefalomielite/tratamento farmacológico , Encefalomielite/prevenção & controle , Feminino , Doenças dos Cavalos/tratamento farmacológico , Doenças dos Cavalos/prevenção & controle , Cavalos , Interferon gama/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Gambás , Sarcocystis/efeitos dos fármacos , Sarcocystis/isolamento & purificação , Sarcocistose/tratamento farmacológico , Sarcocistose/prevenção & controle
19.
Avian Dis ; 42(4): 632-47, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9876829

RESUMO

Avian encephalomyelitis control methods were not developed until the 1950s although the disease had been discovered and described over 20 yr earlier. Inability to transmit the infection by other than intracerebral inoculation, lack of suitable immunologic methods, the unknowing use of immune chickens or embryos for experimental studies, and reliance on a highly adapted strain of virus rather than fresh field isolates were the main reasons for a general lack of progress. In the absence of supportive experimental data, at least two commercial breeding organizations turned to the use of a crude chicken brain-propagated virus for vaccination of breeder replacement flocks in the 1950s. This control procedure turned out to be practical and efficacious. Development of suitable embryo infection methods and immunologic tests and the chance finding that antibody-free flocks were essential for experimental studies led to the development of embryo-susceptibility tests to identify immune breeder flocks and formed the basis for another commercially applied control program, the testing and selection of only immune flocks for hatching purposes. The application of the new testing methods coupled with a switch from the adapted Van Roekel strain of virus to fresh field isolates for experimentation resulted in a rapid unraveling of the epizootiology and pathogenesis of the disease and also to the development of a safe and effective vaccine that was licensed for administration to breeder replacements in 1962.


Assuntos
Doenças das Aves/prevenção & controle , Vírus da Encefalomielite Aviária , Encefalomielite/veterinária , Infecções por Enterovirus/veterinária , Doenças das Aves Domésticas/prevenção & controle , Animais , Doenças das Aves/história , Embrião de Galinha , Galinhas , Encefalomielite/história , Encefalomielite/prevenção & controle , Infecções por Enterovirus/história , Infecções por Enterovirus/prevenção & controle , História do Século XX , Doenças das Aves Domésticas/história , Codorniz , Perus , Vacinas Virais/história
20.
Adv Exp Med Biol ; 218: 297-305, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-2829543

RESUMO

1. Maternal antibody protected C57BL/6 and BALB/c suckling mice from the acute, fatal encephalomyelitis caused by MHV-JHM. 2. 40% of the C57BL/6 mice and 25% of the BALB/c mice which were protected by maternal antibody developed neurological disease days to weeks later. Although the clinical syndromes developed by the two different strains were different, in both cases the mice developed a demyelinating encephalomyelitis with fewer inflammatory changes present in the grey matter. 3. Presence or absence of neutralizing antibody in the sera of maternal antibody-protected C57BL/6 mice did not correlate with the development of clinically evident neurological disease. 4. Infectious virus could only be isolated from C57BL/6 mice with neurological disease, although viral antigen could be detected in most mice whether symptomatic or not. 5. This model should be useful for the study of the viral and immune factors important in MHV-induced viral demyelination.


Assuntos
Infecções por Coronaviridae/prevenção & controle , Encefalomielite/prevenção & controle , Imunidade Materno-Adquirida , Animais , Animais Lactentes , Anticorpos Antivirais/biossíntese , Antígenos Virais/isolamento & purificação , Doenças Desmielinizantes/etiologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/imunologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA