Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 119: 381-393, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38604270

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. Recent evidence suggests that lymphocyte trafficking in the intestines could play a key role in its etiology. Nevertheless, it is not clear how intestinal tissue is involved in the disease onset nor its evolution. In the present study, we aimed to evaluate intestinal inflammation dynamic throughout the disease course and its potential impact on disease progression. METHODS: We used tissue immunophenotyping (immunohistofluorescence and flow cytometry) and a recently described molecular magnetic resonance imaging (MRI) method targeting mucosal addressin cell adhesion molecule-1 (MAdCAM-1) to assess intestinal inflammation in vivo in two distinct animal models of MS (Experimental Autoimmune Encephalomyelitis - EAE) at several time points of disease progression. RESULTS: We report a positive correlation between disease severity and MAdCAM-1 MRI signal in two EAE models. Moreover, high MAdCAM-1 MRI signal during the asymptomatic phase is associated with a delayed disease onset in progressive EAE and to a lower risk of conversion to a secondary-progressive form in relapsing-remitting EAE. During disease evolution, in line with a bi-directional immune communication between the gut and the central nervous system, we observed a decrease in T-CD4+ and B lymphocytes in the ileum concomitantly with their increase in the spinal cord. CONCLUSION: Altogether, these data unveil a crosstalk between intestinal and central inflammation in EAE and support the use of molecular MRI of intestinal MAdCAM-1 as a new biomarker for prognostic in MS patients.


Asunto(s)
Biomarcadores , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Mucoproteínas , Esclerosis Múltiple , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/diagnóstico por imagen , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Imagen por Resonancia Magnética/métodos , Ratones , Biomarcadores/metabolismo , Mucoproteínas/metabolismo , Femenino , Pronóstico , Progresión de la Enfermedad , Moléculas de Adhesión Celular/metabolismo , Intestinos/diagnóstico por imagen , Intestinos/patología , Inmunoglobulinas/metabolismo , Inflamación/metabolismo , Inflamación/diagnóstico por imagen , Mucosa Intestinal/metabolismo , Mucosa Intestinal/diagnóstico por imagen
2.
Sante Publique ; 35(HS2): 107-109, 2024.
Artículo en Francés | MEDLINE | ID: mdl-38360765

RESUMEN

The Groupe de réflexion avec les associations de malades (GRAM)—a joint scientist/association think tank at the French National Institute of Health and Medical Research (INSERM)—has reflected, on the basis of internal surveys and the experiences of its members, on focus points concerning good practice to help partners involved in participatory research projects. In this article, we list these focus points and highlight the need to afford partners opportunities for dialogue, in order to give good chances of success to participatory research projects, in both the interest of associations and researchers.


Asunto(s)
Investigación Participativa Basada en la Comunidad , Humanos , Encuestas y Cuestionarios
3.
Cell Mol Life Sci ; 79(6): 323, 2022 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-35633384

RESUMEN

BACKGROUND: In multiple sclerosis (MS), disturbance of the plasminogen activation system (PAS) and blood brain barrier (BBB) disruption are physiopathological processes that might lead to an abnormal fibrin(ogen) extravasation into the parenchyma. Fibrin(ogen) deposits, usually degraded by the PAS, promote an autoimmune response and subsequent demyelination. However, the PAS disruption is not well understood and not fully characterized in this disorder. METHODS: Here, we characterized the expression of PAS actors during different stages of two mouse models of MS (experimental autoimmune encephalomyelitis-EAE), in the central nervous system (CNS) by quantitative RT-PCR, immunohistofluorescence and fluorescent in situ hybridization (FISH). Thanks to constitutive PAI-1 knockout mice (PAI-1 KO) and an immunotherapy using a blocking PAI-1 antibody, we evaluated the role of PAI-1 in EAE models and its impact on physiopathological processes such as fibrin(ogen) deposits, lymphocyte infiltration and demyelination. RESULTS: We report a striking overexpression of PAI-1 in reactive astrocytes during symptomatic phases, in two EAE mouse models of MS. This increase is concomitant with lymphocyte infiltration and fibrin(ogen) deposits in CNS parenchyma. By genetic invalidation of PAI-1 in mice and immunotherapy using a blocking PAI-1 antibody, we demonstrate that abolition of PAI-1 reduces the severity of EAE and occurrence of relapses in two EAE models. These benefits are correlated with a decrease in fibrin(ogen) deposits, infiltration of T4 lymphocytes, reactive astrogliosis, demyelination and axonal damage. CONCLUSION: These results demonstrate that a deleterious overexpression of PAI-1 by reactive astrocytes leads to intra-parenchymal dysfibrinolysis in MS models and anti-PAI-1 strategies could be a new therapeutic perspective for MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Inhibidor 1 de Activador Plasminogénico , Animales , Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Fibrina , Hibridación Fluorescente in Situ , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Inhibidor 1 de Activador Plasminogénico/genética , Serpina E2
4.
Cell Immunol ; 371: 104451, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34781155

RESUMEN

The COVID-19 pandemic has once again brought to the forefront the existence of a tight link between the coagulation/fibrinolytic system and the immunologic processes. Tissue-type plasminogen activator (tPA) is a serine protease with a key role in fibrinolysis by converting plasminogen into plasmin that can finally degrade fibrin clots. tPA is released in the blood by endothelial cells and hepatocytes but is also produced by various types of immune cells including T cells and monocytes. Beyond its role on hemostasis, tPA is also a potent modulator of inflammation and is involved in the regulation of several inflammatory diseases. Here, after a brief description of tPA structure, we review its new functions in adaptive immunity focusing on T cells and antigen presenting cells. We intend to synthesize the recent knowledge on proteolysis- and receptor-mediated effects of tPA on immune response in physiological and pathological context.


Asunto(s)
Coagulación Sanguínea/inmunología , COVID-19/inmunología , Fibrinólisis/inmunología , Inmunidad/inmunología , SARS-CoV-2/inmunología , Activador de Tejido Plasminógeno/inmunología , Células Presentadoras de Antígenos/inmunología , COVID-19/epidemiología , COVID-19/virología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Modelos Inmunológicos , Pandemias , SARS-CoV-2/fisiología , Linfocitos T/inmunología , Activador de Tejido Plasminógeno/metabolismo
5.
J Neurosci ; 40(8): 1778-1787, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31953371

RESUMEN

Increase in blood-brain barrier (BBB) permeability is a crucial step in neuroinflammatory processes. We previously showed that N Methyl D Aspartate Receptor (NMDARs), expressed on cerebral endothelial cells forming the BBB, regulate immune cell infiltration across this barrier in the mouse. Here, we describe the mechanism responsible for the action of NMDARs on BBB permeabilization. We report that mouse CNS endothelial NMDARs display the regulatory GluN3A subunit. This composition confers to NMDARs' unconventional properties: these receptors do not induce Ca2+ influx but rather show nonionotropic properties. In inflammatory conditions, costimulation of human brain endothelial cells by NMDA agonists (NMDA or glycine) and the serine protease tissue plasminogen activator, previously shown to potentiate NMDAR activity, induces metabotropic signaling via the Rho/ROCK pathway. This pathway leads to an increase in permeability via phosphorylation of myosin light chain and subsequent shrinkage of human brain endothelial cells. Together, these data draw a link between NMDARs and the cytoskeleton in brain endothelial cells that regulates BBB permeability in inflammatory conditions.SIGNIFICANCE STATEMENT The authors describe how NMDARs expressed on endothelial cells regulate blood-brain barrier function via myosin light chain phosphorylation and increase in permeability. They report that these non-neuronal NMDARs display distinct structural, functional, and pharmacological features than their neuronal counterparts.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Miosinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Células Endoteliales/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Masculino , Ratones , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Permeabilidad , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Activador de Tejido Plasminógeno/farmacología , Factor de Necrosis Tumoral alfa/farmacología
6.
J Neuroinflammation ; 18(1): 52, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33610187

RESUMEN

BACKGROUND: Tissue plasminogen activator (tPA) is a serine protease involved in fibrinolysis. It is released by endothelial cells, but also expressed by neurons and glial cells in the central nervous system (CNS). Interestingly, this enzyme also contributes to pathological processes in the CNS such as neuroinflammation by activating microglia and increasing blood-brain barrier permeability. Nevertheless, its role in the control of adaptive and innate immune response remains poorly understood. METHODS: tPA effects on myeloid and lymphoid cell response were studied in vivo in the mouse model of multiple sclerosis experimental autoimmune encephalomyelitis and in vitro in splenocytes. RESULTS: tPA-/- animals exhibited less severe experimental autoimmune encephalomyelitis than their wild-type counterparts. This was accompanied by a reduction in both lymphoid and myeloid cell populations in the spinal cord parenchyma. In parallel, tPA increased T cell activation and proliferation, as well as cytokine production by a protease-dependent mechanism and via plasmin generation. In addition, tPA directly raised the expression of MHC-II and the co-stimulatory molecules CD80 and CD86 at the surface of dendritic cells and macrophages by a direct action dependent of the activation of epidermal growth factor receptor. CONCLUSIONS: Our study provides new insights into the mechanisms responsible for the harmful functions of tPA in multiple sclerosis and its animal models: tPA promotes the proliferation and activation of both lymphoid and myeloid populations by distinct, though complementary, mechanisms.


Asunto(s)
Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Activación de Linfocitos/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Activador de Tejido Plasminógeno/toxicidad , Animales , Femenino , Humanos , Activación de Linfocitos/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Activador de Tejido Plasminógeno/deficiencia
7.
Brain ; 143(10): 2957-2972, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32893288

RESUMEN

Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis is a neuropsychiatric disease characterized by an antibody-mediated autoimmune response against NMDAR. Recent studies have shown that anti-NMDAR antibodies are involved in the pathophysiology of the disease. However, the upstream immune and inflammatory processes responsible for this pathogenic response are still poorly understood. Here, we immunized mice against the region of NMDA receptor containing the N368/G369 amino acids, previously implicated in a pathogenic response. This paradigm induced encephalopathy characterized by blood-brain barrier opening, periventricular T2-MRI hyperintensities and IgG deposits into the brain parenchyma. Two weeks after immunization, mice developed clinical symptoms reminiscent of encephalitis: anxiety- and depressive-like behaviours, spatial memory impairment (without motor disorders) and increased sensitivity to seizures. This response occurred independently of overt T-cell recruitment. However, it was associated with B220+ (B cell) infiltration towards the ventricles, where they differentiated into CD138+ cells (plasmocytes). Interestingly, these B cells originated from peripheral lymphoid organs (spleen and cervical lymphoid nodes). Finally, blocking the B-cell response using a depleting cocktail of antibodies reduced the severity of symptoms in encephalitis mice. This study demonstrates that the B-cell response can lead to an autoimmune reaction against NMDAR that drives encephalitis-like behavioural impairments. It also provides a relevant platform for dissecting encephalitogenic mechanisms in an animal model, and enables the testing of therapeutic strategies targeting the immune system in anti-NMDAR encephalitis.


Asunto(s)
Autoanticuerpos/sangre , Linfocitos B/metabolismo , Encefalitis/sangre , Enfermedad de Hashimoto/sangre , Proteínas del Tejido Nervioso/toxicidad , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Encefalitis/inducido químicamente , Encefalitis/inmunología , Enfermedad de Hashimoto/inducido químicamente , Enfermedad de Hashimoto/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/inmunología , Receptores de N-Metil-D-Aspartato/inmunología
8.
Proc Natl Acad Sci U S A ; 114(23): 6116-6121, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28533365

RESUMEN

New strategies for detecting disease activity in multiple sclerosis are being investigated to ameliorate diagnosis and follow-up of patients. Today, although magnetic resonance imaging (MRI) is widely used to diagnose and monitor multiple sclerosis, no imaging tools exist to predict the evolution of disease and the efficacy of therapeutic strategies. Here, we show that molecular MRI targeting the endothelial adhesion molecule P-selectin unmasks the pathological events that take place in the spinal cord of mice subjected to chronic or relapsing experimental autoimmune encephalomyelitis. This approach provides a quantitative spatiotemporal follow-up of disease course in relation to clinical manifestations. Moreover, it predicts relapse in asymptomatic mice and remission in symptomatic animals. Future molecular MRI targeting P-selectin may be used to improve diagnosis, follow-up of treatment, and management of relapse/remission cycles in multiple sclerosis patients by providing information currently inaccessible through conventional MRI techniques.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/patología , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Encéfalo/patología , Medios de Contraste , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Selectina-P/metabolismo , Recurrencia , Médula Espinal/patología
9.
Biochim Biophys Acta ; 1862(3): 395-402, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26493446

RESUMEN

The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Asunto(s)
Barrera Hematoencefálica/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Inflamación/inmunología , Microglía/inmunología , Plasminógeno/inmunología , Animales , Barrera Hematoencefálica/patología , Enfermedades del Sistema Nervioso Central/patología , Fibrina/inmunología , Fibrinolisina/inmunología , Humanos , Inflamación/patología , Leucocitos/inmunología , Leucocitos/patología , Microglía/patología , Activador de Tejido Plasminógeno/inmunología
10.
Glia ; 65(12): 1961-1975, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28850711

RESUMEN

Myelination is a late developmental process regulated by a set of inhibitory and stimulatory factors, including extracellular matrix components. Accordingly, chondroitin sulfate proteoglycans (CSPGs) act as negative regulators of myelination processes. A disintegrin and metalloproteinase with thrombospondin motifs type 4 (ADAMTS-4) is an extracellular protease capable of degrading CSPGs. Although exogenous ADAMTS-4 has been proven to be beneficial in several models of central nervous system (CNS) injuries, the physiological functions of endogenous ADAMTS-4 remain poorly understood. We first used Adamts4/LacZ reporter mice to reveal that ADAMTS-4 is strongly expressed in the CNS, especially in the white matter, with a cellular profile restricted to mature oligodendrocytes. Interestingly, we evidenced an abnormal myelination in Adamts4-/- mice, characterized by a higher diameter of myelinated axons with a shifting g-ratio. Accordingly, lack of ADAMTS-4 is accompanied by motor deficits and disturbed nervous electrical activity. In conclusion, we demonstrate that ADAMTS-4 is a new marker of mature oligodendrocytes contributing to the myelination processes and thus to the control of motor capacities.


Asunto(s)
Proteína ADAMTS4/metabolismo , Trastornos del Movimiento/genética , Oligodendroglía/metabolismo , Proteína ADAMTS4/genética , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/metabolismo , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Cuerpo Calloso/ultraestructura , Modelos Animales de Enfermedad , Potenciales Evocados Somatosensoriales/genética , Potenciales Evocados Somatosensoriales/fisiología , Trastornos Neurológicos de la Marcha/etiología , Locomoción/genética , Locomoción/fisiología , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica , Trastornos del Movimiento/fisiopatología , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/patología , Oligodendroglía/ultraestructura , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estadísticas no Paramétricas , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
11.
Brain ; 139(Pt 9): 2406-19, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27435092

RESUMEN

Multiple sclerosis is among the most common causes of neurological disability in young adults. Here we provide the preclinical proof of concept of the benefit of a novel strategy of treatment for multiple sclerosis targeting neuroendothelial N-methyl-D-aspartate glutamate receptors. We designed a monoclonal antibody against N-methyl-D-aspartate receptors, which targets a regulatory site of the GluN1 subunit of N-methyl-D-aspartate receptor sensitive to the protease tissue plasminogen activator. This antibody reverted the effect of tissue plasminogen activator on N-methyl-D-aspartate receptor function without affecting basal N-methyl-D-aspartate receptor activity (n = 21, P < 0.01). This antibody bound N-methyl-D-aspartate receptors on the luminal surface of neurovascular endothelium in human tissues and in mouse, at the vicinity of tight junctions of the blood-spinal cord barrier. Noteworthy, it reduced human leucocyte transmigration in an in vitro model of the blood-brain barrier (n = 12, P < 0.05). When injected during the effector phase of MOG-induced experimental autoimmune encephalomyelitis (n = 24), it blocked the progression of neurological impairments, reducing cumulative clinical score (P < 0.001) and mean peak score (P < 0.001). This effect was observed in wild-type animals but not in tissue plasminogen activator knock-out animals (n = 10). This therapeutic effect was associated to a preservation of the blood-spinal cord barrier (n = 6, P < 0.001), leading to reduced leucocyte infiltration (n = 6, P < 0.001). Overall, this study unveils a critical function of endothelial N-methyl-D-aspartate receptor in multiple sclerosis, and highlights the therapeutic potential of strategies targeting the protease-regulated site of N-methyl-D-aspartate receptor.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas del Tejido Nervioso/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Activador de Tejido Plasminógeno/metabolismo , Animales , Células Endoteliales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
J Inflamm (Lond) ; 21(1): 4, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355547

RESUMEN

Tissue-plasminogen activator (tPA) is a serine protease well known for its fibrinolytic function. Recent studies indicate that tPA could also modulate inflammation via plasmin generation and/or by receptor mediated signalling in vitro. However, the contribution of tPA in inflammatory processes in vivo has not been fully addressed. Therefore, using tPA-deficient mice, we have analysed the effect of lipopolysaccharide (LPS) challenge on the phenotype of myeloid cells including neutrophils, macrophages and dendritic cells (DCs) in spleen. We found that LPS treatment upregulated the frequency of major histocompatibility class two (MHCII+) macrophages but also, paradoxically, induced a deep downregulation of MHCII molecule level on macrophages and on conventional dendritic cells 2 (cDC2). Expression level of the CD11b integrin, known as a tPA receptor, was upregulated by LPS on MHCII+ macrophages and cDC2, suggesting that tPA effects could be amplified during inflammation. In tPA-/- mice under inflammatory conditions, expression of costimulatory CD86 molecules on MHCII+ macrophages was decreased compared to WT mice, while in steady state the expression of MHCII molecules was higher on macrophages. Finally, we reported that tPA deficiency slightly modified the phenotype of DCs and T cells in acute inflammatory conditions. Overall, our findings indicate that in vivo, LPS injection had an unexpectedly bimodal effect on MHCII expression on macrophages and DCs that consequently might affect adaptive immunity. tPA could also participate in the regulation of the T cell response by modulating the levels of CD86 and MHCII molecules on macrophages.

13.
J Neurosci ; 32(15): 5186-99, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22496564

RESUMEN

Tissue-type plasminogen activator (tPA) regulates physiological processes in the brain, such as learning and memory, and plays a critical role in neuronal survival and neuroinflammation in pathological conditions. Here we demonstrate, by combining mouse in vitro and in vivo data, that tPA is an important element of the cross talk between neurons and astrocytes. The data show that tPA released by neurons is constitutively endocytosed by astrocytes via the low-density lipoprotein-related protein receptor, and is then exocytosed in a regulated manner. The exocytotic recycling of tPA by astrocytes is inhibited in the presence of extracellular glutamate. Kainate receptors of astrocytes act as sensors of extracellular glutamate and, via a signaling pathway involving protein kinase C, modulate the exocytosis of tPA. Further, by thus capturing extracellular tPA, astrocytes serve to reduce NMDA-mediated responses potentiated by tPA. Overall, this work provides the first demonstration that the neuromodulator, tPA, may also be considered as a gliotransmitter.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ácido Glutámico/farmacología , Ácido Glutámico/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Activador de Tejido Plasminógeno/metabolismo , Albúminas/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Clatrina/fisiología , Dinaminas/fisiología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Citometría de Flujo , Silenciador del Gen , Inmunohistoquímica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Plásmidos/genética , Proteína Quinasa C/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ácido Kaínico/efectos de los fármacos , Receptores de Ácido Kaínico/metabolismo , Receptores de LDL/metabolismo , Sinapsinas/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo , alfa-Macroglobulinas/metabolismo
14.
Glia ; 60(9): 1437-50, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22653796

RESUMEN

The endocannabinoid anandamide (AEA) is released by macrophages and microglia on pathological neuroinflammatory conditions such as multiple sclerosis (MS). CD200 is a membrane glycoprotein expressed in neurons that suppresses immune activity via its receptor (CD200R) mainly located in macrophages/microglia. CD200-CD200R interactions contribute to the brain immune privileged status. In this study, we show that AEA protects neurons from microglia-induced neurotoxicity via CD200-CD200R interaction. AEA increases the expression of CD200R1 in LPS/IFN-γ activated microglia through the activation of CB(2) receptors. The neuroprotective effect of AEA disappears when microglial cells derive from CD200R1(-/-) mice. We also show that engagement of CD200R1 by CD200Fc decreased the production of the proinflammatory cytokines IL-1ß and IL-6, but increased IL-10 in activated microglia. In the chronic phases of Theiler's virus-induced demyelinating disease (TMEV-IDD) the expression of CD200 and CD200R1 was reduced in the spinal cord. AEA-treated animals up-regulated the expression of CD200 and CD200R1, restoring levels found in sham animals together with increased expression of IL-10 and reduced expression of IL-1ß and IL-6. Treated animals also improved their motor behavior. Because AEA up-regulated the expression of CD200R1 in microglia, but failed to enhance CD200 in neurons we suggest that AEA-induced up-regulation of CD200 in TMEV-IDD is likely due to IL-10 as this cytokine increases CD200 in neurons. Our findings provide a new mechanism of action of AEA to limit immune response in the inflamed brain.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Superficie/metabolismo , Ácidos Araquidónicos/uso terapéutico , Encéfalo/metabolismo , Endocannabinoides/uso terapéutico , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Alcamidas Poliinsaturadas/uso terapéutico , Receptores de Superficie Celular/metabolismo , Animales , Ácidos Araquidónicos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Células Cultivadas , Endocannabinoides/farmacología , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Receptores de Orexina , Alcamidas Poliinsaturadas/farmacología
15.
Neuroimage ; 63(2): 760-70, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22813950

RESUMEN

Since endothelial cells can be targeted by large contrast-carrying particles, molecular imaging of cerebrovascular cell activation is highly promising to evaluate the underlying inflammation of the central nervous system (CNS). In this study, we aimed to demonstrate that molecular magnetic resonance imaging (MRI) of cerebrovascular cell activation can reveal CNS disorders in the absence of visible lesions and symptoms. To this aim, we optimized contrast carrying particles targeting vascular cell adhesion molecule-1 and MRI protocols through both in vitro and in vivo experiments. Although, pre-contrast MRI images failed to reveal the ongoing pathology, contrast-enhanced MRI revealed hypoperfusion-triggered CNS injury in vascular dementia, unmasked amyloid-induced cerebrovascular activation in Alzheimer's disease and allowed monitoring of disease activity during experimental autoimmune encephalomyelitis. Moreover, contrast-enhanced MRI revealed the cerebrovascular cell activation associated with known risk factors of CNS disorders such as peripheral inflammation, ethanol consumption, hyperglycemia and aging. By providing a dramatically higher sensitivity than previously reported methods and molecular contrast agents, the technology described in the present study opens new avenues of investigation in the field of neuroinflammation.


Asunto(s)
Enfermedades del Sistema Nervioso Central/diagnóstico , Células Endoteliales/metabolismo , Compuestos Férricos , Imagen por Resonancia Magnética/métodos , Imagen Molecular/métodos , Animales , Western Blotting , Inmunohistoquímica , Masculino , Nanopartículas del Metal , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Neuroinflammation ; 8: 102, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21851608

RESUMEN

BACKGROUND: VCAM-1 represents one of the most important adhesion molecule involved in the transmigration of blood leukocytes across the blood-brain barrier (BBB) that is an essential step in the pathogenesis of MS. Several evidences have suggested the potential therapeutic value of cannabinoids (CBs) in the treatment of MS and their experimental models. However, the effects of endocannabinoids on VCAM-1 regulation are poorly understood. In the present study we investigated the effects of anandamide (AEA) in the regulation of VCAM-1 expression induced by Theiler's virus (TMEV) infection of brain endothelial cells using in vitro and in vivo approaches. METHODS: i) in vitro: VCAM-1 was measured by ELISA in supernatants of brain endothelial cells infected with TMEV and subjected to AEA and/or cannabinoid receptors antagonist treatment. To evaluate the functional effect of VCAM-1 modulation we developed a blood brain barrier model based on a system of astrocytes and brain endothelial cells co-culture. ii) in vivo: CB(1) receptor deficient mice (Cnr1(-/-)) infected with TMEV were treated with the AEA uptake inhibitor UCM-707 for three days. VCAM-1 expression and microglial reactivity were evaluated by immunohistochemistry. RESULTS: Anandamide-induced inhibition of VCAM-1 expression in brain endothelial cell cultures was mediated by activation of CB(1) receptors. The study of leukocyte transmigration confirmed the functional relevance of VCAM-1 inhibition by AEA. In vivo approaches also showed that the inhibition of AEA uptake reduced the expression of brain VCAM-1 in response to TMEV infection. Although a decreased expression of VCAM-1 by UCM-707 was observed in both, wild type and CB(1) receptor deficient mice (Cnr1(-/-)), the magnitude of VCAM-1 inhibition was significantly higher in the wild type mice. Interestingly, Cnr1(-/-) mice showed enhanced microglial reactivity and VCAM-1 expression following TMEV infection, indicating that the lack of CB(1) receptor exacerbated neuroinflammation. CONCLUSIONS: Our results suggest that CB(1) receptor dependent VCAM-1 inhibition is a novel mechanism for AEA-reduced leukocyte transmigration and contribute to a better understanding of the mechanisms underlying the beneficial role of endocannabinoid system in the Theiler's virus model of MS.


Asunto(s)
Ácidos Araquidónicos/farmacología , Barrera Hematoencefálica/metabolismo , Células Endoteliales/efectos de los fármacos , Leucocitos/fisiología , Alcamidas Poliinsaturadas/farmacología , Receptor Cannabinoide CB1/metabolismo , Theilovirus/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/farmacología , Adhesión Celular/efectos de los fármacos , Endocannabinoides , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Humanos , Leucocitos/citología , Leucocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Microglía/metabolismo , Theilovirus/genética
17.
Brain Behav Immun ; 25(4): 736-49, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21310228

RESUMEN

Theiler's virus (TMEV) infection of the central nervous system (CNS) induces an immune-mediated demyelinating disease in susceptible mouse strains and serves as a relevant infection model for human multiple sclerosis (MS). The endocannabinoid system represents a novel therapeutic target for autoimmune and chronic inflammatory diseases due to its anti-inflammatory properties by regulating cytokine network. IL-12p70 and IL-23 are functionally related heterodimeric cytokines that play a crucial role in the pathogenesis of MS. In the present study we showed that the endocannabinoid anandamide (AEA) downregulated the gene expression of IL-12p70 and IL-23 forming subunits mRNAs in the spinal cord of TMEV-infected mice and ameliorated motor disturbances. This was accompanied by significant decreases on the serological levels of IL-12p70/IL-23 and more interestingly, of IL-17A. In contrast, serum levels of IL-10 resulted elevated. In addition, we studied the signalling pathways involved in the regulation of IL-12p70/IL-23 and IL-10 expression in TMEV-infected microglia and addressed the possible interactions of AEA with these pathways. AEA acted through the ERK1/2 and JNK pathways to downregulate IL-12p70 and IL-23 while upregulating IL-10. These effects were partially mediated by CB2 receptor activation. We also described an autocrine circuit of cross-talk between IL-12p70/IL-23 and IL-10, since endogenously produced IL-10 negatively regulates IL-12p70 and IL-23 cytokines in TMEV-infected microglia. This suggests that by altering the cytokine network, AEA could indirectly modify the type of immune responses within the CNS. Accordingly, pharmacological modulation of endocannabinoids might be a useful tool for treating neuroinflammatory diseases.


Asunto(s)
Ácidos Araquidónicos/farmacología , Moduladores de Receptores de Cannabinoides/farmacología , Infecciones por Cardiovirus/inmunología , Endocannabinoides , Interleucinas/inmunología , Microglía/inmunología , Enfermedad Autoinmune Experimental del Sistema Nervioso/inmunología , Alcamidas Poliinsaturadas/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Análisis de Varianza , Animales , Infecciones por Cardiovirus/tratamiento farmacológico , Infecciones por Cardiovirus/virología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-12/metabolismo , Interleucina-23/efectos de los fármacos , Interleucina-23/genética , Interleucina-23/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Ratones , Microglía/metabolismo , Microglía/virología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Enfermedad Autoinmune Experimental del Sistema Nervioso/metabolismo , Enfermedad Autoinmune Experimental del Sistema Nervioso/virología , Neuroinmunomodulación/efectos de los fármacos , Subunidades de Proteína , ARN Mensajero/análisis , Receptor Cross-Talk , Transducción de Señal , Estadísticas no Paramétricas , Theilovirus/inmunología
18.
J Thromb Haemost ; 19(9): 2235-2247, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34060720

RESUMEN

BACKGROUND: Factor XII (FXII) is a serine protease that participates in the intrinsic coagulation pathway. Several studies have shown that plasma FXII exerts a deleterious role in cerebral ischemia and traumatic brain injury by promoting thrombo-inflammation. Nevertheless, the impact of FXII on neuronal cell fate remains unknown. OBJECTIVES: We investigated the role of FXII and FXIIa in neuronal injury and apoptotic cell death. METHODS: We tested the neuroprotective roles of FXII and FXIIa in an experimental model of neuronal injury induced by stereotaxic intracerebral injection of N-methyl-D-aspartic acid (NMDA) in vivo and in a model of apoptotic death of murine primary neuronal cultures through serum deprivation in vitro. RESULTS: Here, we found that exogenous FXII and FXIIa reduce brain lesions induced by NMDA injection in vivo. Furthermore, FXII protects cultured neurons from apoptosis through a growth factor--like effect. This mechanism was triggered by direct interaction with epidermal growth factor (EGF) receptor and subsequent activation of this receptor. Interestingly, the "proteolytically" active and two-chain form of FXII, FXIIa, exerts its protective effects by an alternative signaling pathway. FXIIa activates the pro-form of hepatocyte growth factor (HGF) into HGF, which in turn activated the HGF receptor (HGFR) pathway. CONCLUSION: This study describes two novel mechanisms of action of FXII and identifies neurons as target cells for the protective effects of single and two-chain forms of FXII. Therefore, inhibition of FXII in neurological disorders may have deleterious effects by preventing its beneficial effects on neuronal survival.


Asunto(s)
Factor XII , Proteínas Proto-Oncogénicas c-met , Animales , Apoptosis , Coagulación Sanguínea , Factor XIIa , Ratones , Neuronas
19.
Glia ; 58(2): 135-47, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19565660

RESUMEN

The endocannabinoid system exhibits anti-inflammatory properties by regulating cytokine production. Anandamide (AEA) down-regulates proinflammatory cytokines in a viral model of multiple sclerosis (MS). However, little is known about the mechanisms by which AEA exerts these effects. Microglial cells are the main source of cytokines within the brain and the first barrier of defense against pathogens by acting as antigen presenting cells. IL-10 is a key physiological negative regulator of microglial activation. In this study we show that AEA enhances LPS/IFNgamma-induced IL-10 production in microglia by targeting CB(2) receptors through the activation of ERK1/2 and JNK MAPKs. AEA also inhibits NF-kappaB activation by interfering with the phosphorylation of IkappaBalpha, which may result in an increase of IL-10 production. Moreover, endogenously produced IL-10 negatively regulates IL-12 and IL-23 cytokines, which in its turn modify the pattern of expression of transcription factors involved in Th commitment of splenocytes. This suggests that by altering the cytokine network, AEA could indirectly modify the type of immune responses within the central nervous system (CNS). Accordingly, pharmacological modulation of AEA uptake and degradation might be a useful tool for treating neuroinflammatory diseases.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Interleucina-10/metabolismo , Microglía/enzimología , Microglía/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Receptor Cannabinoide CB2/metabolismo , Animales , Línea Celular , Células Cultivadas , Endocannabinoides , Proteínas I-kappa B/metabolismo , Interferón gamma/toxicidad , Interleucina-12/metabolismo , Interleucina-23/metabolismo , Lipopolisacáridos/toxicidad , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA