Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 16(1): 58, 2019 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-30851725

RESUMEN

BACKGROUND: Corticosteroids dominate in the treatment of chronic autoimmune neuropathies although long-term use is characterized by devastating side effects. METHODS: We introduce the intrathecal application of the synthetic steroid triamcinolone (TRIAM) as a novel therapeutic option in experimental autoimmune neuritis in Lewis rats RESULTS: After immunization with neuritogenic P2 peptide, we show a dose-dependent therapeutic effect of one intrathecal injection of 0.3 or 0.6 mg/kg TRIAM on clinical and electrophysiological parameters of neuritis with a lower degree of inflammatory infiltrates (T cells and macrophages) and demyelination in the sciatic nerve. In vitro studies in Schwann cell cultures showed an increased expression of IL-1 receptor antagonist and reduced expression of Toll-like receptor 4 after incubation with TRIAM as well as a protective effect of TRIAM against oxidative stress after H2O2 exposure. CONCLUSION: Intrathecal TRIAM application could be a novel immunomodulatory and potentially neuroprotective option for autoimmune neuropathies with a direct effect on Schwann cells.


Asunto(s)
Antiinflamatorios/administración & dosificación , Neuritis Autoinmune Experimental/tratamiento farmacológico , Neuritis Autoinmune Experimental/patología , Estrés Oxidativo/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Triamcinolona Acetonida/administración & dosificación , Animales , Antígenos CD/metabolismo , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Espinales/métodos , Ganglios Linfáticos/citología , Masculino , Conducción Nerviosa/efectos de los fármacos , Neuritis Autoinmune Experimental/inducido químicamente , Ratas , Ratas Endogámicas Lew , Factores de Transcripción SOXE/metabolismo , Antígenos Thy-1/metabolismo
2.
J Neuroinflammation ; 15(1): 217, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068351

RESUMEN

BACKGROUND: Autoimmune polyneuropathies are acquired inflammatory disorders of the peripheral nervous system (PNS) characterized by inflammation, demyelination, and axonal degeneration. Although the pathogenesis has not been fully elucidated, T cells recognizing self-antigens are believed to initiate inflammation in a subgroup of patients. However, the route and time of T cell entry into the PNS have not yet been described in detail. In this study, we analyzed both kinetics as well as localization of retrovirally transfected green fluorescent protein (GFP)-expressing neuritogenic T lymphocytes in experimental autoimmune neuritis (EAN). METHODS: T lymphocytes obtained from rats following EAN induction by immunization with peripheral nerve protein peptide P255-78 were retrovirally engineered to express GFP. Non-specific T cells were negatively selected by in vitro restimulation, whereas GFP-expressing neuritogenic T cells (reactive to P255-78) were adoptively transferred into healthy rats (AT-EAN). Antigen-specific T cell tracking and localization was performed by flow cytometry and immunohistochemistry during the course of disease. RESULTS: After induction of autoimmune neuritis, P2-reactive T cells were detectable in the liver, spleen, lymph nodes, lung, peripheral blood, and the sciatic nerves with distinct kinetics. A significant number of GFP+ T cells appeared early in the lung with a peak at day four. In the peripheral nerves within the first days, GFP-negative T cells rapidly accumulated and exceeded the number of GFP-expressing cells, but did not enter the endoneurium. Very early after adoptive transfer, T cells are found in proximity to peripheral nerves and in the epineurium. However, only GFP-expressing neuritogenic T cells are able to enter the endoneurium from day five after transfer. CONCLUSIONS: Our findings suggest that neuritogenic T cells invade the PNS early in the course of disease. However, neuritogenic T cells cross the blood-nerve barrier with a certain delay without preference to dorsal roots. Further understanding of the pathophysiological role of autoagressive T cells may help to improve therapeutic strategies in immune-mediated neuropathies.


Asunto(s)
Neuritis Autoinmune Experimental/inmunología , Neuritis Autoinmune Experimental/patología , Nervios Periféricos/patología , Linfocitos T/fisiología , Traslado Adoptivo , Animales , Antígenos CD4/metabolismo , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/inmunología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína P2 de Mielina/metabolismo , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/cirugía , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Endogámicas Lew , Linfocitos T/metabolismo , Factores de Tiempo , Transducción Genética
3.
J Neuroinflammation ; 15(1): 51, 2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29467007

RESUMEN

BACKGROUND: With the recognition of the key roles of cellular metabolism in immunity, targeting metabolic pathway becomes a new strategy for autoimmune disease treatment. Guillain-Barré syndrome (GBS) is an acute immune-mediated inflammatory demyelinating disease of the peripheral nervous system, characterized by inflammatory cell infiltration. These inflammatory cells, including activated macrophages, Th1 cells, and Th17 cells, generally undergo metabolic reprogramming and rely mainly on glycolysis to exert functions. This study aimed to explore whether enhanced glycolysis contributed to the pathogenesis of experimental autoimmune neuritis (EAN), a classic model of GBS. METHODS: Preventive and therapeutic treatments with glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), were applied to EAN rats. The effects of treatments were determined by clinical scoring, weighting, and tissue examination. Flow cytometry and ELISA were used to evaluate T cell differentiation, autoantibody level, and macrophage functions in vivo and in vitro. RESULTS: Glycolysis inhibition with 2-DG not only inhibited the initiation, but also prevented the progression of EAN, evidenced by the improved clinical scores, weight loss, inflammatory cell infiltration, and demyelination of sciatic nerves. 2-DG inhibited the differentiation of Th1, Th17, and Tfh cells but enhanced Treg cell development, accompanied with reduced autoantibody secretion. Further experiments in vitro proved glycolysis inhibition decreased the nitric oxide production and phagocytosis of macrophages and suppressed the maturation of dendritic cells (DC). CONCLUSION: The effects of glycolysis inhibition on both innate and adaptive immune responses and the alleviation of animal clinical symptoms indicated that enhanced glycolysis contributed to the pathogenesis of EAN. Glycolysis inhibition may be a new therapy for GBS.


Asunto(s)
Glucólisis/fisiología , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/metabolismo , Animales , Desoxiglucosa/farmacología , Desoxiglucosa/uso terapéutico , Glucólisis/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/fisiología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Neuritis Autoinmune Experimental/tratamiento farmacológico , Células RAW 264.7 , Ratas , Ratas Endogámicas Lew
4.
J Vis Exp ; (129)2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29155769

RESUMEN

Experimental autoimmune neuritis (EAN) is a well-appreciated experimental model of autoimmune peripheral demyelinating diseases. EAN disease is induced by immunizing mice with neurogenic peptides to direct an inflammatory attack toward components of the peripheral nervous system (PNS). Recent advances have enabled the induction of EAN in the relatively resistant C57BL/6 mouse line using myelin protein zero (P0)106-125 or P0180-199 peptides delivered in adjuvant combined with the injection of pertussis toxin. The ability to induce EAN in the C57BL/6 strain allows for the use of the numerous genetic tools that exist on this mouse background, and thus allows the sophisticated study of disease pathogenesis and interrogation of the mechanistic action of novel therapeutics in combination with transgenic approaches. In this study, we demonstrate a simple approach to successfully induce EAN using the P0180-199 peptide in C57BL/6 mice. We also outline a protocol for the assessment of functional deficits that occur in this model, accompanied by an array of neuropathological features. Thus, this model is a powerful experimental model to study the pathogenesis of human peripheral demyelinating neuropathies, and to determine the efficacy of potential therapies that aim to promote myelin repair and protect against nerve damage in autoimmune neuritis.


Asunto(s)
Neuritis Autoinmune Experimental/inducido químicamente , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Neuroimmunol ; 305: 72-74, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28284349

RESUMEN

We investigated the effect of 4-aminopyridine (4-AP) on experimental autoimmune neuritis (EAN) using a 4-AP-treated group in which 4-AP was administered in the diet, and a control group (n=10 per group). Electrophysiological and pathological assessment was performed in the sciatic nerve. The EAN clinical scores were significantly lower in the 4-AP-treated group than in the control group (p<0.05). The motor conductance velocity two weeks post-immunization was significantly higher in the 4-AP-treated group (p<0.05). Finally, 4-AP did not lead to pathological changes. Thus, 4-AP might be a potential therapeutic agent in demyelinating neuropathy.


Asunto(s)
4-Aminopiridina/uso terapéutico , Neuritis Autoinmune Experimental/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/uso terapéutico , 4-Aminopiridina/farmacología , Animales , Modelos Animales de Enfermedad , Electromiografía , Potenciales Evocados Motores/efectos de los fármacos , Potenciales Evocados Motores/fisiología , Adyuvante de Freund/toxicidad , Masculino , Proteínas de la Mielina/toxicidad , Conducción Nerviosa/efectos de los fármacos , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/inmunología , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Endogámicas Lew , Estadísticas no Paramétricas , Vacunación/efectos adversos
6.
J Neuropathol Exp Neurol ; 76(2): 89-100, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28082327

RESUMEN

We assessed novel disability-based parameters and neuropathological features of the P0180-190 peptide-induced model of experimental autoimmune neuritis (EAN) in C57BL/6 mice. We show that functional assessments such as running capacity provide a more sensitive method for detecting alterations in disease severity than a classical clinical scoring paradigm. We performed detailed ultrastructural analysis and show for the first time that tomaculous neuropathy is a neuropathological feature of this disease model. In addition, we demonstrate that ultrastructural assessments of myelin pathology are sufficiently sensitive to detect significant differences in both mean G-ratio and mean axon diameter between mice with EAN induced with different doses of pertussis toxin. In summary, we have established a comprehensive assessment paradigm for discriminating variations in disease severity and the extent of myelin pathology in this model. Our findings indicate that this model is a powerful tool to study the pathogenesis of human peripheral demyelinating neuropathies and that this assessment paradigm could be used to determine the efficacy of potential therapies that aim to promote myelin repair and protect against nerve damage in autoimmune neuritides.


Asunto(s)
Marcha/fisiología , Neuritis Autoinmune Experimental/patología , Neuritis Autoinmune Experimental/fisiopatología , Fragmentos de Péptidos/toxicidad , Recuperación de la Función/fisiología , Carrera/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neuritis Autoinmune Experimental/inducido químicamente
7.
Exp Neurol ; 280: 106-14, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27063582

RESUMEN

Guillain-Barre' syndrome (GBS) is an acute, postinfectious, immune-mediated, demyelinating disease of peripheral nerves and nerve roots. As a classical animal model of GBS, experimental autoimmune neuritis (EAN) has become well-accepted. Additionally, the potent immune modulation exerted by mammalian target of rapamycin (mTOR) inhibitors has been used to treat cancers and showed beneficial effects. Here we demonstrate that the mTOR inhibitor RAD001 (everolimus) protected rats from the symptoms of EAN, as shown by decreased paralysis, diminished inflammatory cell infiltration, reductions in demyelination of peripheral nerves and improved nerve conduction. Furthermore, RAD001 shifted macrophage polarization toward the protective M2 phenotype and modified the inflammatory milieu by downregulating the production of pro-inflammatory cytokines including IFN-γ and IL-17as well as upregulating the release of anti-inflammatory cytokines such as IL-4 and TGF-ß. Amounts of the mTOR downstream targets p-P70S6K and p-4E-BP1 in sciatic nerves decreased, whereas the level of its upstream protein p-Akt was elevated. This demonstrated that RAD001 inhibited the mTOR pathway and encouraged the expression of p-Akt, which led to M2 macrophage polarization, thus improved the outcome of EAN in rats. Consequently, RAD001 exhibits strong potential as a therapeutic strategy for ameliorating peripheral poly-neuropathy.


Asunto(s)
Everolimus/uso terapéutico , Inmunosupresores/uso terapéutico , Neuritis Autoinmune Experimental/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Electromiografía , Everolimus/farmacología , Adyuvante de Freund/toxicidad , Inmunosupresores/farmacología , Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Conducción Nerviosa/efectos de los fármacos , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/metabolismo , Ratas , Ratas Endogámicas Lew , Factores de Tiempo
8.
ASN Neuro ; 7(4)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26186926

RESUMEN

A short-term exposure to moderately intense physical exercise affords a novel measure of protection against autoimmune-mediated peripheral nerve injury. Here, we investigated the mechanism by which forced exercise attenuates the development and progression of experimental autoimmune neuritis (EAN), an established animal model of Guillain-Barré syndrome. Adult male Lewis rats remained sedentary (control) or were preconditioned with forced exercise (1.2 km/day × 3 weeks) prior to P2-antigen induction of EAN. Sedentary rats developed a monophasic course of EAN beginning on postimmunization day 12.3 ± 0.2 and reaching peak severity on day 17.0 ± 0.3 (N = 12). By comparison, forced-exercise preconditioned rats exhibited a similar monophasic course but with significant (p < .05) reduction of disease severity. Analysis of popliteal lymph nodes revealed a protective effect of exercise preconditioning on leukocyte composition and egress. Compared with sedentary controls, forced exercise preconditioning promoted a sustained twofold retention of P2-antigen responsive leukocytes. The percentage distribution of pro-inflammatory (Th1) lymphocytes retained in the nodes from sedentary EAN rats (5.1 ± 0.9%) was significantly greater than that present in nodes from forced-exercise preconditioned EAN rats (2.9 ± 0.6%) or from adjuvant controls (2.0 ± 0.3%). In contrast, the percentage of anti-inflammatory (Th2) lymphocytes (7-10%) and that of cytotoxic T lymphocytes (∼20%) remained unaltered by forced exercise preconditioning. These data do not support an exercise-inducible shift in Th1:Th2 cell bias. Rather, preconditioning with forced exercise elicits a sustained attenuation of EAN severity, in part, by altering the composition and egress of autoreactive proinflammatory (Th1) lymphocytes from draining lymph nodes.


Asunto(s)
Ganglios Linfáticos/patología , Neuritis Autoinmune Experimental/patología , Neuritis Autoinmune Experimental/prevención & control , Condicionamiento Físico Animal/métodos , Células TH1/fisiología , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Citocinas , Modelos Animales de Enfermedad , Citometría de Flujo , Leucocitos/patología , Masculino , Proteína P2 de Mielina/química , Proteína P2 de Mielina/toxicidad , Neuritis Autoinmune Experimental/inducido químicamente , Ratas , Ratas Endogámicas Lew
9.
Exp Neurol ; 271: 279-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26033473

RESUMEN

Multiple sclerosis (MS) is classically defined by motor deficits, but it is also associated with the secondary symptoms of pain, depression, and anxiety. Up to this point modifying these secondary symptoms has been difficult. There is evidence that both MS and the animal model experimental autoimmune encephalomyelitis (EAE), commonly used to study the pathophysiology of the disease, can be modulated by exercise. To examine whether limited voluntary wheel running could modulate EAE disease progression and the co-morbid symptoms of pain, mice with EAE were allowed access to running wheels for 1h every day. Allowing only 1h every day of voluntary running led to a significant delay in the onset of clinical signs of the disease. The development of mechanical allodynia was assessed using Von Frey hairs and indicated that wheel running had a modest positive effect on the pain hypersensitivity associated with EAE. These behavioral changes were associated with reduced numbers of cFOS and phosphorylated NR1 positive cells in the dorsal horn of the spinal cord compared to no-run EAE controls. In addition, within the dorsal horn, voluntary wheel running reduced the number of infiltrating CD3(+) T-cells and reduced the overall levels of Iba1 immunoreactivity. Using high performance liquid chromatography (HPLC), we observed that wheel-running lead to significant changes in the spinal cord levels of the antioxidant glutathione. Oxidative stress has separately been shown to contribute to EAE disease progression and neuropathic pain. Together these results indicate that in mice with EAE, voluntary motor activity can delay the onset of clinical signs and reduce pain symptoms associated with the disease.


Asunto(s)
Terapia por Ejercicio/métodos , Neuralgia/etiología , Neuralgia/rehabilitación , Neuritis Autoinmune Experimental/complicaciones , Umbral del Dolor/fisiología , Carrera/fisiología , Animales , Antígenos CD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Hiperalgesia/fisiopatología , Hiperalgesia/rehabilitación , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Actividad Motora/fisiología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/rehabilitación , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Estadísticas no Paramétricas
10.
J Neuroimmunol ; 277(1-2): 13-7, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25262157

RESUMEN

Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy are autoimmune disorders of the peripheral nervous system in which autoantibodies are implicated in the disease pathogenesis. Recent work has focused on the nodal regions of the myelinated axon as potential autoantibody targets. Here we screened patient sera for autoantibodies to neurofascin and assessed the pathophysiological relevance of anti-neurofascin antibodies in vivo. Levels of anti-neurofascin antibodies were higher in sera from patients with Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy when compared with those of controls. Anti-neurofascin antibodies exacerbated and prolonged adoptive transfer experimental autoimmune neuritis and caused conduction defects when injected intraneurally.


Asunto(s)
Autoanticuerpos/sangre , Moléculas de Adhesión Celular/inmunología , Síndrome de Guillain-Barré/sangre , Inmunización Pasiva/efectos adversos , Factores de Crecimiento Nervioso/inmunología , Neuritis Autoinmune Experimental/inducido químicamente , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/sangre , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Gangliósidos/inmunología , Humanos , Masculino , Glicoproteína Mielina-Oligodendrócito/toxicidad , Conducción Nerviosa/efectos de los fármacos , Neuritis Autoinmune Experimental/patología , Neurofibromina 1/inmunología , Ratas , Ratas Endogámicas Lew , Nervio Ciático/efectos de los fármacos , Nervio Ciático/fisiopatología , Estadísticas no Paramétricas , Linfocitos T/inmunología , Linfocitos T/patología , Factores de Tiempo
11.
J Neuropathol Exp Neurol ; 73(6): 507-18, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24806299

RESUMEN

Optic neuritis is a common clinical manifestation of the chronic inflammatory CNS disease multiple sclerosis that can result in persistent visual impairment caused by degeneration of optic nerve axons and apoptosis of retinal ganglion cells (RGCs). Using a model of experimental autoimmune encephalomyelitis with optic neuritis (Brown Norway rats), we show that administration of the N-methyl-D-aspartate (NMDA) receptor antagonists memantine or MK801 results in RGC protection, axon protection, and reduced demyelination of optic nerves. Calcium imaging revealed that RGC responses to glutamate stimulation predominantly occurred via NMDA receptors and were inhibited by memantine in a dose-dependent manner. In contrast, oligodendrocytes were mainly responsive through the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainate receptor. This suggests that NMDA receptor blockade protected RGCs directly and that the protection was independent of effects on oligodendrocytes. Moreover, increased RGC survival was observed before the onset of optic nerve demyelination--when RGC degeneration had already started. These results indicate an important pathophysiologic role for NMDA receptor-mediated glutamate toxicity during the induction phase of this disease model and highlight a potential target for therapeutic neuroprotection in human optic neuritis.


Asunto(s)
Maleato de Dizocilpina/uso terapéutico , Memantina/uso terapéutico , Neuritis Autoinmune Experimental/complicaciones , Fármacos Neuroprotectores/uso terapéutico , Neuritis Óptica/complicaciones , Neuritis Óptica/tratamiento farmacológico , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Citocinas/sangre , Femenino , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Neuritis Autoinmune Experimental/inducido químicamente , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Nervio Óptico/patología , Neuritis Óptica/patología , Ratas , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Estilbamidinas
12.
PLoS One ; 9(3): e90942, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24603865

RESUMEN

Experimental autoimmune neuritis (EAN) is an autoantigen-specific T-cell-mediated disease model for human demyelinating inflammatory disease of the peripheral nervous system. Erythropoietin (EPO) has been known to promote EAN recovery but its haematopoiesis stimulating effects may limit its clinic application. Here we investigated the effects and potential mechanisms of an EPO-derived nonerythropoietic peptide, ARA 290, in EAN. Exogenous ARA 290 intervention greatly improved EAN recovery, improved nerve regeneration and remyelination, and suppressed nerve inflammation. Furthermore, haematopoiesis was not induced by ARA 290 during EAN treatment. ARA 290 intervention suppressed lymphocyte proliferation and altered helper T cell differentiation by inducing increase of Foxp3+/CD4+ regulatory T cells and IL-4+/CD4+ Th2 cells and decrease of IFN-γ+/CD4+ Th1 cells in EAN. In addition, ARA 290 inhibited inflammatory macrophage activation and promoted its phagocytic activity. In vitro, ARA 290 was shown to promote Schwann cell proliferation and inhibit its inflammatory activation. In summary, our data demonstrated that ARA 290 could effectively suppress EAN by attenuating inflammation and exerting direct cell protection, indicating that ARA 290 could be a potent candidate for treatment of autoimmune neuropathies.


Asunto(s)
Eritropoyetina/química , Regeneración Nerviosa/efectos de los fármacos , Neuritis Autoinmune Experimental/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Masculino , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/inmunología , Neuritis Autoinmune Experimental/patología , Neuropéptidos/efectos adversos , Fármacos Neuroprotectores/síntesis química , Oligopéptidos/síntesis química , Ratas , Ratas Endogámicas Lew , Nervio Ciático/efectos de los fármacos , Nervio Ciático/inmunología , Nervio Ciático/patología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Balance Th1 - Th2/efectos de los fármacos
13.
Immunol Res ; 56(2-3): 334-40, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23579773

RESUMEN

Experimental autoimmune neuritis (EAN) serves as an animal model for human Gullain-Barre syndrome (GBS), an autoimmune disease causing demyelination and inflammation of peripheral nerves. Macrophages, which play a major role in this autoimmune inflammatory process, can be selectively targeted by high doses of bisphophonates. The goal of this study was to examine the effect of the bisphosphonate, clodronate, on the severity of the EAN model. EAN was induced in female adult rats by immunization with bovine peripheral myelin. A number of treatment protocols with clodronate were used based on the common dosage regimen of 20 mg/kg in humans starting with the appearance of clinical signs on day 10 post-immunization. The clinical parameters measured included a clinical score, a motor performance test performed on a Rotarod and body weight. The expression of the matrix metaloprotease (MMP-9) in the sciatic nerves was measured as a marker of inflammatory macrophages. Treatment with clodronate, 20 mg/kg daily and 40 mg/kg every 2 days, significantly reduced the disease severity (a 75% decrease in severity, p < 0.01 by ANOVA) as measured by the clinical score compared to controls. Performance on the Rotarod test and body weight confirmed the clinical score findings. MMP-9 expression levels were significantly lower in the sciatic nerves of clodronate-treated rats. The present findings support the efficiency of clodronate in inflammatory diseases of the peripheral nervous system. The mechanism of action includes inhibition of inflammatory macrophages. The results suggest the use of bisphosphonates be considered in humans with GBS.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Ácido Clodrónico/administración & dosificación , Enfermedades Desmielinizantes/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Neuritis Autoinmune Experimental/tratamiento farmacológico , Animales , Biomarcadores/metabolismo , Ácido Clodrónico/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Macrófagos/inmunología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Actividad Motora/efectos de los fármacos , Proteína P2 de Mielina/inmunología , Neuritis Autoinmune Experimental/inducido químicamente , Ratas , Ratas Endogámicas Lew , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Nervio Ciático/patología , Síndrome
14.
Neuropathol Appl Neurobiol ; 39(7): 772-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23452257

RESUMEN

AIM: The role of chemokines and their receptors, which regulate trafficking and homing of leucocytes to inflamed organs in human or murine autoimmune neuritis, has not yet been elucidated in detail, Therefore, the role of the chemokine receptors CXCR4 and CXCR7 and their ligand CXCL12 was studied in autoimmune-mediated inflammation of the peripheral nervous system. METHODS: CXCL12/CXCR4 and/or CXCL12/CXCR7 interactions were specifically inhibited by the compounds AMD3100 or CCX771, respectively, in experimental autoimmune neuritis (EAN) of C57BL/6J mice immunized with P0106-125 peptide. RESULTS: Disease activity was significantly suppressed by blocking CXCR7 while antagonization of CXCR4 enhanced disease activity. Enhanced disease activity was accompanied by significantly increased transcription of IFN-γ, IL-12 and TNF-α mRNA in regional lymph nodes and spleen as well as by increased serum levels of IFN-γ. Furthermore, by blocking CXCR4, expression of the cell adhesion molecules ICAM-1 and VCAM-1 was upregulated on vascular endothelial cells of the sciatic nerve, which coincided with significantly increased infiltration of the sciatic nerve by CD4+ T cells and macrophages. Remarkably, combined antagonization of both CXCR4 and CXCR7 significantly suppressed disease activity. This was accompanied by increased frequencies of activated and highly IFN-γ-expressing, P0106-125 -specific T cells in regional lymph nodes and spleen; however, these cells were unable to infiltrate the sciatic nerve. CONCLUSION: These data suggest differential and hierarchically ordered roles for CXCR4/CXCL12- vs. CXCR7/CXCL12-dependent effects during EAN: CXCR7/CXCL12 interaction is a gatekeeper for pathogenic cells, regardless of their CXCR4/CXCL12-dependent state of activation.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neuritis Autoinmune Experimental/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/inmunología , Propanolaminas/farmacología , Linfocitos T/citología , Linfocitos T/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Glycobiology ; 22(12): 1768-74, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22843673

RESUMEN

Immunization of rabbits with bovine brain gangliosides induced an experimental neuropathy, with clinical signs resembling Guillain-Barré syndrome. All the immunized animals developed immunoglobulin G immunoreactivity to GM1 ganglioside. In a few (4 of 27) animals, an additional anti-ganglioside antibody population showing an unusual binding behavior was detected. Enzyme-linked immunosorbent assay and thin-layer chromatography immunostaining analyses showed that the binding of these unusual antibodies required the presence of two co-localized gangliosides. Maximal interaction was observed to a mixture of GM1 and GD1b, but the antibodies also showed "density-dependent" binding to GD1b. The antibodies were purified by affinity chromatography and displayed the ability to target antigens in biological membranes (rat synaptosomes).


Asunto(s)
Gangliósido G(M1)/inmunología , Gangliósidos/inmunología , Inmunoglobulina G/inmunología , Animales , Química Encefálica , Bovinos , Síndrome de Guillain-Barré/inducido químicamente , Síndrome de Guillain-Barré/inmunología , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/inmunología , Conejos , Ratas
16.
Neurobiol Aging ; 33(12): 2942-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22440674

RESUMEN

Amyloid plaques associated with Alzheimer's disease (AD) induce inflammatory responses associated with activated microglia and reactive astrocytes, which exacerbate neurodegeneration through release of inflammatory cytokines, reactive oxygen species, and other factors. Inflammation contributes to neurodegeneration at later stages of AD, but it may also play a role in early disease pathogenesis. We found that before plaque deposition, amyloid precursor protein (APP)/presenilin 1 (PSEN1) transgenic mice (PSAPP mice), a well-characterized model of AD, exhibit evidence of cerebrovascular inflammation. Expression of the endothelial cell-specific antigen MECA-32 (mouse endothelial cell antigen-32) was upregulated in the cerebrovasculature of young PSAPP mice (3 months old) and was similar to that observed in mice with experimental autoimmune encephalomyelitis, a model of multiple sclerosis characterized by neuroinflammation. MECA-32 is normally expressed in central and peripheral vasculature throughout development, but expression in the cerebrovasculature is downregulated on establishment of the blood-brain barrier (BBB). However, CNS inflammation triggers re-expression of MECA-32 in compromised cerebrovasculature. Our study indicates that MECA-32 may be a robust marker of cerebrovascular inflammation and compromised BBB integrity, triggered by soluble amyloid-ß early in disease pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Vasculitis del Sistema Nervioso Central/etiología , Vasculitis del Sistema Nervioso Central/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Antígenos de Superficie/metabolismo , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/metabolismo , Presenilina-1/genética , Regulación hacia Arriba/fisiología , Vasculitis del Sistema Nervioso Central/patología
17.
Immunol Invest ; 41(2): 171-82, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21864114

RESUMEN

Susceptible-strain animals immunized with P2 peptide could generate the disease of experimental autoimmune neuritis (EAN) with inflammation and demyelination of peripheral nerve. A myriad of transcription factors and inflammatory cytokines have been found to participate in this process; however, the roles of toll-like receptors (TLRs) are poorly understood in EAN. The aim of this study is to explore the role of TLR9 in the pathogenesis of EAN. The EAN was induced in Lewis rat by immunization with P2(53-78) and complete Freund's adjuvant. CpG oligodeoxynucleotides (ODN) (cODN), a suppressive ODN (sODN) and a control non-specific ODN (nODN) were respectively administered to explore the role of TLR9 in EAN both in vivo and vitro. Following immunization up to the peak phase of EAN, EAN rats inoculated with sODN had remarkably better clinical score of EAN and expressed a significantly inhibited TLR9 signaling pathway. Our study suggests that TLR9 may be involved in the pathogenesis of EAN.


Asunto(s)
Síndrome de Guillain-Barré/inmunología , Neuritis Autoinmune Experimental/inmunología , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Receptor Toll-Like 9/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Adyuvante de Freund/administración & dosificación , Síndrome de Guillain-Barré/terapia , Humanos , Inmunización , Masculino , Proteína P2 de Mielina/administración & dosificación , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/genética , Neuritis Autoinmune Experimental/fisiopatología , Neuritis Autoinmune Experimental/terapia , Oligodesoxirribonucleótidos Antisentido/efectos adversos , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Endogámicas Lew , Receptor Toll-Like 9/genética
18.
J Neurol Sci ; 306(1-2): 115-20, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21501850

RESUMEN

We studied the effects of fasudil, a selective Rho-kinase inhibitor, on experimental autoimmune neuritis (EAN). Continuous parenteral administration of fasudil prevented the development of EAN induced by P0 peptide 180-199 in Lewis rats while it also reduced EAN severity when administered after disease onset. Immunohistochemical examination disclosed a marked decrease in the amount of inflammatory cell infiltration and attenuation of demyelination and axonal degeneration. Specific proliferation of lymphocytes from fasudil-treated rats in response to P0 peptide was significantly reduced as compared with those from phosphate-buffered saline (PBS)-treated rats. Fasudil treatment was associated with a significant reduction in secretion of IFN-γ; by contrast, secretion of IL-4 was almost the same in the fasudil- and PBS-treated groups. As a result, the IFN-γ/IL-4 ratio in the supernatant was significantly deceased in fasudil-treated rats compared with PBS-treated ones. Therefore, our results indicate a beneficial effect of selective blockade of Rho-kinase in animals with autoimmune inflammation of the peripheral nerves, and may provide a rationale for the selective blockade of Rho-kinase as a new therapy for Guillain-Barré syndrome.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Neuritis Autoinmune Experimental/tratamiento farmacológico , Neuritis Autoinmune Experimental/prevención & control , Inhibidores de Proteínas Quinasas/administración & dosificación , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/fisiología , Proteínas Portadoras/metabolismo , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Sistemas de Liberación de Medicamentos , Ganglios Linfáticos/metabolismo , Masculino , Proteína P0 de la Mielina/efectos adversos , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/patología , Infiltración Neutrófila/efectos de los fármacos , Fosfoproteínas/metabolismo , Ratas , Ratas Endogámicas Lew , Intercambiadores de Sodio-Hidrógeno , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
19.
Brain Res ; 1333: 82-90, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20361946

RESUMEN

The glycoprotein erythropoietin (EPO) is a multifunctional cytokine involved in erythropoiesis. Recent data have suggested that EPO and EPO receptors are expressed in the central nervous system, where EPO exerts neuroprotective effects. However, peripheral nervous system (PNS) EPO and EPO receptor expression has not been widely studied. EPO and EPO receptor expression was examined in the PNS in an experimental autoimmune neuritis (EAN) rat model in the present study to elucidate EPO/EPO-receptor binding pathway involvement in injured PNS tissue. Western blot analysis demonstrated that EPO was significantly increased in the PNS at the paralytic stage on day 14 post-immunization (PI); levels were significantly decreased at day 30 PI. EPO was identified in PNS-derived vascular endothelial cells, Schwann cells, and axons in normal control rats. Most inflammatory cells in EAN lesions were EPO immunopositive at day 14 PI. In addition, the intensity of EPO immunoreactivity in both Schwann and vascular endothelial cells was greater than that of normal controls at this stage; intensity declined at day 30 PI. These findings suggest that EPO is transiently upregulated in EAN lesions and that the EPO/EPO-receptor binding pathway is associated with neuroprotection in EAN-affected PNS tissues.


Asunto(s)
Eritropoyetina/metabolismo , Neuritis Autoinmune Experimental/patología , Sistema Nervioso Periférico/metabolismo , Regulación hacia Arriba/fisiología , Animales , Modelos Animales de Enfermedad , Ecdisona/metabolismo , Femenino , Proteína P2 de Mielina/inmunología , Neuritis Autoinmune Experimental/inducido químicamente , Neuroglía/metabolismo , Neuronas/metabolismo , Sistema Nervioso Periférico/patología , Ratas , Ratas Endogámicas Lew , Receptores de Eritropoyetina/metabolismo , Proteínas S100/metabolismo , Factores de Tiempo , Regulación hacia Arriba/inmunología
20.
J Neurosci Res ; 88(8): 1651-63, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20077429

RESUMEN

Campylobacteriosis is a frequent antecedent event in Guillain-Barré syndrome (GBS), inducing high-titer serum antibodies for ganglioside antigens in the peripheral nervous system (PNS). Molecular mimicry between the lipooligosaccharide (LOS) component of Campylobacter jejuni and human peripheral nerve gangliosides is believed to play an important role in the pathogenesis of GBS. Conventional treatment strategies for patients with GBS include plasmapheresis, intravenous immunoglobulin (IVIG), and immunosuppression, which are invasive or relatively ineffective. In this study, we used our animal model of GBS, in which Lewis rats were immunized with GD3-like LOS isolated from C.jejuni. The animals developed anti-GD3 ganglioside antibodies and manifested neuromuscular dysfunction. To develop novel therapeutic strategies, we treated the animals by intraperitoneal administration of an anti-GD3 antiidiotype monoclonal antibody (BEC2) that specifically interacts with the pathogenic antibody. The treated animals had a remarkable reduction of anti-GD3 antibody titers and improvement of motor nerve functions. The results suggest that ganglioside mimics, such as antiidiotype antibodies, may be powerful reagents for therapeutic intervention in GBS by neutralizing specific pathogenic antiganglioside antibodies.


Asunto(s)
Anticuerpos Antiidiotipos/uso terapéutico , Gangliósidos/inmunología , Neuritis Autoinmune Experimental/terapia , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Biotinilación/métodos , Campylobacter jejuni/inmunología , Técnicas de Cocultivo/métodos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Canales de Potasio Éter-A-Go-Go/inmunología , Canales de Potasio Éter-A-Go-Go/metabolismo , Canales de Potasio Éter-A-Go-Go/farmacocinética , Femenino , Adyuvante de Freund/inmunología , Lipopolisacáridos , Neuronas Motoras/patología , Neuronas Motoras/ultraestructura , Músculo Esquelético/fisiología , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacocinética , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/complicaciones , Neuritis Autoinmune Experimental/inmunología , Enfermedades de la Unión Neuromuscular/tratamiento farmacológico , Enfermedades de la Unión Neuromuscular/etiología , Enfermedades de la Unión Neuromuscular/metabolismo , Técnicas de Cultivo de Órganos , Ratas , Prueba de Desempeño de Rotación con Aceleración Constante/métodos , Nervio Ciático/patología , Nervio Ciático/ultraestructura , Médula Espinal/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA