134 resultados para Neuroinflammation
Resumo:
In recent years, previously unsuspected roles of astrocytes have been revealed, largely owing to the development of new tools enabling their selective study in situ. These exciting findings add to the large body of evidence demonstrating that astrocytes play a central role in brain homeostasis, in particular via the numerous cooperative metabolic processes they establish with neurons, such as the supply of energy metabolites and neurotransmitter recycling functions. Furthermore, impairments in astrocytic function are increasingly being recognized as an important contributor to neuronal dysfunction and, in particular, neurodegenerative processes. In this review, we discuss recent evidence supporting important roles for astrocytes in neuropathological conditions such as neuroinflammation, amyotrophic lateral sclerosis and Alzheimer's disease. We also explore the potential for neuroprotective therapeutics based on the modulation of astrocytic functions.
Resumo:
ABSTRACT: BACKGROUND: Neuroprotective and neurotrophic properties of leukemia inhibitory factor (LIF) have been widely reported. In the central nervous system (CNS), astrocytes are the major source for LIF, expression of which is enhanced following disturbances leading to neuronal damage. How astrocytic LIF expression is regulated, however, has remained an unanswered question. Since neuronal stress is associated with production of extracellular adenosine, we investigated whether LIF expression in astrocytes was mediated through adenosine receptor signaling. METHODS: Mouse cortical neuronal and astrocyte cultures from wild-type and adenosine A2B receptor knock-out animals, as well as adenosine receptor agonists/antagonists and various enzymatic inhibitors, were used to study LIF expression and release in astrocytes. When needed, a one-way analysis of variance (ANOVA) followed by Bonferroni post-hoc test was used for statistical analysis. RESULTS: We show here that glutamate-stressed cortical neurons induce LIF expression through activation of adenosine A2B receptor subtype in cultured astrocytes and require signaling of protein kinase C (PKC), mitogen-activated protein kinases (MAPKs: p38 and ERK1/2), and the nuclear transcription factor (NF)-κB. Moreover, LIF concentration in the supernatant in response to 5'-N-ethylcarboxamide (NECA) stimulation was directly correlated to de novo protein synthesis, suggesting that LIF release did not occur through a regulated release pathway. Immunocytochemistry experiments show that LIF-containing vesicles co-localize with clathrin and Rab11, but not with pHogrin, Chromogranin (Cg)A and CgB, suggesting that LIF might be secreted through recycling endosomes. We further show that pre-treatment with supernatants from NECA-treated astrocytes increased survival of cultured cortical neurons against glutamate, which was absent when the supernatants were pre-treated with an anti-LIF neutralizing antibody. CONCLUSIONS: Adenosine from glutamate-stressed neurons induces rapid LIF release in astrocytes. This rapid release of LIF promotes the survival of cortical neurons against excitotoxicity.
Resumo:
Neuroinflammation is observed in many brain pathologies: in neurodegenerative diseases and multiple sclerosis as well as in chemically induced lesions. It is characterized by the reactivity of microglial cells and astrocytes, activation of inducible NO-synthase (i-NOS), and increased expression and/or release of cytokines and chemokines. Clearly, cell-to-cell signaling between the different brain cell types plays an important role in the initiation and propagation of neuroinflammation, but despite the growing list of known molecular actors, the underlying pathways and the sequence of events remain to be fully elucidated. The present chapter presents an example of how to assess neuroinflammation in complex brain tissues, using aggregating brain cell cultures as an in vitro model. This three-dimensional cell culture system provides optimal cell-to-cell interactions crucial for histotypic cellular maturation and control of neuroinflammatory processes. The techniques described here comprise immunocytochemistry to assess the reactivity of microglia and astrocytes and the expression of cytokines; quantitative RT-PCR to measure the mRNA expression of cytokines (TNF-α, IL-1β, IL-6, IL-1ra, TGF-β, IL-15, IFN-γ), chemokines (ccl5, cxcl1, cxcl2), and i-NOS; and immunoblotting to assess MAP kinase pathway activation (phosphorylation of p38 and p44/42 MAP kinases).
Resumo:
BACKGROUND: Cerebral ischemia is associated with the activation of glial cells, infiltration of leukocytes and an increase in inflammatory mediators in the ischemic brain and systemic circulation. How this inflammatory response influences lesion size and neurological outcome remains unclear. D-JNKI1, an inhibitor of the c-Jun N-terminal kinase pathway, is strongly neuroprotective in animal models of stroke. Intriguingly, the protection mediated by D-JNKI1 is high even with intravenous administration at very low doses with undetectable drug levels in the brain, pointing to a systemic mode of action, perhaps on inflammation. FINDINGS: We evaluated whether D-JNKI1, administered intravenously 3 h after the onset of middle cerebral artery occlusion (MCAO), modulates secretion of the inflammatory mediators interleukin-6 and keratinocyte-derived chemokine in the plasma and from the spleen and brain at several time points after MCAO. We found an early release of both mediators in the systemic circulation followed by an increase in the brain and went on to show a later systemic increase in vehicle-treated mice. Release of interleukin-6 and keratinocyte-derived chemokine from the spleen of mice with MCAO was not significantly different from sham mice. Interestingly, the secretion of these inflammatory mediators was not altered in the systemic circulation or brain after successful neuroprotection with D-JNKI1. CONCLUSIONS: We demonstrate that neuroprotection with D-JNKI1 after experimental cerebral ischemia is independent of systemic and brain release of interleukin-6 and keratinocyte-derived chemokine. Furthermore, our findings suggest that the early systemic release of interleukin-6 and keratinocyte-derived chemokine may not necessarily predict an unfavorable outcome in this model.
Resumo:
Background. Microglia and astrocytes respond to homeostatic disturbances with profound changes of gene expression. This response, known as glial activation or neuroinflammation, can be detrimental to the surrounding tissue. The transcription factor CCAAT/enhancer binding protein ß (C/EBPß) is an important regulator of gene expression in inflammation but little is known about its involvement in glial activation. To explore the functional role of C/EBPß in glial activation we have analyzed pro-inflammatory gene expression and neurotoxicity in murine wild type and C/EBPß-null glial cultures. Methods. Due to fertility and mortality problems associated with the C/EBPß-null genotype we developed a protocol to prepare mixed glial cultures from cerebral cortex of a single mouse embryo with high yield. Wild-type and C/EBPß-null glial cultures were compared in terms of total cell density by Hoechst-33258 staining; microglial content by CD11b immunocytochemistry; astroglial content by GFAP western blot; gene expression by quantitative real-time PCR, western blot, immunocytochemistry and Griess reaction; and microglial neurotoxicity by estimating MAP2 content in neuronal/microglial cocultures. C/EBPß DNA binding activity was evaluated by electrophoretic mobility shift assay and quantitative chromatin immunoprecipitation. Results. C/EBPß mRNA and protein levels, as well as DNA binding, were increased in glial cultures by treatment with lipopolysaccharide (LPS) or LPS + interferon ¿ (IFN¿). Quantitative chromatin immunoprecipitation showed binding of C/EBPß to pro-inflammatory gene promoters in glial activation in a stimulus- and gene-dependent manner. In agreement with these results, LPS and LPS+IFN¿ induced different transcriptional patterns between pro-inflammatory cytokines and NO synthase-2 genes. Furthermore, the expressions of IL-1ß and NO synthase-2, and consequent NO production, were reduced in the absence of C/EBPß. In addition, neurotoxicity elicited by LPS+IFN¿-treated microglia co-cultured with neurons was completely abolished by the absence of C/EBPß in microglia.
Resumo:
BACKGROUND: Brain inflammation plays a central role in numerous brain pathologies, including multiple sclerosis (MS). Microglial cells and astrocytes are the effector cells of neuroinflammation. They can be activated also by agents such as interferon-gamma (IFN-gamma) and lipopolysaccharide (LPS). Peroxisome proliferator-associated receptor (PPAR) pathways are involved in the control of the inflammatory processes, and PPAR-beta seems to play an important role in the regulation of central inflammation. In addition, PPAR-beta agonists were shown to have trophic effects on oligodendrocytes in vitro, and to confer partial protection in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In the present work, a three-dimensional brain cell culture system was used as in vitro model to study antibody-induced demyelination and inflammatory responses. GW 501516, a specific PPAR-beta agonist, was examined for its capacity to protect from antibody-mediated demyelination and to prevent inflammatory responses induced by IFN-gamma and LPS. METHODS: Aggregating brain cells cultures were prepared from embryonal rat brain, and used to study the inflammatory responses triggered by IFN-gamma and LPS and by antibody-mediated demyelination induced by antibodies directed against myelin-oligodendrocyte glycoprotein (MOG). The effects of GW 501516 on cellular responses were characterized by the quantification of the mRNA expression of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), inducible NO synthase (i-NOS), PPAR-beta, PPAR-gamma, glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), and high molecular weight neurofilament protein (NF-H). GFAP expression was also examined by immunocytochemistry, and microglial cells were visualized by isolectin B4 (IB4) and ED1 labeling. RESULTS: GW 501516 decreased the IFN-gamma-induced up-regulation of TNF-alpha and iNOS in accord with the proposed anti-inflammatory effects of this PPAR-beta agonist. However, it increased IL-6 m-RNA expression. In demyelinating cultures, reactivity of both microglial cells and astrocytes was observed, while the expression of the inflammatory cytokines and iNOS remained unaffected. Furthermore, GW 501516 did not protect against the demyelination-induced changes in gene expression. CONCLUSION: Although GW 501516 showed anti-inflammatory activity, it did not protect against antibody-mediated demyelination. This suggests that the protective effects of PPAR-beta agonists observed in vivo can be attributed to their anti-inflammatory properties rather than to a direct protective or trophic effect on oligodendrocytes.
Resumo:
BACKGROUND: Fragile X-associated tremor/ataxia syndrome (FXTAS) is an inherited late-onset neurodegenerative disorder, characterized both by neurological and cognitive deficits. It is caused by the expansion of CGG repeats (55 to 200 repeats) in the noncoding region of the fragile X mental retardation 1 (FMR1) gene. Abnormal immunological patterns are often associated with neurodegenerative disorders and implicated in their etiology. We therefore investigated the immune status of FXTAS patients, which had not been assessed prior to this study. METHOD: Peripheral blood mononuclear cells (PBMCs) were collected from 15 asymptomatic FMR1 premutation carriers and 20 age-matched controls. Concentrations of three cytokines (IL-6, IL-8, IL-10) were measured in PBMC supernatants using ELISA assays. RESULTS: We found a significant increase in the concentration of the major anti-inflammatory cytokine IL-10 in supernatants of PBMCs derived from premutation carriers, when compared with controls (P = 0.019). This increase correlated significantly with the number of CGG repeats (P = 0.002). CONCLUSIONS: Elevated IL-10 levels were observed in all premutation carriers, before appearance of the classical neurological symptoms; therefore, IL-10 may be one of the early biomarkers of FXTAS.
Resumo:
Xenobiotic exposure is a risk factor in the etiology of neurodegenerative disease. It was recently hypothesized that restricted exposure during brain development could predispose for a neurodegenerative disease later in life. As neuroinflammation contributes to progressive neurodegeneration, it is suspected that neurodevelopmental xenobiotic exposure could elicit a neuroinflammatory process, which over time may assume a detrimental character. We investigated the neurotoxic effects of paraquat (PQ) in three-dimensional whole rat brain cell cultures, exposed during an early differentiation stage, comparing immediate effects-directly post exposure-with long-term effects, 20 days after interrupted PQ-administration. Adverse effects and neuroinflammatory responses were assessed by measuring changes in gene- and protein-expression as well as by determining cell morphology changes. Differentiating neural cultures were highly susceptible to PQ and showed neuronal damage and strong astrogliosis. After the 20-day washout period, neurons partially recovered, whereas astrogliosis persisted, and was accompanied by microglial activation of a neurodegenerative phenotype. Our data shows that immediate and long-term effects of subchronic PQ-exposure differ. Also, PQ-exposure during this window of extensive neuronal differentiation led to a delayed microglial activation, of a character that could promote further pro-inflammatory signals that enable prolonged inflammation, thereby fueling further neurodegeneration.
Resumo:
Résumé large public: Une altération localisée du métabolisme du glucose, le substrat énergétique préférentiellement utilisé dans le cerveau, est un trait caractéristique précoce de la maladie d'Alzheimer (MA). Il est maintenant largement admis que le beta-amyloïde, la neuroinflammation et le stress oxydatif participent au développement de la MA. Cependant les mécanismes cellulaires de la pathogenèse restent à identifier. Le métabolisme cérébral a ceci de remarquable qu'il repose sur la coopération entre deux types cellulaires, ainsi les astrocytes et les neurones constituent une unité métabolique. Les astrocytes sont notamment responsables de fournir aux neurones des substrats énergétiques, ainsi que des précurseurs du glutathion pour la défense contre le stress oxydatif. Ces fonctions astrocytaires sont essentielles au bon fonctionnement et à la survie neuronale; par conséquent, une altération de ces fonctions astrocytaires pourrait participer au développement de certaines maladies cérébrales. Le but de ce travail est, dans un premier temps, d'explorer les effets de médiateurs de la neuroinflammation (les cytokines pro-inflammatoires) et du peptide beta-amyloïde sur le métabolisme des astrocytes corticaux, en se focalisant sur les éléments en lien avec le métabolisme énergétique et le stress oxydatif. Puis, dans un second temps, de caractériser les conséquences pour les neurones des modifications du métabolisme astrocytaire induites par ces substances. Les résultats obtenus ici montrent que les cytokines pro-inflammatoires et le beta-amyloïde induisent une profonde altération du métabolisme astrocytaire, selon deux profils distincts. Les cytokines pro-inflammatoires, particulièrement en combinaison, agissent comme « découpleurs » du métabolisme énergétique du glucose, en diminuant l'apport potentiel de substrats énergétiques aux neurones. En plus de son effet propre, le peptide beta-amyloïde potentialise les effets des cytokines pro-inflammatoires. Or, dans le cerveau de patients atteints de la MA, les astrocytes sont exposés simultanément à ces deux types de substances. Les deux types de substances ont un effet ambivalent en termes de stress oxydatif. Ils induisent à la fois une augmentation de la libération de glutathion (potentiellement protecteur pour les neurones voisins) et la production d'espèces réactives de l'oxygène (potentiellement toxiques). Etant donné l'importance de la coopération entre astrocytes et neurones, ces modulations du métabolisme astrocytaire pourraient donc avoir un retentissement majeur sur les cellules environnantes, et en particulier sur la fonction et la survie neuronale. Résumé Les astrocytes et les neurones constituent une unité métabolique. Les astrocytes sont notamment responsables de fournir aux neurones des substrats énergétiques, tels que le lactate, ainsi que des précurseurs du glutathion pour la défense contre le stress oxydatif. Une altération localisée du métabolisme du glucose, le substrat énergétique préférentiellement utilisé dans le cerveau, est un trait caractéristique, précoce, de la maladie d'Alzheimer (MA). Il est maintenant largement admis que le beta-amyloïde, la neuroinflammation et le stress oxydatif participent au développement de la MA. Cependant, les mécanismes cellulaires de la pathogenèse restent à identifier. Le but de ce travail est d'explorer les effets des cytokines pro-inflammatoires (Il-1 ß et TNFα) et du beta-amyloïde (Aß) sur le métabolisme du glucose des astrocytes corticaux en culture primaire ainsi que de caractériser les conséquences, pour la viabilité des neurones voisins, des modifications du métabolisme astrocytaire induites par ces substances. Les résultats obtenus montrent que les cytokines pro-inflammatoires et le beta-amyloïde induisent une profonde altération du métabolisme astrocytaire, selon deux profils distincts. Les cytokines pro-inflammatoires, particulièrement en combinaison, agissent comme « découpleurs » du métabolisme glycolytique astrocytaire. Après 48 heures, le traitement avec TNFα et Il-lß cause une augmentation de la capture de glucose et de son métabolisme dans la voie des pentoses phosphates et dans le cycle de Krebs. A l'inverse, il cause une diminution de la libération de lactate et des stocks cellulaires de glycogène. En combinaison avec les cytokines tel qu'in vivo dans les cerveaux de patients atteints de MA, le peptide betaamyloïde potentialise les effets décrits ci-dessus. Isolément, le Aß cause une augmentation coordonnée de la capture de glucose et de toutes les voies de son métabolisme (libération de lactate, glycogenèse, voie des pentoses phosphate et cycle de Krebs). Les traitements altèrent peu les taux de glutathion intracellulaires, par contre ils augmentent massivement la libération de glutathion dans le milieu extracellulaire. A l'inverse, les deux types de traitements augmentent la production intracellulaire d'espèces réactives de l'oxygène (ROS). De plus, les cytokines pro-inflammatoires en combinaison augmentent massivement la production des ROS dans l'espace extracellulaire. Afin de caractériser l'impact de ces altérations métaboliques sur la viabilité des neurones environnants, un modèle de co-culture et des milieux conditionnés astrocytaires ont été utilisés. Les résultats montrent qu'en l'absence d'une source exogène d'antioxydants, la présence d'astrocytes favorise la viabilité neuronale ainsi que leur défense contre le stress oxydatif. Cette propriété n'est cependant pas modulée par les différents traitements. D'autre part, la présence d'astrocytes, et non de milieu conditionné, protège les neurones contre l'excitotoxicité due au glutamate. Les astrocytes prétraités (aussi bien avec le beta-amyloïde qu'avec les cytokines pro-inflammatoires) perdent cette propriété. Cet élément suggère que la perturbation du métabolisme astrocytaire causé par les cytokines pro-inflammatoires ou le beta-amyloïde pourrait participer à l'atteinte de la viabilité neuronale associée à certaines pathologies neurodégénératives.
Resumo:
Neuropathic pain is a clinical manifestation of nerve injury difficult to treat even with potent analgesic compounds. Here, we used different lines of genetically modified mice to clarify the role played by CB2 cannabinoid receptors in the regulation of the central immune responses leading to the development of neuropathic pain. CB2 knock-out mice and wild-type littermates were exposed to sciatic nerve injury, and both genotypes developed a similar hyperalgesia and allodynia in the ipsilateral paw. Most strikingly, knock-outs also developed a contralateral mirror image pain, associated with an enhanced microglial and astrocytic expression in the contralateral spinal horn. In agreement, hyperalgesia, allodynia, and microglial and astrocytic activation induced by sciatic nerve injury were attenuated in transgenic mice overexpressing CB2 receptors. These results demonstrate the crucial role of CB2 cannabinoid receptor in modulating glial activation in response to nerve injury. The enhanced manifestations of neuropathic pain were replicated in irradiated wild-type mice reconstituted with bone marrow cells from CB2 knock-outs, thus demonstrating the implication of the CB2 receptor expressed in hematopoietic cells in the development of neuropathic pain at the spinal cord.
Resumo:
Introduction : Doublecortin (DCX) is a microtubule associated protein expressed by migrating neural precursors. DCX is also expressed in approximately 4% of all cortical cells in adult normal primate brain. DCX expression is also enhanced locally in response to an acute insult made to the brain. This is thought to play a role in plasticity or neural repair. That being said, it would be interesting to know how the expression of DCX is modified in a more chronic insult, like in neurodegeneration such as in Parkinson's Disease (PD) and Alzheimer's Disease (AD). The aim of my study is to study the expression of DCX cells in the cortex of patients having a neurodegenerative disease, compared to control patients. Method: DCX cells quantification on 9 DCX‐stained 5 μm thick formalin fixed paraffin embedded brain sections: 3 Alzheimer's disease patients, 3 Parkinson's disease patients and 3 control patients. Each patient had several sections that we could stain with different stainings (GALLYA, TAU, DCX). By using a computerized image analysis system (Explora Nova, La Rochelle, France), cortical columns were selected on areas on the cortex with a lot of degeneration subjectively observed on GALLYA stained sections and on TAU stained sections. Then total number of cells was counted on TAU sections, where all nuclei were colored in blue. Then the DCX cells were counted on the corresponding DCX sections. These values were standardized to a reference surface area. The ratio of DCX cells over total cells was then calculated. Results : There is a difference of DCX cell expression between Alzheimer's Disease patients and control patients. The percentage of dcx cells in the cortex of an Alzheimer's patient is around 12.54% ± 2.17%, where as in the cortex of control patients, it is around 5.47% ± 0.83%. On the other hand, there is no significant difference in the ratio of DCX cells over total cells between parkinson's patients and control patients, both having around 5% of DCX cells. Discussion: There is a dramatic increase of DCX expression in AD (12.5%) compared to PD and controls (5.5%). The increase in DCX ratio in AD may have two potential causes: 1.The increased ratio is due to DCX cells being more resistant to degeneration compared to surrounding cells which are degenerating due to AD, leading to the cortical atrophy observed in AD patients. So the decrease of total cells without any change in the number of DCX cells makes the ratio bigger in AD compared to the controls. 2.The increased ratio is due to an actual increase in DCX cells. This means that there is some neural repair to compensate the degenerative process, just like the repair process observed in acute lesions to the brain. This second idea can be integrated in the broader point of view of neuroinflammation. The progression of the disease would trigger neuroinflammation and the process following the primary inflammatory response which is neural repair. So our study can show that the increase in DCX cells is an attempt to repair the degenerated neurons, in the context of neuroinflammation triggered by the physiopathological progression of the disease.
Resumo:
Delta 9-tetrahydrocannabinol (THC) has been proposed as therapeutic agent in the treatment of multiple sclerosis. In the present study, we examined whether a modulation of brain inflammatory by THC may protect against demyelination. Myelinating aggregating brain cell cultures were subjected to demyelination by a repeated treatment (3x) with the two inflammatory agents interferon-y (IFN-y) and lipopolysaccharide (LPS). The effects of THC on an acute inflammatory reponse were also examined by treating the aggregates with a single application of the two inflammatory agents. THC effects on the demyelinating process and on several mediators of the inflammatory reponse were analyzed. THC treatment partially prevented the decreased immunoreactivity for MBP, and the decrease in MBP content measured by immunoblotting. It prevented IFN-y + LPS -induced microglial reactivity; and decreased the IFN-y + LPS-induced i8ncreased phosphorylation of p44/42 MAP kinase. The other inflammatory markers, I-NOS and TNF-a mRNA expression, and p38 MAP kinase phosphorylation of p44/42 MAP kinase. The other inflammatory markers, I-NOS and TNF-a mRNA expression, and p38 MAP kinase phosphorylation were downregulated by THC treatment following a single application of the inflammatory agents, but not after repeated applications. THC protected partially against the IFN-y + LPS-induced demyelination. The protective effect of THC on IFN-y + LPS-induced demyelination may be due to a decrease of the inflammatory reponse. However, the anti-inflammatory effect of THC on some inflammatory markers is lost when the inflammatory response is more proeminent and of longer duration, suggesting either that the anti-inflammatory effect of a molecule may depend on the properties of the inflammatory response, or that the anti-inflammatory potential of THC decreases in case of repeated exposure.
Resumo:
BACKGROUND: The long latent stage seen in syphilis, followed by chronic central nervous system infection and inflammation, can be explained by the persistence of atypical cystic and granular forms of Treponema pallidum. We investigated whether a similar situation may occur in Lyme neuroborreliosis. METHOD: Atypical forms of Borrelia burgdorferi spirochetes were induced exposing cultures of Borrelia burgdorferi (strains B31 and ADB1) to such unfavorable conditions as osmotic and heat shock, and exposure to the binding agents Thioflavin S and Congo red. We also analyzed whether these forms may be induced in vitro, following infection of primary chicken and rat neurons, as well as rat and human astrocytes. We further analyzed whether atypical forms similar to those induced in vitro may also occur in vivo, in brains of three patients with Lyme neuroborreliosis. We used immunohistochemical methods to detect evidence of neuroinflammation in the form of reactive microglia and astrocytes. RESULTS: Under these conditions we observed atypical cystic, rolled and granular forms of these spirochetes. We characterized these abnormal forms by histochemical, immunohistochemical, dark field and atomic force microscopy (AFM) methods. The atypical and cystic forms found in the brains of three patients with neuropathologically confirmed Lyme neuroborreliosis were identical to those induced in vitro. We also observed nuclear fragmentation of the infected astrocytes using the TUNEL method. Abundant HLA-DR positive microglia and GFAP positive reactive astrocytes were present in the cerebral cortex. CONCLUSION: The results indicate that atypical extra- and intracellular pleomorphic and cystic forms of Borrelia burgdorferi and local neuroinflammation occur in the brain in chronic Lyme neuroborreliosis. The persistence of these more resistant spirochete forms, and their intracellular location in neurons and glial cells, may explain the long latent stage and persistence of Borrelia infection. The results also suggest that Borrelia burgdorferi may induce cellular dysfunction and apoptosis. The detection and recognition of atypical, cystic and granular forms in infected tissues is essential for the diagnosis and the treatment as they can occur in the absence of the typical spiral Borrelia form.
Resumo:
Neuroinflammation is the local reaction of the brain to infection, trauma, toxic molecules or protein aggregates. The brain resident macrophages, microglia, are able to trigger an appropriate response involving secretion of cytokines and chemokines, resulting in the activation of astrocytes and recruitment of peripheral immune cells. IL-1β plays an important role in this response; yet its production and mode of action in the brain are not fully understood and its precise implication in neurodegenerative diseases needs further characterization. Our results indicate that the capacity to form a functional NLRP3 inflammasome and secretion of IL-1β is limited to the microglial compartment in the mouse brain. We were not able to observe IL-1β secretion from astrocytes, nor do they express all NLRP3 inflammasome components. Microglia were able to produce IL-1β in response to different classical inflammasome activators, such as ATP, Nigericin or Alum. Similarly, microglia secreted IL-18 and IL-1α, two other inflammasome-linked pro-inflammatory factors. Cell stimulation with α-synuclein, a neurodegenerative disease-related peptide, did not result in the release of active IL-1β by microglia, despite a weak pro-inflammatory effect. Amyloid-β peptides were able to activate the NLRP3 inflammasome in microglia and IL-1β secretion occurred in a P2X7 receptor-independent manner. Thus microglia-dependent inflammasome activation can play an important role in the brain and especially in neuroinflammatory conditions.
Resumo:
BACKGROUND: Increasing evidences link T helper 17 (Th17) cells with multiple sclerosis (MS). In this context, interleukin-22 (IL-22), a Th17-linked cytokine, has been implicated in blood brain barrier breakdown and lymphocyte infiltration. Furthermore, polymorphism between MS patients and controls has been recently described in the gene coding for IL-22 binding protein (IL-22BP). Here, we aimed to better characterize IL-22 in the context of MS. METHODS: IL-22 and IL-22BP expressions were assessed by ELISA and qPCR in the following compartments of MS patients and control subjects: (1) the serum, (2) the cerebrospinal fluid, and (3) immune cells of peripheral blood. Identification of the IL-22 receptor subunit, IL-22R1, was performed by immunohistochemistry and immunofluorescence in human brain tissues and human primary astrocytes. The role of IL-22 on human primary astrocytes was evaluated using 7-AAD and annexin V, markers of cell viability and apoptosis, respectively. RESULTS: In a cohort of 141 MS patients and healthy control (HC) subjects, we found that serum levels of IL-22 were significantly higher in relapsing MS patients than in HC but also remitting and progressive MS patients. Monocytes and monocyte-derived dendritic cells contained an enhanced expression of mRNA coding for IL-22BP as compared to HC. Using immunohistochemistry and confocal microscopy, we found that IL-22 and its receptor were detected on astrocytes of brain tissues from both control subjects and MS patients, although in the latter, the expression was higher around blood vessels and in MS plaques. Cytometry-based functional assays revealed that addition of IL-22 improved the survival of human primary astrocytes. Furthermore, tumor necrosis factor α-treated astrocytes had a better long-term survival capacity upon IL-22 co-treatment. This protective effect of IL-22 seemed to be conferred, at least partially, by a decreased apoptosis. CONCLUSIONS: We show that (1) there is a dysregulation in the expression of IL-22 and its antagonist, IL-22BP, in MS patients, (2) IL-22 targets specifically astrocytes in the human brain, and (3) this cytokine confers an increased survival of the latter cells.