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










Base de datos
Intervalo de año de publicación
1.
Vet Immunol Immunopathol ; 202: 130-140, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30078587

RESUMEN

In recent years stem cell therapies have been broadly applied in various disease models specifically immune mediated and degenerative diseases. Whether adipose-derived stem cells might represent a useful therapeutic option in virus-triggered central nervous system diseases has not been investigated so far. Theiler's murine encephalomyelitis (TME) and canine distemper encephalitis are established, virus-mediated animal models sharing many similarities with multiple sclerosis (MS). Canine adipose-derived stem cells (ASC) were selected since dogs might serve as an important translational model for further therapeutic applications. The aim of the present study was to investigate whether canine ASC influence clinical signs, axonal damage, demyelination and inflammation during TME. ASC were transplanted intravenously (iv) or intra-cerebroventricularly (icv) at 7 (early) or 42 (late) days post infection (dpi) in TME virus (TMEV) infected mice. TMEV/ASC iv animals transplanted at 7dpi displayed a transient clinical deterioration in rotarod performance compared to TMEV/control animals. Worsening of clinical signs was associated with significantly increased numbers of microglia/macrophages and demyelination in the spinal cord. In contrast, late transplantation had no influence on clinical findings of TMEV-infected animals. However, late TMEV/ASC iv transplanted animals showed reduced axonal damage compared to TMEV/control animals. Screening of spinal cord and peripheral organs for transplanted ASC revealed no positive cells. Surprisingly, iv transplanted animals showed pulmonary follicular aggregates consisting of T- and B-lymphocytes. Thus, our data suggest that following intravenous application, the lung as priming organ for lymphocytes seems to play a pivotal role in the pathogenesis of TME. Consequences of T-lymphocyte priming in the lung depend on the disease phase and may be responsible for disease modifying effects of ASC.


Asunto(s)
Infecciones por Cardiovirus/terapia , Enfermedades Desmielinizantes/virología , Pulmón/inmunología , Trasplante de Células Madre/efectos adversos , Células Madre/citología , Tejido Adiposo/citología , Animales , Linfocitos T CD4-Positivos/inmunología , Infecciones por Cardiovirus/inmunología , Modelos Animales de Enfermedad , Virus del Moquillo Canino , Perros , Infusiones Intravenosas , Pulmón/patología , Ratones , Esclerosis Múltiple , Theilovirus
2.
J Virol Methods ; 185(2): 204-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22790206

RESUMEN

Encephalomyocarditis virus (EMCV) infection can cause myocarditis and sudden death in pre-weaned piglets and severe reproductive failure in sows. There are no specific antiviral drugs for the treatment of the virus infection. In this study, four recombinant adenoviruses expressing small interfering RNAs (siRNAs) targeting to 1D or 3AB protein genes of EMCV were constructed and their inhibition efficiency on the replication of EMCV was evaluated in both Marc-145 cells and mice. The results showed that the rAd5 expressing siRNAs (rAd5) could inhibit EMCV replication in Marc-145 cells in protein and mRNA levels, as well as the virus yield by approximately 100-1000 times. And the inhibition of viral replication was sustained for 72h and dose-dependent. Animal experiment results showed that EMCV VP1 mRNA level in the brain of mice in the rAd5 groups were obviously lower than those in rAd-G1 and challenge control groups. The virus yields in rAd-1D-2 and rAd-3AB-1 groups were markedly decreased by more than 90.0%. The survival rates of mice in rAd-1D-2 group were significantly higher than those in challenge control groups. Furthermore, the survival mice only showed minor microscopic lesions in brain and minor edema of nerve cell, which was obviously slighter than those in challenge control groups. These results indicated that siRNAs mediated by the adenovirus could provide protective efficacy against EMCV challenge in mice. It might provide a potential strategy for combating EMCV.


Asunto(s)
Adenoviridae/genética , Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Animales , Encéfalo/patología , Encéfalo/virología , Edema Encefálico/patología , Edema Encefálico/virología , Proteínas de la Cápside/genética , Infecciones por Cardiovirus/virología , Línea Celular , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/fisiología , Femenino , Genes Virales , Ingeniería Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética , Plásmidos/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , Factores de Tiempo , Carga Viral , Replicación Viral
3.
J Clin Immunol ; 30 Suppl 1: S50-5, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20387101

RESUMEN

INTRODUCTION: Natural autoantibodies are part of the normal human immunoglobulin repertoire. These antibodies react to self-antigens, are usually polyreactive with relatively low affinity, and typically are of the IgM isotype. Natural IgMs in mice that stimulated remyelination in central nervous system (CNS) demyelinating disease all shared the characteristics of binding to the surface of live oligodendrocytes and myelinated tracts in living slices of CNS tissue. METHODS: A screen for human IgMs with similar character resulted in two human natural antibodies, which when injected peripherally into animal models of demyelination induced remyelination. A recombinant human IgM (rHIgM22) that also promoted remyelination in vivo was constructed. RESULTS: Very small doses of this IgM are required for the promotion of remyelination (EC50 is 460 ng per 20-g mouse). It is clear that after peripheral delivery, rHIgM22 enters the CNS and accumulates in CNS lesions. rHIgM22 was tracked in living mice using ferritin-labeled antihuman mu chain antibodies visualized by magnetic resonance imaging and traditional immunocytochemistry. Although the exact antigen recognized by rHIgM22 is not known, all mouse IgMs that promote remyelination bind to myelin membrane lipids, suggesting the antigen for rHIgM22 is similar. CONCLUSIONS: We propose that the IgMs bind to CNS cells and reorganize the membrane, initiating a signal that results in oligodendrocyte proliferation and/or protection with an end result of increased myelin. Recombinant natural human antibodies are potentially important therapeutic molecules that may modulate a wide spectrum of human disease.


Asunto(s)
Autoanticuerpos/inmunología , Infecciones por Cardiovirus/terapia , Enfermedades Autoinmunes Desmielinizantes SNC/terapia , Inmunoglobulina M/inmunología , Vaina de Mielina/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Barrera Hematoencefálica , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Enfermedades Autoinmunes Desmielinizantes SNC/virología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Humanos , Inmunoglobulina M/uso terapéutico , Lípidos de la Membrana/inmunología , Ratones , Esclerosis Múltiple , Vaina de Mielina/inmunología , Oligodendroglía/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Regeneración , Médula Espinal/inmunología , Theilovirus
4.
J Neurovirol ; 15(1): 90-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19115130

RESUMEN

Most natural killer (NK) T cells express an invariant Valpha14 T-cell receptor. To explore the contribution of NKT cells in an animal model for multiple sclerosis, Theiler's murine encephalomyelitis virus (TMEV) infection, TMEV-infected mice were treated with Valpha14 antibody. Treatment during the early stage of infection delayed the onset of demyelinating disease with higher interleukin-4 production, whereas administration during the late stage or weekly resulted in more severe demyelination with enhanced virus persistence. The effect of in vivo depletion of NKT cells differed depending on the stage of infection, suggesting contrasting roles for NKT cells over the disease course.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Theilovirus , Animales , Anticuerpos/administración & dosificación , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/terapia , Modelos Animales de Enfermedad , Procedimientos de Reducción del Leucocitos , Ratones , Ratones Endogámicos , Esclerosis Múltiple/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/antagonistas & inhibidores , Factores de Tiempo
5.
J Neurovirol ; 12(4): 251-61, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16966216

RESUMEN

Theiler's murine encephalomyelitis virus induces a demyelinating disease (TMEV-IDD) of the central nervous system (CNS) in susceptible mouse strains with accompanying histopathology characterized by mononuclear cell infiltrates. In susceptible strains of mice such as SJL, virus establishes a persistent infection in macrophages, induces a CNS infiltration by macrophages, T cells, and B cells, which results in chronic-progressive paralysis. In the present report the authors have investigated the functional role of CCL2 (monocyte chemotactic protein-1) in the induction and progression of demyelinating disease. Treatment of infected mice at day 0, 14, or 28 with anti-CCL2 resulted in a significant decrease in the clinical disease progression. Further analysis of anti-CCL2-treated mice revealed decreased CNS inflammation and mononuclear cell infiltration with an accompanying change in inflammatory cytokine responses. There was an overall decrease in the absolute numbers of CNS-infiltrating CD4+ T cells, macrophages, and B cells. Finally, anti-CCL2 treatment resulted in decreased viral load in the CNS. These data directly demonstrate a role for CCL2 in the pathogenesis of TMEV-IDD.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Infecciones por Cardiovirus/terapia , Quimiocina CCL2/antagonistas & inhibidores , Enfermedades Desmielinizantes/terapia , Theilovirus/crecimiento & desarrollo , Animales , Anticuerpos Monoclonales/inmunología , Infecciones por Cardiovirus/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Quimiocina CCL2/inmunología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Femenino , Inflamación/inmunología , Inflamación/patología , Inflamación/virología , Leucocitos Mononucleares/inmunología , Ratones , Linfocitos T/inmunología
6.
J Neuroimmunol ; 165(1-2): 1-10, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16106527

RESUMEN

In 1972 Guido Biozzi selectively bred mice to study the immunopathological mechanisms underlying polygenic diseases. One line, the Biozzi antibody high (AB/H) mouse (now designated the ABH strain) was later found to be highly susceptible to many experimentally induced diseases such as autoimmune encephalomyelitis, autoimmune neuritis, autoimmune uveitis, as well as virus-induced demyelination and has thus been a key mouse strain to study human inflammatory neurological diseases. In this paper we discuss the background of the Biozzi ABH mouse and review how studies with these mice have shed light on the pathogenic mechanisms operating in chronic neurological disease.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Biozzi/inmunología , Esclerosis Múltiple/inmunología , Enfermedad Autoinmune Experimental del Sistema Nervioso/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/patología , Infecciones por Alphavirus/terapia , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/terapia , Enfermedad Crónica , Humanos , Ratones , Ratones Biozzi/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Enfermedad Autoinmune Experimental del Sistema Nervioso/genética , Enfermedad Autoinmune Experimental del Sistema Nervioso/patología , Enfermedad Autoinmune Experimental del Sistema Nervioso/terapia , Virus de los Bosques Semliki
7.
J Virol ; 77(3): 2247-50, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12525660

RESUMEN

Transient CD154 blockade at the onset of Theiler's murine encephalomyelitis virus-induced demyelinating disease ameliorated disease progression for 80 days, reduced immune cell infiltration, and transiently increased viral loads in the central nervous system. Peripheral antiviral and autoimmune T-cell responses were normal, and disease severity returned to control levels by day 120.


Asunto(s)
Anticuerpos/uso terapéutico , Ligando de CD40/fisiología , Infecciones por Cardiovirus/terapia , Enfermedades Desmielinizantes/terapia , Theilovirus , Secuencia de Aminoácidos , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Hipersensibilidad Tardía/etiología , Ratones , Datos de Secuencia Molecular , Médula Espinal/patología , Médula Espinal/virología , Células TH1/inmunología , Carga Viral
8.
Eur J Immunol ; 32(3): 606-15, 2002 03.
Artículo en Inglés | MEDLINE | ID: mdl-11857334

RESUMEN

IFN-gamma is an anti-viral and immunomodulatory cytokine critical for resistance to multiple pathogens. Using mice with targeted disruption of the gene for IFN-gamma, we previously demonstrated that this cytokine is critical for resistance to viral persistence and demyelination in the Theiler's virus model of multiple sclerosis. During viral infections, IFN-gamma is produced by natural killer (NK) cells, CD4(+) and CD8(+) T cells; however, the proportions of lymphocyte subsets responding to virus infection influences the contributions to IFN-gamma-mediated protection. To determine the lymphocyte subsets that produce IFN-gamma to maintain resistance, we used adoptive transfer strategies to generate mice with lymphocyte-specific deficiencies in IFN-gamma-production. We demonstrate that IFN-gamma production by both CD4(+) and CD8(+) T cell subsets is critical for resistance to Theiler's murine encephalomyelitis virus (TMEV)-induced demyelination and neurological disease, and that CD4(+) T cells make a greater contribution to IFN-gamma-mediated protection. To determine the cellular targets of IFN-gamma-mediated responses, we used adoptive transfer studies and bone marrow chimerism to generate mice in which either hematopoietic or somatic cells lacked the ability to express IFN-gamma receptor. We demonstrate that IFN-gamma receptor must be present on central nervous system glia, but not bone marrow-derived lymphocytes, in order to maintain resistance to TMEV-induced demyelination.


Asunto(s)
Infecciones por Cardiovirus/terapia , Enfermedades Desmielinizantes/terapia , Inmunoterapia Adoptiva , Interferón gamma/fisiología , Subgrupos Linfocitarios/inmunología , Theilovirus/fisiología , Animales , Antígenos Virales/análisis , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Cruzamientos Genéticos , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Interferón gamma/deficiencia , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple , Desempeño Psicomotor , ARN Viral/análisis , Quimera por Radiación , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/fisiología , Médula Espinal/patología , Theilovirus/inmunología , Receptor de Interferón gamma
9.
Cell Transplant ; 11(8): 753-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12588107

RESUMEN

We used mice to test our hypothesis that in response to viral invasion, stem cells may migrate into the heart and attenuate the effect of viral myocarditis. Male BALB/c mice were divided into three groups: mouse embryonic stem (ES) cell control, encephalomyocarditis virus (EMCV), and EMCV + ES cells. After administration of ES cells via tail vein, mice were immediately inoculated with EMCV. Mice were sacrificed at different days after EMCV inoculation. Mortality was recorded. Inflammatory cell infiltration and necrosis (major pathological changes of viral myocarditis) were evaluated by hematoxylin-eosin staining. ES cell migration and differentiation were identified by immunofluorescence. The survival rate in the EMCV + ES cell group (80%) was significantly increased (p < 0.05) over the EMCV-alone group (64%). Also, the incidence of inflammatory cell infiltration and myocardial lesions was lower in the EMCV + ES cell mice. Furthermore, the result of green fluorescent protein (GFP) and alpha-actinin analysis indicated that ES cells migrated into the heart and differentiated into myocytes after virus inoculation. In conclusion, ES cells significantly increased the survival of viral myocarditis mice and also decreased the necrosis and infiltration of inflammatory cells. These results demonstrated the ability of stem cells to mitigate the effects of viral infection on the heart and illustrated their potential therapeutic application to other mammalian species, including humans.


Asunto(s)
Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis , Miocarditis/terapia , Trasplante de Células Madre , Animales , Infecciones por Cardiovirus/mortalidad , Diferenciación Celular , Línea Celular , Movimiento Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/mortalidad , Miocarditis/virología , Miocitos Cardíacos/citología , Células Madre/citología
10.
J Interferon Cytokine Res ; 21(10): 785-92, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11710989

RESUMEN

Interferon-beta (IFN-beta) has been used successfully to treat patients with relapsing-remitting multiple sclerosis (MS). IFN-tau is a new class of type I IFN that is secreted by the trophoblast and is the signal for maternal recognition of pregnancy in sheep. IFN-tau has potent immunosuppressive and antiviral activities similar to other type I IFN but is less cytotoxic than IFN-alpha/beta. The current investigation concerns the effect of recombinant ovine IFN-tau (rOvIFN-tau) on the modulation of MHC class I and II expression on cloned mouse cerebrovascular endothelial (CVE) cells. IFN-tau induced tyrosine phosphorylation of Stat1 and upregulated the expression of MHC class I on CVE. One proposed action by which type I IFN reduce the relapse rate in MS is via interference with IFN-gamma-induced MHC class II expression. IFN-tau was shown to downregulate IFN-gamma-induced MHC class II expression on CVE and, hence, may be of potential therapeutic value in downregulating inflammation in the central nervous system (CNS). IFN-tau did not upregulate the expression of MHC class II on CVE. IFN-tau also inhibited the replication of Theiler's virus in CVE. These in vitro results suggest that IFN-tau may be of therapeutic value in the treatment of virus-induced demyelinating disease.


Asunto(s)
Antivirales/farmacología , Endotelio Vascular/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Interferón Tipo I/farmacología , Miocardio/citología , Proteínas Gestacionales/farmacología , Theilovirus/efectos de los fármacos , Animales , Infecciones por Cardiovirus/terapia , Células Clonales , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/virología , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Esclerosis Múltiple/terapia , Fosforilación , Factor de Transcripción STAT1 , Ovinos , Theilovirus/crecimiento & desarrollo , Transactivadores/metabolismo , Regulación hacia Arriba , Replicación Viral/efectos de los fármacos
11.
J Pharmacol Exp Ther ; 299(2): 645-51, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11602677

RESUMEN

We have previously shown that immunoglobulin therapy suppressed murine coxsackievirus B3 myocarditis. In the present study, we examined the effects of immunoglobulin upon murine myocarditis induced by encephalomyocarditis virus, which is not pathogenic to humans. Antiviral activity of immunoglobulin (Venilon) against encephalomyocarditis virus could not be detected in vitro. The production of cytokines was decreased in virus-infected macrophages by the treatment of immunoglobulin in vitro. Immunoglobulin (1 g/kg/day) was administered intraperitoneally to the virus-infected C3H/He mice daily for 2 weeks, beginning simultaneously with virus inoculation in experiment I and on day 14 after virus inoculation in experiment II. In experiment I, survival rate did not differ significantly between immunoglobulin-treated and untreated groups. In experiment II, survival rate was higher in immunoglobulin compared with control groups. Immunoglobulin administration suppressed the development of myocardial necrosis with T-lymphocyte infiltrates in mice not only in the acute viremic but in the chronic aviremic stages concomitantly associated with the reduction of inflammatory cytokines, i.e., tumor necrosis factor-alpha, interferon-gamma, macrophage inflammatory protein-2, and interleukin-6. Taken together, immunoglobulin therapy could have the potential to prevent congestive heart failure.


Asunto(s)
Infecciones por Cardiovirus/terapia , Citocinas/biosíntesis , Virus de la Encefalomiocarditis , Insuficiencia Cardíaca/prevención & control , Inmunización Pasiva , Animales , Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/metabolismo , Línea Celular , Citocinas/sangre , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Ratones , Miocardio/patología , Tamaño de los Órganos/efectos de los fármacos
12.
J Virol ; 75(3): 1294-300, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11152502

RESUMEN

Interleukin-8 (IL-8) is a chemotactic cytokine for neutrophils and lymphocytes. Macrophage inflammatory protein 2 (MIP-2) is a murine counterpart of IL-8. The present study was performed to determine whether MIP-2 aggravates murine myocarditis. We examined (i) the MIP-2-producing activity of encephalomyocarditis (EMC) virus-infected cultured macrophages, (ii) serial plasma MIP-2 levels in EMC virus-induced mice by enzyme-linked immunosorbent assay, and (iii) the effects of antimouse MIP-2 monoclonal antibody (MAb) in vivo upon myocarditis. The production of MIP-2 increased in an infection dose- and time-dependent manner in virus-infected RAW 264. 7 macrophages. Five-week-old C(3)H/He mice were inoculated with EMC virus. Plasma MIP-2 levels were significantly elevated in mice on days 7 and 14 postinfection. Mice were injected subcutaneously with anti-MIP-2 MAb at 10 microg/day (group 2) or 100 microg/day (group 3) on days 0 to 5 and were observed until day 21. Uninfected control mice (group 1) were prepared. The survival rate was higher in the anti-MIP-2-treated group (group 3), but not in group 2, than in the control group. Histopathological analysis revealed that cellular infiltration and myocardial necrosis with macrophage and T-cell accumulation were less prominent in the anti-MIP-2 MAb-treated group, but not in group 2, compared to the level in the controls. MIP-2 is an important naturally occurring inflammatory cytokine in myocarditis, and anti-MIP-2 MAb treatment may prevent the inflammatory response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Infecciones por Cardiovirus/metabolismo , Quimiocinas/biosíntesis , Virus de la Encefalomiocarditis/fisiología , Miocarditis/metabolismo , Animales , Western Blotting , Infecciones por Cardiovirus/terapia , Línea Celular , Quimiocina CXCL2 , Quimiocinas/sangre , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C3H , Miocarditis/terapia , Conejos
13.
J Neuroimmunol ; 108(1-2): 22-8, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10900333

RESUMEN

We studied the effect of high-dose mouse IgG on TMEV-induced demyelinating disease (TMEV-IDD). We injected TMEV intracerebrally into susceptible SJL/J mice and induced TMEV-IDD. Mouse IgG were injected intraperitonealy, and clinical course and various immunological indicators were studied. The results show that TMEV-IDD was significantly suppressed both clinically and histologically (P<0.01) when IgG were administered in the effector phase. The delayed type hypersensitivity and T cell proliferative response specific for TMEV were decreased by this treatment. In an ELISPOT assay, the number of TNF-alpha producing lymphocytes in the spinal cords was low in high-dose IgG treated mice compared with PBS treated control mice. These data suggest that administration of IgG suppresses TMEV-IDD and may be promising treatment to prevent exacerbation of human multiple sclerosis.


Asunto(s)
Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/terapia , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/farmacología , Theilovirus/efectos de los fármacos , Theilovirus/fisiología , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/terapia , Infecciones por Cardiovirus/virología , Enfermedades Desmielinizantes/virología , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad Tardía/inmunología , Inmunoglobulina G/inmunología , Interferón gamma/metabolismo , Interleucinas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Médula Espinal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Blood ; 94(8): 2915-22, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10515897

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) establishes a persistent infection in the central nervous system (CNS) leading to an inflammatory demyelinating disease of the CNS in which the histology and clinical course is similar to multiple sclerosis (MS). Disease pathogenesis is primarily due to T-cell-mediated destruction of myelin, which has been attributed to cytopathic effects of the virus, but immune-mediated destruction of myelin mediated via both virus-specific and myelin-specific T cells appear to play the major role. To determine if bone marrow transplantation would be an effective therapy for a virus-initiated autoimmune disease and to better separate viral cytopathic effects from immune-mediated demyelination, we ablated the immune system of TMEV-infected animals with 1,100 cGy total body irradiation, and then the animal's immunity was reconstituted by transplantation of disease-susceptible SJL/J mice with syngeneic marrow or disease-susceptible DBA/2J with marrow from disease-resistant (C57Bl/6 x DBA/2)F1 (B6D2) donors. Hematopoietic transplant performed after onset of disease resulted in 42% mortality in SJL/J syngeneic transplants, 47% mortality in diseased DBA2 recipients restored with marrow from naive B6D2 donors, and 12% in diseased DBA2 recipients receiving marrow from B6D2 donors previously infected with TMEV. Delayed type hypersensitivity (DTH) to both virion and myelin proteins was decreased in surviving mice that underwent transplantation; however, CNS viral titers were significantly elevated compared with nontransplanted controls. We conclude that a functional immune system with appropriate T-cell responses are important in prevention of lethal cytopathic CNS effects from TMEV. Relevant to the clinical use of bone marrow transplantation, attempts to ablate the immune system in viral-mediated immune diseases or virus-initiated autoimmune disease may have acute and lethal consequences. Our results raise concern about the attempted use of autologous hematopoietic transplantation in patients with MS, an autoimmune disease with a suspected virus etiology, particularly if the graft is aggressively depleted of lymphocytes.


Asunto(s)
Enfermedades Autoinmunes/terapia , Infecciones por Cardiovirus/terapia , Sistema Nervioso Central/virología , Enfermedades Desmielinizantes/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Depleción Linfocítica/efectos adversos , Esclerosis Múltiple/terapia , Theilovirus/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Contraindicaciones , Citocinas/biosíntesis , Efecto Citopatogénico Viral , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos , Vaina de Mielina/inmunología , Médula Espinal/metabolismo , Médula Espinal/virología , Linfocitos T Citotóxicos/inmunología , Theilovirus/inmunología , Theilovirus/aislamiento & purificación , Latencia del Virus , Replicación Viral , Irradiación Corporal Total
15.
J Neuroimmunol ; 97(1-2): 110-8, 1999 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10408964

RESUMEN

An augmentation of experimental allergic encephalomyelitis (EAE) was observed when monoclonal antibody (mAb) to intercellular adhesion molecule 1 (ICAM-1) was administered after adoptive transfer. Clinical disease was more severe in the ICAM-1 specific mAb-treated EAE mice and included prominent ataxia compared to the PBS-treated controls or Theiler's murine encephalomyelitis virus (TMEV) infected mice treated with ICAM-1 specific mAb. Neuropathologic evaluation demonstrated a distinctly different distribution of lesions in the anti-ICAM-1-treated EAE mice which featured prominent demyelination and inflammation in the cerebellum, brainstem and cerebrum. These structures were minimally involved in the control mice and mAb treatment did not alter the neuropathology in TMEV-infected mice. These results indicate that anti-ICAM-1 can alter trafficking of lymphocytes and mononuclear cells in EAE but not TMEV-induced demyelinating disease.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Infecciones por Cardiovirus/terapia , Encefalomielitis Autoinmune Experimental/terapia , Theilovirus , Animales , Antígenos Virales/inmunología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/terapia , Enfermedades Desmielinizantes/virología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Molécula 1 de Adhesión Intercelular/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/farmacología , Recurrencia
16.
Int J Immunopharmacol ; 18(3): 183-92, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8796446

RESUMEN

The use of interferon-alpha (IFN-alpha) in the treatment of infectious diseases has shown limited efficacy and dose-limiting toxicity. We have selected safe immunomodulators of the muramyl peptide family with the potential of enhancing the efficacy of IFN-alpha without resulting in increased toxicity. One of these synthetic muramyl dipeptide (MDP) derivatives, namely murabutide which is in a clinical stage of development, has been recently found to synergize with IFN-alpha 2a in the selective induction of anti-inflammatory mediators and to enhance the biological activities of the therapeutic cytokine. The present study was performed to assess the antiviral activity of such muramyl peptides and a possible potentiation of the antiviral activity of IFN-alpha/beta by associated therapy using the classical assay of Encephalomyocarditis virus (EMCV) infection. In vitro, pretreatment of Moloney Sarcoma virus (MSV)-transformed cell line with MDP derivatives followed by treatment with IFN-alpha/beta showed a synergistic protection against the cytopathogenic effect of a subsequent EMCV infection. None of the MSV cultures could be protected by stimulation with muramyl peptides alone. In vivo, all of the muramyl peptide derivatives tested were found to be more potent than the parent molecule MDP in inducing protection against death or in the prolongation of the mean survival time of infected mice. Sequential administration of suboptimal doses of exogenous IFN-alpha/beta and muramyl peptides established a strong antiviral state and considerably improved the protective effect of the cytokine, frequently leading to an abortive infection. Our findings suggest that combination therapy with safe muramyl peptides and IFN-alpha/beta could constitute a highly effective and new regimen for the treatment of viral infections in humans.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis , Interferón beta/uso terapéutico , Interferón gamma/uso terapéutico , Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Animales , Línea Celular/efectos de los fármacos , Sinergismo Farmacológico , Masculino , Ratones
17.
Eur Heart J ; 16 Suppl O: 140-3, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8682082

RESUMEN

We have developed murine models of viral myocarditis induced by the encephalomyocarditis (EMC) virus in which there is a high incidence of severe myocarditis, congestive heart failure and dilated cardiomyopathy. From these models, we have learned of their natural history and pathogenesis and assessed new diagnostic methods and therapeutic and preventive interventions. Our recent studies showed that increased circulating cytokines have been detected in patients with acute myocarditis and cardiomyopathy and suggest that cytokines may play some role in the pathogenesis of myocardial injury in these diseases. In our animal model of EMC virus myocarditis, plasma tumour necrosis factor-(TNF)-alpha was elevated in the acute stage and exogenously administered anti-TNF-alpha antibody improved survival and reduced the myocardial lesion, suggesting the importance of TNF-alpha in the pathogenesis. A recently developed positive inotropic agent, vesnarinone, was effective in the treatment of EMC virus myocarditis by its immunomodulating effects such as inhibition of production of TNF-alpha The plasma angiotensin II level was increased in EMC virus myocarditis, and a new angiotensin II type I antagonist, TCV-116, prevented development of myocarditis.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Cardiomiopatía Dilatada/terapia , Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis , Insuficiencia Cardíaca/terapia , Miocarditis/terapia , Tetrazoles , Angiotensina II/sangre , Antagonistas de Receptores de Angiotensina , Animales , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Cardiomiopatía Dilatada/inmunología , Cardiotónicos/uso terapéutico , Infecciones por Cardiovirus/inmunología , Citocinas/antagonistas & inhibidores , Citocinas/sangre , Virus de la Encefalomiocarditis/efectos de los fármacos , Insuficiencia Cardíaca/inmunología , Humanos , Ratones , Miocarditis/inmunología , Profármacos/uso terapéutico , Pirazinas , Quinolinas/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Virol ; 69(12): 8147-50, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7494339

RESUMEN

The injection of 20 ng of mouse interleukin-12 (IL-12) protects mice from a lethal infection with encephalomyocarditis virus. In vitro, an anti-gamma interferon (anti-IFN-gamma) monoclonal antibody but not an anti-IL-12 monoclonal antibody neutralizes the antiviral activity present in the supernatants of splenocytes stimulated with IL-12. Finally, IL-12 fails to protect 129 Sv/Ev IFN-gamma R0/0 mice against encephalomyocarditis virus infection. These results demonstrate that IL-12 exerts its antiviral activity through the induction of endogenous IFN-gamma.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis , Interferón gamma/farmacología , Interleucina-12/uso terapéutico , Animales , Anticuerpos Monoclonales/farmacología , Antivirales/inmunología , Infecciones por Cardiovirus/inmunología , Relación Dosis-Respuesta a Droga , Humanos , Interferón Tipo I/inmunología , Interferón Tipo I/farmacología , Interferón gamma/inmunología , Interleucina-12/inmunología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Pruebas de Neutralización , Proteínas Recombinantes , Bazo/inmunología , Factores de Tiempo
19.
J Pharmacol Exp Ther ; 274(1): 494-8, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7616436

RESUMEN

The effects of combination therapy with the immunomodulators OK432 (derived from the Su strain of Streptococcus pyogenes A3; 1 unit corresponds to 0.1 mg of dried streptococci dissolved in 0.1 ml of saline) and human recombinant interferon-alpha A/D (IFN) on cardiac atrial natriuretic peptide (ANP) gene expression and myocardial hypertrophy were examined in a murine model of viral myocarditis with congestive heart failure. Therapy was started 24 h after inoculation with encephalomyocarditis virus and was continued for 14 days. The plasma ANP concentration in untreated infected mice was significantly (P < .01) increased on day 10 (115 +/- 48 pg/ml) and day 30 (43 +/- 22 pg/ml) after inoculation relative to that in uninfected controls (5 +/- 4 pg/ml), whereas plasma ANP levels in treated mice were significantly (P < .01) reduced on day 10 (14 +/- 13 pg/ml) and day 30 (11 +/- 9 pg/ml) in comparison with untreated infected mice. The atrial and ventricular ANP messenger RNA (mRNA) concentrations in untreated mice showed increases of approximately 1.4- and 29.3-fold, respectively, on day 10 and increases of 1.8- and 34-fold, respectively, on day 30 compared with the concentration in uninfected controls. Combined OK432 and IFN significantly (P < .01) reduced the increase in ANP mRNA concentration in ventricles to 6.0- and 6.7-fold on days 10 and 30, respectively. Neither OK432 nor IFN monotherapy reduced the ANP mRNA concentrations in atria and ventricles compared with those in untreated controls.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Factor Natriurético Atrial/genética , Infecciones por Cardiovirus/terapia , Regulación de la Expresión Génica , Interferón Tipo I/uso terapéutico , Miocarditis/terapia , Picibanil/uso terapéutico , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/fisiopatología , Virus de la Encefalomiocarditis , Femenino , Humanos , Interferón-alfa , Ratones , Ratones Endogámicos C3H , Miocarditis/genética , Miocarditis/fisiopatología , Miocarditis/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes
20.
J Clin Invest ; 94(3): 1212-7, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8083362

RESUMEN

Controversy still exists concerning the therapy for viral myocarditis which manifests a wide variety of clinical symptoms. Vesnarinone, a quinolinone derivative that was developed as a positive inotropic agent with complex actions, including phosphodiesterase inhibition and cation channel modification, has recently been confirmed to improve the prognosis of patients with chronic heart failure. However, the precise mechanism of this beneficial effect is not yet clearly understood. In this study, using a murine model of acute viral myocarditis resulting from encephalomyocarditis virus infection, survival and myocardial damage were markedly improved by treatment with vesnarinone. In contrast, survival was not improved by treatment with amrinone, a phosphodiesterase inhibitor. Although vesnarinone did not inhibit viral replication or protect myocytes from viral direct cell injury, it did inhibit the increase in natural killer cell activity after viral infection. On the other hand, amrinone failed to inhibit natural killer cell activity. Both vesnarinone and amrinone suppressed the production of tumor necrosis factor-alpha. Therefore, we postulate that vesnarinone exerted its beneficial effects through an inhibition of natural killer cell activity, and that it serves as an immunomodulator providing new therapeutic possibilities for the treatment of viral myocarditis and/or immunological disorders.


Asunto(s)
Cardiomiopatías/terapia , Infecciones por Cardiovirus/terapia , Virus de la Encefalomiocarditis , Inmunosupresores/uso terapéutico , Células Asesinas Naturales/inmunología , Miocardio/patología , Quinolinas/toxicidad , Factor de Necrosis Tumoral alfa/biosíntesis , Amrinona/uso terapéutico , Animales , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Femenino , Células Asesinas Naturales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos , Embarazo , Pirazinas , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA