Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
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.
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
3.
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 , Vírus Sindbis/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 , Vírus Sindbis/patogenicidade , Análise de Sobrevida , Células Th17/metabolismo
4.
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
5.
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
6.
Shock ; 40(6): 471-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24088994

RESUMO

Traumatic brain injury (TBI) is a leading cause of mortality and disability. Acute postinjury insults after TBI, such as hypoxia, contribute to secondary brain injury and worse clinical outcomes. The functional and neuroinflammatory effects of brief episodes of hypoxia experienced following TBI have not been evaluated. Our previous studies have identified interleukin 6 (IL-6) as a potential mediator of mild TBI-induced pathology. In the present study, we sought to determine the effects of brief hypoxia on mild TBI and whether IL-6 played a role in the neuroinflammatory and functional deficits after injury. A murine model of mild TBI was induced by a weight drop (500 g from 1.5 cm). After injury, mice were exposed to immediate hypoxia (FIO2 = 15.1%) or normoxia (FIO2 = 21%) for 30 min. Serum and brain samples were analyzed for inflammatory cytokines 24 h after TBI. Neuron-specific enolase was measured as a serum biomarker of brain injury. Evaluation of motor coordination was performed for 5 days after TBI using a rotarod device. In some animals, anti-IL-6 was administered following TBI and hypoxia to neutralize systemic IL-6. Mice undergoing TBI had significant increases in brain injury. Exposure to brief hypoxia after TBI resulted in a more than 5-fold increase in serum neuron-specific enolase. This increase was associated with increases in serum and brain cytokine expression, suggesting that brief hypoxia exacerbates systemic and brain inflammation. Neutralization of IL-6 suppressed postinjury neuroinflammation and neuronal injury. In addition, TBI and hypoxia induced significant motor coordination deficits that were completely abrogated by IL-6 blockade. Exposure to hypoxia after TBI induces neuroinflammation and brain injury. These changes can be mitigated by neutralization of systemic IL-6. Interleukin 6 blockade also corrected the TBI-induced deficit in motor coordination. These data suggest that systemic IL-6 modulates the degree of neuroinflammation and contributes to reduced motor coordination after mild TBI.


Assuntos
Lesões Encefálicas/complicações , Encefalomielite/etiologia , Hipóxia Encefálica/complicações , Interleucina-6/fisiologia , Transtornos das Habilidades Motoras/etiologia , Animais , Anticorpos Neutralizantes/uso terapêutico , Biomarcadores/sangue , Lesões Encefálicas/sangue , Modelos Animais de Doenças , Encefalomielite/sangue , Encefalomielite/prevenção & controle , Hipóxia Encefálica/sangue , Mediadores da Inflamação/sangue , Mediadores da Inflamação/fisiologia , Interleucina-6/antagonistas & inibidores , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos das Habilidades Motoras/sangue , Transtornos das Habilidades Motoras/prevenção & controle , Fosfopiruvato Hidratase/sangue
7.
Am J Vet Res ; 74(2): 248-56, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23363350

RESUMO

OBJECTIVE: To evaluate metaphylactic RNA interference to prevent equine herpesvirus type 1 (EHV-1) infection in experimental herpesvirus myeloencephalopathy in horses and to determine whether horses infected with a neuropathogenic strain of the virus that develop equine herpesvirus myeloencephalopathy (EHM) have differences in viremia. ANIMALS: 13 seronegative horses. PROCEDURES: EHV-1 strain Ab4 was administered intranasally on day 0, and small interfering RNAs (siRNAs [EHV-1 specific siRNAs {n = 7} or an irrelevant siRNA {6}]) were administered intranasally 24 hours before and 12, 24, 36, and 48 hours after infection. Physical and neurologic examinations, nasal swab specimens, and blood samples were collected for virus isolation and quantitative PCR assay. Data from the study were combined with data from a previous study of 14 horses. RESULTS: No significant difference was detected in clinical variables, viremia, or detection of EHV-1 in nasal swab specimens of horses treated with the EHV-1 targeted siRNAs (sigB3-siOri2) versus controls. No significant differences in viremia were detected between horses that developed EHM and those that did not. CONCLUSIONS AND CLINICAL RELEVANCE: Administration of siRNAs targeted against EHV-1 around the time of EHV-1 infection was not protective with this experimental design. Horses infected with the neuropathogenic EHV-1 strain Ab4 that developed EHM did not have a more pronounced viremia.


Assuntos
Encefalomielite/veterinária , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/fisiologia , Doenças dos Cavalos/genética , Doenças dos Cavalos/prevenção & controle , RNA Interferente Pequeno/uso terapêutico , Viremia/virologia , Animais , DNA Viral/genética , Encefalomielite/genética , Encefalomielite/prevenção & controle , Encefalomielite/virologia , Feminino , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/classificação , Herpesvirus Equídeo 1/patogenicidade , Doenças dos Cavalos/virologia , Cavalos , Masculino , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Fatores de Tempo
8.
Transbound Emerg Dis ; 59(3): 256-63, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21975071

RESUMO

Equid herpesvirus 1 (EHV-1)-associated myeloencephalopathy (EHM) is a disease affecting the central nervous system of horses. Despite the constantly increasing interest about this syndrome, epidemiological data are limited especially when related to the description of large outbreaks. The aim of this article is to describe clinical, virological and molecular data obtained throughout a severe outbreak of EHM, with emphasis on laboratory diagnostic methods. The epidemic disease concerned a riding school in France where 7/66 horses aged 12-22 years developed signs of neurological disease in July 2009. Diagnosis of EHM was supported by EHV-1 detection using both real-time PCR and virus culture, and SNP-PCR test for viral strain characterization. EHM morbidity was 10.6% (7/66), mortality was 7.5% (5/66) and case fatality rate was 71.4% (5/7). Clinical presentation of the disease was characterized by the fact that fever was systematically present within 2 days before the severe neurological signs were noted. EHV-1 was detected by PCR in each available blood and nasal swab samples. Neuropathogenic strain only (G(2254) ) was isolated during the current outbreak; C(t) values, used as an indicative level of the viral load, ranged 26.0-37.0 among the six sampled horses. The amount of virus in biological samples was not systematically related to the intensity of the clinical signs being observed. In conclusion, this article described a severe outbreak of EHM while limited in time and restricted to one premise. Molecular data strongly suggested taking into account any low viral load as being a potential risk factor for neurological manifestations.


Assuntos
Surtos de Doenças/veterinária , Encefalomielite/veterinária , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1 , Doenças dos Cavalos/epidemiologia , Animais , Encefalomielite/epidemiologia , Encefalomielite/prevenção & controle , Encefalomielite/virologia , Feminino , França/epidemiologia , Infecções por Herpesviridae/epidemiologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Doenças dos Cavalos/prevenção & controle , Cavalos , Masculino , Vacinas Virais/imunologia
9.
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
10.
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
11.
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
12.
Can J Vet Res ; 73(3): 176-83, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19794889

RESUMO

Sarcocystis neurona is the principal etiologic agent of equine protozoal myeloencephalitis (EPM). An immunodominant protein of S. neurona, SnSAG-1, is expressed by the majority of S. neurona merozoites isolated from spinal tissues of horses diagnosed with EPM and may be a candidate for diagnostic tests and prophylaxis for EPM. Five horses were vaccinated with adjuvanted recombinant SnSAG1 (rSnSAG1) and 5 control (sham vaccinated) horses were vaccinated with adjuvant only. Serum was evaluated pre- and post-vaccination, prior to challenge, for antibodies against rSnSAG1 and inhibitory effects on the infectivity of S. neurona by an in vitro serum neutralization assay. The effect of vaccination with rSnSAG1 on in vivo infection by S. neurona was evaluated by challenging all the horses with S. neurona merozoites. Blinded daily examinations and 4 blinded neurological examinations were used to evaluate the presence of clinical signs of EPM. The 5 vaccinated horses developed serum and cerebrospinal fluid (CSF) titers of SnSAG1, detected by enzyme-linked immunosorbent assay (ELISA), post-vaccination. Post-vaccination serum from vaccinated horses was found to have an inhibitory effect on merozoites, demonstrated by in vitro bioassay. Following the challenge, the 5 control horses displayed clinical signs of EPM, including ataxia. While 4 of the 5 vaccinated horses did not become ataxic. One rSnSAG-1 vaccinated horse showed paresis in 1 limb with muscle atrophy. All horses showed mild, transient, cranial nerve deficits; however, disease did not progress to ataxia in rSnSAG-1 vaccinated horses. The study showed that vaccination with rSnSAG-1 produced antibodies in horses that neutralized merozoites when tested by in vitro culture and significantly reduced clinical signs demonstrated by in vivo challenge.


Assuntos
Antígenos de Protozoários/farmacologia , Encefalomielite/veterinária , Doenças dos Cavalos/parasitologia , Proteínas de Protozoários/farmacologia , Proteínas Recombinantes/farmacologia , Sarcocystis/imunologia , Sarcocistose/veterinária , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/líquido cefalorraquidiano , Encefalomielite/imunologia , Encefalomielite/parasitologia , Encefalomielite/prevenção & controle , Feminino , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/prevenção & controle , Cavalos , Masculino , Testes de Neutralização/veterinária , Sarcocistose/imunologia , Sarcocistose/parasitologia , Sarcocistose/prevenção & controle , Método Simples-Cego , Vacinação/veterinária
13.
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
14.
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
15.
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 , Vírus Sindbis , Viroses/tratamento farmacológico , Viroses/patologia
16.
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
17.
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
18.
J Immunol ; 171(4): 2006-13, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902505

RESUMO

CTL escape mutations have been identified in several chronic infections, including mice infected with mouse hepatitis virus strain JHM. One outstanding question in understanding CTL escape is whether a CD8 T cell response to two or more immunodominant CTL epitopes would prevent CTL escape. Although CTL escape at multiple epitopes seems intuitively unlikely, CTL escape at multiple CD8 T cell epitopes has been documented in some chronically infected individual animals. To resolve this apparent contradiction, we engineered a recombinant variant of JHM that expressed the well-characterized gp33 epitope of lymphocytic choriomeningitis virus, an epitope with high functional avidity. The results show that the presence of a host response to this second epitope protected mice against CTL escape at the immunodominant JHM-specific CD8 T cell epitope, the persistence of infectious virus, and the development of clinical disease.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Encefalomielite/imunologia , Encefalomielite/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Latência Viral/imunologia , Sequência de Aminoácidos , Animais , Antígenos Virais/administração & dosagem , Antígenos Virais/análise , Antígenos Virais/biossíntese , Antígenos Virais/genética , Doença Crônica , Infecções por Coronavirus/genética , Modelos Animais de Doenças , Encefalomielite/genética , Encefalomielite/virologia , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/análise , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/genética , Feminino , Deleção de Genes , Glicoproteínas/administração & dosagem , Glicoproteínas/análise , Glicoproteínas/biossíntese , Glicoproteínas/genética , Células HeLa , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/biossíntese , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia , Proteínas Virais/administração & dosagem , Proteínas Virais/análise , Proteínas Virais/biossíntese , Proteínas Virais/genética , Latência Viral/genética
19.
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
20.
J Immunol ; 170(6): 3204-13, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12626579

RESUMO

CD8(+) T cells infiltrating the CNS control infection by the neurotropic JHM strain of mouse hepatitis virus. Differential susceptibility of infected cell types to clearance by perforin or IFN-gamma uncovered distinct, nonredundant roles for these antiviral mechanisms. To separately evaluate each effector function specifically in the context of CD8(+) T cells, pathogenesis was analyzed in mice deficient in both perforin and IFN-gamma (PKO/GKO) or selectively reconstituted for each function by transfer of CD8(+) T cells. Untreated PKO/GKO mice were unable to control the infection and died of lethal encephalomyelitis within 16 days, despite substantially higher CD8(+) T cell accumulation in the CNS compared with controls. Uncontrolled infection was associated with limited MHC class I up-regulation and an absence of class II expression on microglia, coinciding with decreased CD4(+) T cells in CNS infiltrates. CD8(+) T cells from perforin-deficient and wild-type donors reduced virus replication in PKO/GKO recipients. By contrast, IFN-gamma-deficient donor CD8(+) T cells did not affect virus replication. The inability of perforin-mediated mechanisms to control virus in the absence of IFN-gamma coincided with reduced class I expression. These data not only confirm direct antiviral activity of IFN-gamma within the CNS but also demonstrate IFN-gamma-dependent MHC surface expression to guarantee local T cell effector function in tissues inherently low in MHC expression. The data further imply that IFN-gamma plays a crucial role in pathogenesis by regulating the balance between virus replication in oligodendrocytes, CD8(+) T cell effector function, and demyelination.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Viroses do Sistema Nervoso Central/imunologia , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe I/biossíntese , Interferon gama/fisiologia , Glicoproteínas de Membrana/fisiologia , Regulação para Cima/imunologia , Doença Aguda , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/transplante , Viroses do Sistema Nervoso Central/genética , Viroses do Sistema Nervoso Central/mortalidade , Viroses do Sistema Nervoso Central/prevenção & controle , Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/prevenção & controle , Citotoxicidade Imunológica/genética , Encefalomielite/genética , Encefalomielite/imunologia , Encefalomielite/mortalidade , Encefalomielite/prevenção & controle , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe II/genética , Interferon gama/deficiência , Interferon gama/genética , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/patogenicidade , Perforina , Proteínas Citotóxicas Formadoras de Poros
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...