Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 119: 381-393, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38604270

RESUMO

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.

2.
Cell Mol Life Sci ; 79(6): 323, 2022 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-35633384

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Inibidor 1 de Ativador de Plasminogênio , Animais , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Fibrina , Hibridização in Situ Fluorescente , Camundongos , Camundongos Knockout , Esclerose Múltipla/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Serpina E2
3.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34948279

RESUMO

Tissue-type plasminogen activator (tPA) plays roles in the development and the plasticity of the nervous system. Here, we demonstrate in neurons, that by opposition to the single chain form (sc-tPA), the two-chains form of tPA (tc-tPA) activates the MET receptor, leading to the recruitment of N-Methyl-d-Aspartate receptors (NMDARs) and to the endocytosis and proteasome-dependent degradation of NMDARs containing the GluN2B subunit. Accordingly, tc-tPA down-regulated GluN2B-NMDAR-driven signalling, a process prevented by blockers of HGFR/MET and mimicked by its agonists, leading to a modulation of neuronal death. Thus, our present study unmasks a new mechanism of action of tPA, with its two-chains form mediating a crosstalk between MET and the GluN2B subunit of NMDARs to control neuronal survival.


Assuntos
Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feto , Camundongos , Cultura Primária de Células , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptor Cross-Talk/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Ativador de Plasminogênio Tecidual/fisiologia
4.
J Neuroinflammation ; 18(1): 52, 2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33610187

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/induzido quimicamente , Ativação Linfocitária/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/toxicidade , Animais , Feminino , Humanos , Ativação Linfocitária/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/metabolismo , Ativador de Plasminogênio Tecidual/deficiência
5.
Mult Scler J Exp Transl Clin ; 6(4): 2055217320959806, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33101703

RESUMO

BACKGROUND: Clinical observations support the hypothesis that stressful events increase relapse occurrence in multiple sclerosis patients, while stress-reduction strategies can modulate this effect. However, a direct cause-effect relationship between stress level and relapse cannot be firmly established from these data. OBJECTIVES: The purpose of this work was to address whether modulation of stress could interfere with symptom relapse in an animal model of multiple sclerosis with relapsing-remitting course. METHODS: Mice bred in standard or enriched environment were subjected to repeated acute stress during the remission phase of relapsing-remitting PLP-induced experimental autoimmune encephalomyelitis. RESULTS: We report that repeated acute stress induced a twofold increase in relapse incidence in experimental autoimmune encephalomyelitis. On the other hand, environmental enrichment reduced relapse incidence and severity, and reversed the effects of repeated acute stress. CONCLUSION: These data provide the platform for further studies on the biological processes that link stress and multiple sclerosis relapses in a suitable animal model.

6.
Brain ; 143(10): 2957-2972, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32893288

RESUMO

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.


Assuntos
Autoanticorpos/sangue , Linfócitos B/metabolismo , Encefalite/sangue , Doença de Hashimoto/sangue , Proteínas do Tecido Nervoso/toxicidade , Animais , Autoanticorpos/imunologia , Linfócitos B/imunologia , Encefalite/induzido quimicamente , Encefalite/imunologia , Doença de Hashimoto/induzido quimicamente , Doença de Hashimoto/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/imunologia , Receptores de N-Metil-D-Aspartato/imunologia
7.
J Neurosci ; 40(8): 1778-1787, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-31953371

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Miosinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Linhagem Celular , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Células Endoteliais/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Camundongos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Permeabilidade , Fosforilação/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tecidual/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
8.
Cereb Cortex ; 29(6): 2482-2498, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29878094

RESUMO

Modifications of neuronal migration during development, including processes that control cortical lamination are associated with functional deficits at adult stage. Here, we report for the first time that the lack of the serine protease tissue-type Plasminogen Activator (tPA), previously characterized as a neuromodulator and a gliotransmitter, leads to an altered cortical lamination in adult. This results in a neuronal migration defect of tPA deficient neurons which are stopped in the intermediate zone at E16. This phenotype is rescued by re-expressing a wild-type tPA in cortical neurons at E14 but not by a tPA that cannot interact with NMDAR. We thus hypothetized that the tPA produced by cortical neuronal progenitors can control their own radial migration through a mechanism dependent of NMDAR expressed at the surface of radial glial cells (RGC). Accordingly, conditional deletion of tPA in neuronal progenitors at E14 or overexpression of a dominant-negative NMDAR that cannot bind tPA in RGC also delayed neuronal migration. Moreover, the lack of tPA lead to an impaired maturation and orientation of RGC. These data provide the first demonstration that the neuronal serine protease tPA is an actor of a proper corticogenesis by its ability to control NMDAR signaling in RGC.


Assuntos
Córtex Cerebral/embriologia , Células Ependimogliais/metabolismo , Neurogênese/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Movimento Celular/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia
9.
Cell Death Differ ; 24(9): 1518-1529, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28644439

RESUMO

The unfolded protein response (UPR) is an endoplasmic reticulum (ER) -related stress conserved pathway that aims to protect cells from being overwhelmed. However, when prolonged, UPR activation converts to a death signal, which relies on its PERK-eIF2α branch. Overactivation of the UPR has been implicated in many neurological diseases, including cerebral ischaemia. Here, by using an in vivo thromboembolic model of stroke on transgenic ER stress-reporter mice and neuronal in vitro models of ischaemia, we demonstrate that ischaemic stress leads to the deleterious activation of the PERK branch of the UPR. Moreover, we show that the serine protease tissue-type plasminogen activator (tPA) can bind to cell surface Grp78 (78 kD glucose-regulated protein), leading to a decrease of the PERK pathway activation, thus a decrease of the deleterious factor CHOP, and finally promotes neuroprotection. Altogether, this work highlights a new role and a therapeutic potential of the chaperone protein Grp78 as a membrane receptor of tPA capable to prevent from ER stress overactivation.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Fibrinolíticos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tromboembolia/terapia , Ativador de Plasminogênio Tecidual/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
10.
Cell Discov ; 3: 17001, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28417010

RESUMO

Plasminogen activation is involved in many processes within the central nervous system, including synaptic plasticity, neuroinflammation and neurodegeneration. However, the mechanisms that regulate plasminogen activation in the brain still remain unknown. Here we demonstrate that astrocytes participate in this regulation by two mechanisms. First, the astrocyte plasma membrane serves as a surface for plasminogen activation by tissue-type plasminogen activator. This activation triggers downstream plasmin-dependent processes with important impacts in brain health and disease, such as fibrinolysis and brain-derived neurotrophic factor conversion. Second, astrocytes take up plasminogen and plasmin in a regulated manner through a novel mechanism involving endocytosis mediated by cell-surface actin and triggered by extracellular plasmin activity at the surface of astrocytes. Following endocytosis, plasminogen and plasmin are targeted to lysosomes for degradation. Thus, cell-surface actin acts as a sensor of plasmin activity to induce a negative feedback through plasmin endocytosis. This study provides evidence that astrocytes control the balance between plasmin formation and plasmin elimination in the brain parenchyma.

11.
Blood ; 128(20): 2423-2434, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27531677

RESUMO

Hyperfibrinolysis is a systemic condition occurring in various clinical disorders such as trauma, liver cirrhosis, and leukemia. Apart from increased bleeding tendency, the pathophysiological consequences of hyperfibrinolysis remain largely unknown. Our aim was to develop an experimental model of hyperfibrinolysis and to study its effects on the homeostasis of the blood-brain barrier (BBB). We induced a sustained hyperfibrinolytic state in mice by hydrodynamic transfection of a plasmid encoding for tissue-type plasminogen activator (tPA). As revealed by near-infrared fluorescence imaging, hyperfibrinolytic mice presented a significant increase in BBB permeability. Using a set of deletion variants of tPA and pharmacological approaches, we demonstrated that this effect was independent of N-methyl-D-aspartate receptor, low-density lipoprotein-related protein, protease-activated receptor-1, or matrix metalloproteinases. In contrast, we provide evidence that hyperfibrinolysis-induced BBB leakage is dependent on plasmin-mediated generation of bradykinin and subsequent activation of bradykinin B2 receptors. Accordingly, this effect was prevented by icatibant, a clinically available B2 receptor antagonist. In agreement with these preclinical data, bradykinin generation was also observed in humans in a context of acute pharmacological hyperfibrinolysis. Altogether, these results suggest that B2 receptor blockade may be a promising strategy to prevent the deleterious effects of hyperfibrinolysis on the homeostasis of the BBB.


Assuntos
Barreira Hematoencefálica/metabolismo , Bradicinina/fisiologia , Permeabilidade Capilar/fisiologia , Fibrinolisina/fisiologia , Fibrinólise/fisiologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Bradicinina/metabolismo , Antagonistas de Receptor B2 da Bradicinina/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/genética , Fibrinolisina/metabolismo , Fibrinólise/efeitos dos fármacos , Fibrinólise/genética , Hidrodinâmica , Camundongos , Camundongos Transgênicos , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo
12.
Brain ; 139(Pt 9): 2406-19, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27435092

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas do Tecido Nervoso/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Células Endoteliais , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Stroke ; 46(2): 477-84, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25503547

RESUMO

BACKGROUND AND PURPOSE: Despite the effectiveness of recombinant tissue-type plasminogen activator (r-tPA) during the acute phase of ischemic stroke, the therapy remains limited by a narrow time window and the occurrence of occasional vascular side effects, particularly symptomatic hemorrhages. Our aim was to investigate the mechanisms underlying the endothelial damage resulting from r-tPA treatment in ischemic-like conditions. METHODS: Microarray analyses were performed on cerebral endothelial cells submitted to r-tPA treatment during oxygen and glucose deprivation to identify novel biomarker candidates. Validation was then performed in vivo in a mouse model of thromboembolic stroke and culminated in an analysis in a clinical cohort of patients with ischemic stroke treated with thrombolysis. RESULTS: The transcription factor NURR1 (NR4A2) was identified as a downstream target induced by r-tPA during oxygen and glucose deprivation. Silencing NURR1 expression reversed the endothelial-toxicity induced by the combined stimuli, a protective effect attributable to reduced levels of proinflammatory mediators, such as nuclear factor-kappa-beta 2 (NF-κ-B2), interleukin 1 alpha (IL1α), intercellular adhesion molecule 1 (ICAM1), SMAD family member 3 (SMAD3), colony stimulating factor 2 (granulocyte-macrophage; CSF2). The detrimental effect of delayed thrombolysis, in conditions in which NURR1 gene expression was enhanced, was confirmed in the preclinical stroke model. Finally, we determined that patients with stroke who had a symptomatic hemorrhagic transformation after r-tPA treatment exhibited higher baseline serum NURR1 levels than did patients with an asymptomatic or absence of cerebral bleedings. CONCLUSIONS: Our results suggest that NURR1 upregulation by r-tPA during ischemic stroke is associated with endothelial dysfunction and inflammation and the enhancement of hemorrhagic complications associated to thrombolysis.


Assuntos
Isquemia Encefálica/sangue , Isquemia Encefálica/tratamento farmacológico , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/sangue , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Animais , Biomarcadores/sangue , Isquemia Encefálica/diagnóstico , Linhagem Celular , Feminino , Fibrinolíticos/efeitos adversos , Fibrinolíticos/uso terapêutico , Humanos , Inflamação/sangue , Inflamação/induzido quimicamente , Inflamação/diagnóstico , Masculino , Camundongos , Pessoa de Meia-Idade , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/biossíntese , Acidente Vascular Cerebral/diagnóstico , Ativador de Plasminogênio Tecidual/efeitos adversos , Resultado do Tratamento
14.
J Neuroimmunol ; 267(1-2): 86-91, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24393520

RESUMO

The pro-inflammatory cytokine IL-1ß is known to play a role in several models of aging, neuroinflammation, and neurodegenerative diseases. Here, we document a detailed time- and age-dependent pattern of pro- and anti-inflammatory biomarkers following bilateral intrahippocampal injection of interleukin-1ß. During the first 12h several pro- and anti-inflammatory cytokines increased in the aged (24 mo old) rats, some of which returned to baseline levels by 24h post-injection while others remained elevated for 72 h post-injection. In contrast, no such increases were observed in the young (3 mo old) rats. Interestingly, young rats up-regulated mRNA of two pro-inflammatory cytokines, interleukin-1ß and tumor necrosis factor-α, but did not translate these transcripts into functional proteins, which may be related to expression of suppressor of cytokine signaling type-2. These results contribute to our understanding of how neuroinflammation may contribute to the pathogenesis of age-related neurodegenerative disorders due to an age-related bias towards a hyper-reactive immune response that is not selective for a pro- or anti-inflammatory phenotype following an inflammatory stimulus.


Assuntos
Envelhecimento , Citocinas/metabolismo , Hipocampo/efeitos dos fármacos , Interleucina-1beta/farmacologia , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Neurobiol Aging ; 35(5): 1065-73, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24315728

RESUMO

Neuroinflammation and degeneration of ascending catecholaminergic systems occur early in the neurodegenerative process. Age and the duration of a pro-inflammatory environment induced by continuous intraventricular lipopolysaccharide (LPS) differentially affect the expression profile of pro- and anti-inflammatory genes and proteins as well as the number of activated microglia (express major histocompatibility complex II; MHC II) and the integrity and density of ascending catecholaminergic neural systems originating from the locus coeruleus (LC) and substantia nigra pars compacta (SNpc) in rats. LPS infusion increased gene expression and/or protein levels for both pro- and anti-inflammatory biomarkers. Although LPS infusion stimulated a robust increase in IL-1ß gene and protein expression, this increase was blunted with age. LPS infusion also increased the density of activated microglia cells throughout the midbrain and brainstem. Corresponding to the development of a pro-inflammatory environment, LC and SNpc neurons immunopositive for tyrosine-hydroxylase (the rate-limiting synthetic enzyme for dopamine and norepinephrine) decreased in number, along with a decrease in tyrosine-hydroxylase gene expression in the midbrain and/or brainstem region. Our data support the concept that continuous exposure to a pro-inflammatory environment drives exaggerated changes in the production and release of inflammatory mediators that interact with age to impair functional capacity of the SNpc and LC.


Assuntos
Envelhecimento/imunologia , Envelhecimento/patologia , Catecolaminas/fisiologia , Inflamação/genética , Inflamação/patologia , Locus Cerúleo/imunologia , Locus Cerúleo/patologia , Neuroimunomodulação/genética , Neurônios/imunologia , Neurônios/patologia , Substância Negra/imunologia , Substância Negra/patologia , Envelhecimento/genética , Animais , Expressão Gênica , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Interleucina-1beta/metabolismo , Lipopolissacarídeos/imunologia , Masculino , Microglia/imunologia , Microglia/patologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/imunologia
16.
Neurobiol Aging ; 34(10): 2293-301, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23639208

RESUMO

The current study investigated the hypothesis that the duration of the proinflammatory environment plays a critical role in the brain's response that results in negative consequences on cognition, biochemistry, and pathology. Lipopolysaccharide or artificial cerebrospinal fluid was slowly (250 ηg/h) infused into the fourth ventricle of young (3-month-old), adult (9-month-old), or aged (23-month-old) male F-344 rats for 21 or 56 days. The rats were then tested in the water pool task and endogenous hippocampal levels of pro- and anti-inflammatory proteins and genes and indicators of glutamatergic function were determined. The duration of the lipopolysaccharide infusion, compared with the age of the rat, had the greatest effect on (1) spatial working memory; (2) the density and distribution of activated microglia within the hippocampus; and (3) the cytokine protein and gene expression profiles within the hippocampus. The duration- and age-dependent consequences of neuroinflammation might explain why human adults respond positively to anti-inflammatory therapies and aged humans do not.


Assuntos
Envelhecimento/metabolismo , Hipocampo/fisiopatologia , Lipopolissacarídeos , Inflamação Neurogênica/induzido quimicamente , Inflamação Neurogênica/fisiopatologia , Envelhecimento/patologia , Envelhecimento/psicologia , Animais , Líquido Cefalorraquidiano , Citocinas/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Memória , Microglia/patologia , Inflamação Neurogênica/metabolismo , Inflamação Neurogênica/patologia , Ratos , Ratos Endogâmicos F344
17.
J Neuroimmune Pharmacol ; 8(5): 1098-105, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23709339

RESUMO

Impaired memory may result from synaptic glutamatergic dysregulation related to chronic neuroinflammation. GLT1 is the primary excitatory amino acid transporter responsible for regulating extracellular glutamate levels in the hippocampus. We tested the hypothesis that if impaired spatial memory results from increased extracellular glutamate due to age or experimentally induced chronic neuroinflammation in the hippocampus, then pharmacological augmentation of the glutamate transporter GLT1 will attenuate deficits in a hippocampal-dependent spatial memory task. The profile of inflammation-related genes and proteins associated with normal aging, or chronic neuroinflammation experimentally-induced via a four-week LPS infusion into the IV(th) ventricle, were correlated with performance in the Morris water maze following treatment with Riluzole, a drug that can enhance glutamate clearance by increasing GLT1 expression. Age-associated inflammation was qualitatively different from LPS-induced neuro-inflammation in young rats. LPS produced a pro-inflammatory phenotype characterized by increased IL-1ß expression in the hippocampus, whereas aging was not associated with a strong central pro-inflammatory response but with a mixed peripheral immune phenotype. Riluzole attenuated the spatial memory impairment, the elevation of serum cytokines and the decrease in GLT1 gene expression in Aged rats, but had no effect on young rats infused with LPS. Our findings highlight the therapeutic potential of reducing glutamatergic function upon memory impairment in neurodegenerative diseases associated with aging.


Assuntos
Envelhecimento/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Hipocampo/efeitos dos fármacos , Transtornos da Memória/metabolismo , Fármacos Neuroprotetores/farmacologia , Riluzol/farmacologia , Animais , Hipocampo/metabolismo , Lipopolissacarídeos/toxicidade , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Alzheimers Dis Parkinsonism ; 3: 110, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24600537

RESUMO

Chronic neuroinflammation is characteristic of neurodegenerative diseases and is present during very early stages, yet significant pathology and behavioral deficits do not manifest until advanced age. We investigated the consequences of experimentally-induced chronic neuroinflammation within the hippocampus and brainstem of young (4 mo) F-344 rats. Lipopolysaccharide (LPS) was infused continuously into the IVth ventricle for 2, 4 or 8 weeks. The number of MHC II immunoreactive microglia in the brain continued to increase throughout the infusion period. In contrast, performance in the Morris water maze was impaired after 4 weeks but recovered by 8 weeks. Likewise, a transient loss of tyrosine hydroxylase immunoreactivity in the substantia nigra and locus coeruleus was observed after 2 weeks, but returned to control levels by 4 weeks of continuous LPS infusion. These data suggest that direct activation of microglia is sufficient to drive, but not sustain, spatial memory impairment and a decrease in tyrosine hydroxylase production in young rats. Our previous studies suggest that chronic neuroinflammation elevates extracellular glutamate and that this elevation underlies the spatial memory impairment. In the current study, increased levels of GLT1 and SNAP25 in the hippocampus corresponded with the resolution of performance deficit. Increased expression of SNAP25 is consistent with reduced glutamate release from axonal terminals while increased GLT1 is consistent with enhanced clearance of extracellular glutamate. These data demonstrate the capacity of the brain to compensate for the presence of chronic neuroinflammation, despite continued activation of microglia, through changes in the regulation of the glutamatergic system.

19.
J Neurosci ; 32(15): 5186-99, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496564

RESUMO

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.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Ácido Glutâmico/farmacologia , Ácido Glutâmico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tecidual/metabolismo , Albuminas/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Clatrina/fisiologia , Dinaminas/fisiologia , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Citometria de Fluxo , Inativação Gênica , Imuno-Histoquímica , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Plasmídeos/genética , Proteína Quinase C/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Ácido Caínico/efeitos dos fármacos , Receptores de Ácido Caínico/metabolismo , Receptores de LDL/metabolismo , Sinapsinas/metabolismo , Transfecção , Proteínas Supressoras de Tumor/metabolismo , alfa-Macroglobulinas/metabolismo
20.
Neurobiol Learn Mem ; 96(2): 121-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21440650

RESUMO

Although tissue type plasminogen activator (tPA) and brain derived neurotrophic factor (BDNF) have been extensively described to influence brain outcomes in a number of disorders, their roles during physiological aging are poorly investigated. In the present study, we investigated whether maintenance of mice in different environmental conditions could influence age-associated changes in hippocampal tPA expression and BDNF maturation in relation with modifications of their cognitive performances. Our data indicate that maintenance in enriched housing led to a reversal of age-associated decrease in expression of hippocampal tPA. A subsequent increase in the level of mature BDNF and an improvement in emotional and spatial memories were observed. Taken together, these data suggest that the tPA-BDNF axis could play a critical role in the control of cognitive functions influenced both by the age and housing conditions.


Assuntos
Envelhecimento/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/fisiologia , Meio Ambiente , Neurônios/fisiologia , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Aprendizagem por Associação/fisiologia , Comportamento Animal/fisiologia , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Feminino , Abrigo para Animais , Memória/fisiologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...