Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Glia ; 70(11): 2032-2044, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35770802

RESUMO

Leigh syndrome is a mitochondrial disease characterized by neurodegeneration, neuroinflammation, and early death. Mice lacking NDUFS4, a mitochondrial complex I subunit (Ndufs4 KO mice), have been established as a good animal model for studying human pathology associated with Leigh syndrome. As the disease progresses, there is an increase in neurodegeneration and neuroinflammation, thereby leading to deteriorating neurological symptoms, including motor deficits, breathing alterations, and eventually, death of the animal. However, despite the magnitude of neuroinflammation associated with brain lesions, the role of neuroinflammatory pathways and their main cellular components have not been addressed directly as relevant players in the disease pathology. Here, we investigate the role of microglial cells, the main immune cells of the CNS, in Leigh-like syndrome pathology, by pharmacologically depleting them using the colony-stimulating factor 1 receptor antagonist PLX3397. Microglial depletion extended lifespan and delayed motor symptoms in Ndufs4 KO mice, likely by preventing neuronal loss. Next, we investigated the role of the major cytokine interleukin-6 (IL-6) in the disease progression. IL-6 deficiency partially rescued breathing abnormalities and modulated gliosis but did not extend the lifespan or rescue motor decline in Ndufs4 KO mice. The present results show that microglial accumulation is pathogenic, in a process independent of IL-6, and hints toward a contributing role of neuroinflammation in the disease of Ndufs4 KO mice and potentially in patients with Leigh syndrome.


Assuntos
Doença de Leigh , Animais , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Interleucina-6/metabolismo , Doença de Leigh/genética , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Camundongos , Camundongos Knockout , Microglia/metabolismo
2.
Brain ; 145(1): 45-63, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-34849584

RESUMO

Mitochondria are small cellular constituents that generate cellular energy (ATP) by oxidative phosphorylation (OXPHOS). Dysfunction of these organelles is linked to a heterogeneous group of multisystemic disorders, including diabetes, cancer, ageing-related pathologies and rare mitochondrial diseases. With respect to the latter, mutations in subunit-encoding genes and assembly factors of the first OXPHOS complex (complex I) induce isolated complex I deficiency and Leigh syndrome. This syndrome is an early-onset, often fatal, encephalopathy with a variable clinical presentation and poor prognosis due to the lack of effective intervention strategies. Mutations in the nuclear DNA-encoded NDUFS4 gene, encoding the NADH:ubiquinone oxidoreductase subunit S4 (NDUFS4) of complex I, induce 'mitochondrial complex I deficiency, nuclear type 1' (MC1DN1) and Leigh syndrome in paediatric patients. A variety of (tissue-specific) Ndufs4 knockout mouse models were developed to study the Leigh syndrome pathomechanism and intervention testing. Here, we review and discuss the role of complex I and NDUFS4 mutations in human mitochondrial disease, and review how the analysis of Ndufs4 knockout mouse models has generated new insights into the MC1ND1/Leigh syndrome pathomechanism and its therapeutic targeting.


Assuntos
Complexo I de Transporte de Elétrons , Doença de Leigh , Doenças Mitocondriais , Animais , Complexo I de Transporte de Elétrons/genética , Humanos , Doença de Leigh/genética , Camundongos , Camundongos Knockout , Doenças Mitocondriais/genética , Fosforilação Oxidativa
3.
Theranostics ; 11(20): 9805-9820, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34815787

RESUMO

Background: Microglia and macrophages adopt a pro-inflammatory phenotype after spinal cord injury (SCI), what is thought to contribute to secondary tissue degeneration. We previously reported that this is due, in part, to the low levels of anti-inflammatory cytokines, such as IL-4. Since IL-13 and IL-4 share receptors and both cytokines drive microglia and macrophages towards an anti-inflammatory phenotype in vitro, here we studied whether administration of IL-13 and IL-4 after SCI leads to beneficial effects. Methods: We injected mice with recombinant IL-13 or IL-4 at 48 h after SCI and assessed their effects on microglia and macrophage phenotype and functional outcomes. We also performed RNA sequencing analysis of macrophages and microglia sorted from the injured spinal cords of mice treated with IL-13 or IL-4 and evaluated the metabolic state of these cells by using Seahorse technology. Results: We observed that IL-13 induced the expression of anti-inflammatory markers in microglia and macrophages after SCI but, in contrast to IL-4, it failed to mediate functional recovery. We found that these two cytokines induced different gene signatures in microglia and macrophages after SCI and that IL-4, in contrast to IL-13, shifted microglia and macrophage metabolism from glycolytic to oxidative phosphorylation. These findings were further confirmed by measuring the metabolic profile of these cells. Importantly, we also revealed that macrophages stimulated with IL-4 or IL-13 are not deleterious to neurons, but they become cytotoxic when oxidative metabolism is blocked. This suggests that the metabolic shift, from glycolysis to oxidative phosphorylation, is required to minimize the cytotoxic responses of microglia and macrophages. Conclusions: These results reveal that the metabolic fitness of microglia and macrophages after SCI contributes to secondary damage and that strategies aimed at boosting oxidative phosphorylation might be a novel approach to minimize the deleterious actions of microglia and macrophages in neurotrauma.


Assuntos
Interleucina-13/metabolismo , Interleucina-4/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Interleucina-13/imunologia , Interleucina-13/farmacologia , Interleucina-4/imunologia , Interleucina-4/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento
4.
Mol Cell Proteomics ; 15(2): 445-61, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26450614

RESUMO

Elevated fumarate concentrations as a result of Krebs cycle inhibition lead to increases in protein succination, an irreversible post-translational modification that occurs when fumarate reacts with cysteine residues to generate S-(2-succino)cysteine (2SC). Metabolic events that reduce NADH re-oxidation can block Krebs cycle activity; therefore we hypothesized that oxidative phosphorylation deficiencies, such as those observed in some mitochondrial diseases, would also lead to increased protein succination. Using the Ndufs4 knockout (Ndufs4 KO) mouse, a model of Leigh syndrome, we demonstrate for the first time that protein succination is increased in the brainstem (BS), particularly in the vestibular nucleus. Importantly, the brainstem is the most affected region exhibiting neurodegeneration and astrocyte and microglial proliferation, and these mice typically die of respiratory failure attributed to vestibular nucleus pathology. In contrast, no increases in protein succination were observed in the skeletal muscle, corresponding with the lack of muscle pathology observed in this model. 2D SDS-PAGE followed by immunoblotting for succinated proteins and MS/MS analysis of BS proteins allowed us to identify the voltage-dependent anion channels 1 and 2 as specific targets of succination in the Ndufs4 knockout. Using targeted mass spectrometry, Cys(77) and Cys(48) were identified as endogenous sites of succination in voltage-dependent anion channels 2. Given the important role of voltage-dependent anion channels isoforms in the exchange of ADP/ATP between the cytosol and the mitochondria, and the already decreased capacity for ATP synthesis in the Ndufs4 KO mice, we propose that the increased protein succination observed in the BS of these animals would further decrease the already compromised mitochondrial function. These data suggest that fumarate is a novel biochemical link that may contribute to the progression of the neuropathology in this mitochondrial disease model.


Assuntos
Complexo I de Transporte de Elétrons/genética , Doença de Leigh/genética , Proteômica , Succinatos/metabolismo , Animais , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Ciclo do Ácido Cítrico , Cisteína/metabolismo , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Fumaratos/metabolismo , Humanos , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Processamento de Proteína Pós-Traducional/genética , Espectrometria de Massas em Tandem
5.
J Neurosci ; 35(14): 5549-56, 2015 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-25855171

RESUMO

Hypothalamic neuronal populations are central regulators of energy homeostasis and reproductive function. However, the ontogeny of these critical hypothalamic neuronal populations is largely unknown. We developed a novel approach to examine the developmental pathways that link specific subtypes of neurons by combining embryonic and adult ribosome-tagging strategies in mice. This new method shows that Pomc-expressing precursors not only differentiate into discrete neuronal populations that mediate energy balance (POMC and AgRP neurons), but also into neurons critical for puberty onset and the regulation of reproductive function (Kiss1 neurons). These results demonstrate a developmental link between nutrient-sensing and reproductive neuropeptide synthesizing neuronal populations and suggest a potential pathway that could link maternal nutrition to reproductive development in the offspring.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Hipotálamo/citologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Células-Tronco/fisiologia , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Dependovirus/genética , Embrião de Mamíferos , Vetores Genéticos/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imunoprecipitação , Kisspeptinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries , Pró-Opiomelanocortina/genética , RNA Mensageiro/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
6.
Brain Behav Immun ; 40: 121-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24632224

RESUMO

Interleukin-6 (IL-6) is a major cytokine controlling not only the immune system but also basic physiological variables such as body weight and metabolism. While central IL-6 is clearly implicated in the latter, the putative role of peripheral IL-6 controlling body weight remains unclear. We herewith report results obtained in muscle-specific IL-6 KO (mIL-6 KO) mice. mIL-6 KO male mice fed a high-fat diet (HFD, 58.4% kcal from fat) or a control diet (18%) gained less weight and body fat than littermate floxed male mice, while the opposite pattern was observed in female mice. Food intake was not affected by muscle IL-6 deficiency, but male and female mIL-6 KO mice were more and less active, respectively, in the hole-board test. Moreover, female mIL-6 KO mice did not control adequately their body temperature upon exposure to 4°C, suggesting a role of muscle IL-6 in energy expenditure. At least part of this regulatory role of muscle IL-6 may be mediated by the hypothalamus, as IL-6 deficiency regulated the expression of critical hypothalamic neuropeptides (NPY, AgRP, POMC, CRH and preproOX). Leptin and insulin changes cannot explain the phenotype of these mice. In summary, the present results demonstrate that muscle IL-6 controls body weight and body fat in a sex-specific fashion, influencing the expression of the main neuropeptides involved in energy homeostasis.


Assuntos
Tecido Adiposo/metabolismo , Peso Corporal/genética , Interleucina-6/genética , Obesidade/genética , Animais , Glicemia/metabolismo , Regulação da Temperatura Corporal , Metabolismo Energético , Feminino , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Neuropeptídeos/metabolismo , Obesidade/metabolismo , Fatores Sexuais
7.
Science ; 342(6165): 1524-8, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24231806

RESUMO

Mitochondrial dysfunction contributes to numerous health problems, including neurological and muscular degeneration, cardiomyopathies, cancer, diabetes, and pathologies of aging. Severe mitochondrial defects can result in childhood disorders such as Leigh syndrome, for which there are no effective therapies. We found that rapamycin, a specific inhibitor of the mechanistic target of rapamycin (mTOR) signaling pathway, robustly enhances survival and attenuates disease progression in a mouse model of Leigh syndrome. Administration of rapamycin to these mice, which are deficient in the mitochondrial respiratory chain subunit Ndufs4 [NADH dehydrogenase (ubiquinone) Fe-S protein 4], delays onset of neurological symptoms, reduces neuroinflammation, and prevents brain lesions. Although the precise mechanism of rescue remains to be determined, rapamycin induces a metabolic shift toward amino acid catabolism and away from glycolysis, alleviating the buildup of glycolytic intermediates. This therapeutic strategy may prove relevant for a broad range of mitochondrial diseases.


Assuntos
Doença de Leigh/tratamento farmacológico , Doenças Mitocondriais/tratamento farmacológico , Terapia de Alvo Molecular , Complexos Multiproteicos/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Glicólise/efeitos dos fármacos , Doença de Leigh/genética , Doença de Leigh/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia
8.
Glia ; 61(4): 587-600, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23322593

RESUMO

Interleukin (IL)-6 is crucial for the induction of many murine models of autoimmunity including experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. While IL-6-deficient mice (IL-6 KO) are resistant to EAE, we showed previously that in transgenic mice with astrocyte-targeted production of IL-6-restricted to the cerebellum (GFAP-IL6), EAE induced with MOG(35-55) was redirected away from the spinal cord to the cerebellum. To further establish the importance of IL-6 produced in the central nervous system, we have generated mice producing IL-6 essentially only in the brain by crossing the GFAP-IL6 mice with IL-6 KO mice. Interestingly, GFAP-IL6-IL-6 KO mice showed a milder but almost identical phenotype as the GFAP-IL6 mice, which correlated with a lower load of inflammatory cells and decreased microglial reactivity. These results indicate that not only is cerebellar IL-6 production and eventual leakage into the peripheral compartment the dominating factor controlling this type of EAE but that it can also facilitate induction of autoimmunity in the absence of normal systemic IL-6 production.


Assuntos
Astrócitos/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Interleucina-6/biossíntese , Animais , Astrócitos/metabolismo , Células Cultivadas , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Interleucina-6/deficiência , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
9.
Brain Behav Immun ; 27(1): 162-73, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23085146

RESUMO

Interleukin-6 (IL-6) is a major cytokine which controls not only the immune system but also exhibits many other functions including effects in the central nervous system (CNS). IL-6 is known to be produced by different cells in the CNS, and all the major CNS do respond to IL-6, which makes it difficult to dissect the specific roles of each cell type when assessing the role of IL-6 in the brain. We have produced for the first time floxed mice for IL-6 and have crossed them with GFAP-Cre mice to delete IL-6 in astrocytes (Ast-IL-6 KO mice), and have compared their phenotype with that of mice with deletion of IL-6 receptor in astrocytes (Ast-IL6R KO mice). Our results indicate a major prosurvival role of the astrocyte IL-6 system at early ages (intrauterine life), which was also involved to various degrees in the control of adult body weight, locomotor activity, anxiety and exploratory behaviors. In some occasions deleting IL-6R in astrocytes mimicked the phenotype of Ast-IL-6 KO mice (i.e. activity), while in others the opposite was observed (i.e. exploration), suggesting autocrine and paracrine (presumably on neurons) roles of astrocyte IL-6. Our results suggest important roles of the astrocyte IL-6 system on normal brain physiology, in some cases totally unexpected from previous results with total IL-6 KO mice.


Assuntos
Astrócitos/metabolismo , Comportamento Animal/fisiologia , Peso Corporal/fisiologia , Interleucina-6/fisiologia , Receptores de Interleucina-6/metabolismo , Animais , Ansiedade/genética , Ansiedade/metabolismo , Astrócitos/citologia , Peso Corporal/genética , Comportamento Exploratório/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Atividade Motora/fisiologia , Reação em Cadeia da Polimerase , RNA/análise , Receptores de Interleucina-6/genética , Fatores Sexuais
10.
Int J Biol Sci ; 8(9): 1254-66, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23136554

RESUMO

Interleukin-6 (IL-6) is a cytokine originally identified almost 30 years ago as a B-cell differentiation factor, capable of inducing the maturation of B cells into antibody-producing cells. As with many other cytokines, it was soon realized that IL-6 was not a factor only involved in the immune response, but with many critical roles in major physiological systems including the nervous system. IL-6 is now known to participate in neurogenesis (influencing both neurons and glial cells), and in the response of mature neurons and glial cells in normal conditions and following a wide arrange of injury models. In many respects, IL-6 behaves in a neurotrophin-like fashion, and seemingly makes understandable why the cytokine family that it belongs to is known as neuropoietins. Its expression is affected in several of the main brain diseases, and animal models strongly suggest that IL-6 could have a role in the observed neuropathology and that therefore it is a clear target of strategic therapies.


Assuntos
Sistema Nervoso Central/metabolismo , Interleucina-6/fisiologia , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Modelos Biológicos , Neurogênese , Receptores de Interleucina-6/metabolismo , Receptores de Interleucina-6/fisiologia
11.
J Clin Invest ; 122(7): 2359-68, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22653057

RESUMO

Leigh syndrome (LS) is a subacute necrotizing encephalomyelopathy with gliosis in several brain regions that usually results in infantile death. Loss of murine Ndufs4, which encodes NADH dehydrogenase (ubiquinone) iron-sulfur protein 4, results in compromised activity of mitochondrial complex I as well as progressive neurodegenerative and behavioral changes that resemble LS. Here, we report the development of breathing abnormalities in a murine model of LS. Magnetic resonance imaging revealed hyperintense bilateral lesions in the dorsal brain stem vestibular nucleus (VN) and cerebellum of severely affected mice. The mutant mice manifested a progressive increase in apnea and had aberrant responses to hypoxia. Electrophysiological recordings within the ventral brain stem pre-Bötzinger respiratory complex were also abnormal. Selective inactivation of Ndufs4 in the VN, one of the principle sites of gliosis, also led to breathing abnormalities and premature death. Conversely, Ndufs4 restoration in the VN corrected breathing deficits and prolonged the life span of knockout mice. These data demonstrate that mitochondrial dysfunction within the VN results in aberrant regulation of respiration and contributes to the lethality of Ndufs4-knockout mice.


Assuntos
Complexo I de Transporte de Elétrons/genética , Doença de Leigh/genética , Insuficiência Respiratória/genética , Potenciais de Ação , Análise de Variância , Animais , Apneia/genética , Tronco Encefálico/patologia , Tronco Encefálico/fisiopatologia , Dependovirus/genética , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Terapia Genética , Vetores Genéticos , Gliose/genética , Gliose/patologia , Frequência Cardíaca , Humanos , Técnicas In Vitro , Doença de Leigh/fisiopatologia , Doença de Leigh/terapia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/sangue , Oxigênio/metabolismo , Insuficiência Respiratória/fisiopatologia , Insuficiência Respiratória/terapia , Taxa Respiratória , Núcleos Vestibulares/metabolismo , Núcleos Vestibulares/patologia , Núcleos Vestibulares/fisiopatologia
12.
J Immunol ; 183(3): 2079-88, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19597000

RESUMO

IL-6 is crucial for the induction of many murine models of autoimmunity including experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. To establish the role of site-specific production of IL-6 in autoimmunity, we examined myelin oligodendrocyte glycoprotein immunization-induced EAE in transgenic mice (GFAP-IL6) with IL-6 production restricted to the cerebellum. Myelin oligodendrocyte glycoprotein-immunized (Mi-) GFAP-IL6 mice developed severe ataxia but no physical signs of spinal cord involvement, which was in sharp contrast to Mi-wild type (WT) animals that developed classical EAE with ascending paralysis. Immune pathology and demyelination were nearly absent from the spinal cord, but significantly increased in the cerebellum of Mi-GFAP-IL6 mice. Tissue damage in the cerebellum in the Mi-GFAP-IL6 mice was accompanied by increased total numbers of infiltrating leukocytes and increased proportions of both neutrophils and B-cells. With the exception of IL-17 mRNA, which was elevated in both control immunized and Mi-GFAP-IL6 cerebellum, the level of other cytokine and chemokine mRNAs were comparable with Mi-WT cerebellum whereas significantly higher levels of IFN-gamma and TNF-alpha mRNA were found in Mi-WT spinal cord. Thus, site-specific production of IL-6 in the cerebellum redirects trafficking away from the normally preferred antigenic site the spinal cord and acts as a leukocyte "sink" that markedly enhances the inflammatory cell accumulation and disease. The mechanisms underlying this process likely include the induction of specific chemokines, activation of microglia, and activation and loss of integrity of the blood-brain barrier present in the cerebellum of the GFAP-IL6 mice before the induction of EAE.


Assuntos
Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/patologia , Inflamação/etiologia , Interleucina-6/biossíntese , Animais , Autoimunidade , Sistema Nervoso Central/metabolismo , Cerebelo/patologia , Quimiotaxia , Citocinas/análise , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/fisiopatologia , Interleucina-6/imunologia , Camundongos , Camundongos Transgênicos , Proteínas da Mielina , Glicoproteína Associada a Mielina/efeitos adversos , Glicoproteína Mielina-Oligodendrócito , Paralisia
13.
Mol Cell Neurosci ; 41(1): 19-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19386233

RESUMO

Endoplasmic reticulum (ER) stress has recently been proposed as one of the factors contributing to apoptotic cell death in Parkinson's disease (PD). Although MAO-B inhibitors have been suggested to exert neuroprotective effects in several experimental models of PD, their effectiveness against ER stress has not been fully determined. Therefore, we have studied the potential usefulness of PF9601N, a non-amphetamine-like MAO-B inhibitor, in preventing cell death in a cell culture model of ER stress. Exposure of human dopaminergic cell line SH-SY5Y to the ER stressor brefeldin A led to Golgi disassembly, activation of the unfolded protein response (UPR), and subsequent expression of the proapoptotic mediator GADD153/CHOP. In this context, PF9601N pretreatment prevented brefeldin A-induced UPR responses, thus blocking the expression of GADD153/CHOP and resulting apoptotic features. In summary, our data suggests that PF9601N is able to block the responses elicited by ER stress, thus preventing apoptotic cell death in brefeldin A-treated cells.


Assuntos
Morte Celular/fisiologia , Retículo Endoplasmático/metabolismo , Indóis/metabolismo , Monoaminoxidase/metabolismo , Fármacos Neuroprotetores/metabolismo , Estresse Fisiológico , Animais , Brefeldina A/metabolismo , Caspase 2/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Síntese de Proteínas/metabolismo , Fatores de Transcrição de Fator Regulador X , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tunicamicina/metabolismo
14.
J Neurosci Res ; 87(3): 784-94, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18831059

RESUMO

Cryolesion of the frontoparietal cortex in mice is a well-described brain injury paradigm that results in increased astrogliosis surrounding the lesion site and is accompanied by a prominent increase in the MAO-B levels in astrocytes. Whether these increased MAO-B levels contribute to cellular damage or modulate reactive astrocytosis remains unclear. MAO-B activity may contribute to cellular damage, since its metabolism products are highly toxic to the cells. Additionally, it has been suggested that MAO-B inhibition may regulate astrocytic reaction. In this study, we have determined the relative contribution of MAO-B activity to the outcome following freeze injury. Freeze injury induced a prominent increase of several inflammatory markers, including ICAM, Mac-1, EB22, and GFAP. Inhibition of MAO-B activity using the selective inhibitor PF9601N did not reduce this cryolesion-induced inflammatory response. Additional data revealed that the expression of several cryolesion-induced cell death genes, such as Fas, Rip, p53, and ICE, was not reduced in PF9601N-treated mice, evidencing that MAO-B activity did not contribute to cryolesion-induced cell death. Definitive functional analysis of the mice using the ladder beam task revealed that MAO-B inhibition did not improve the cryolesion-induced motor impairment. These data strongly suggest that, although MAO-B is highly expressed in the area surrounding the lesion site, its activity does not contribute to the cellular damage or play any role in regulating astrocytic reactivity.


Assuntos
Lesões Encefálicas/imunologia , Monoaminoxidase/metabolismo , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/enzimologia , Lesões Encefálicas/genética , Caspase 1/metabolismo , Moléculas de Adesão Celular/metabolismo , Morte Celular/fisiologia , Encefalite/tratamento farmacológico , Encefalite/enzimologia , Congelamento , Genes p53/efeitos dos fármacos , Proteína Glial Fibrilar Ácida , Indóis/farmacologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Endogâmicos , Inibidores da Monoaminoxidase/farmacologia , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptor fas/metabolismo
15.
J Neurochem ; 105(6): 2404-17, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18331475

RESUMO

PF9601N [N-(2-propynyl) 2-(5-benzyloxyindol) methylamine] is a non-amphetamine type MAO-B inhibitor that has shown neuroprotective properties in vivo using different experimental models of Parkinson's disease. The mechanisms underlying its neuroprotective effects are poorly understood, but appear to be independent of MAO-B inhibition. We have studied its neuroprotective properties using the human SH-SY5Y dopaminergic cell line exposed to 1-methyl-4-phenylpyridinium (MPP(+)), a cellular model of Parkinson's disease. PF9601N pre-treatment significantly reduced MPP(+)-induced cell death and decreased the activation of one of the main executioner caspases, caspase-3. MPP(+) induced stabilization of transcription factor p53, which led to increased levels of this transcription factor, its nuclear translocation and transactivation of p53 response elements. PF9601N prevented this increase, thus reducing its transcriptional activity. Additional results showed that p53 may mediate its pro-apoptotic actions through caspase-2 under our experimental conditions. PUMA-alpha may also contribute to the p53-induced cell death. Since PF9601N significantly reduced MPP(+)-induced caspase-2 activity and PUMA-alpha levels, this reduction may lead to increased cell survival. Thus, PF9601N is a novel molecule with an apparently novel mechanism of action which has a promising potential as a therapeutic agent in the treatment of neurodegenerative diseases.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Apoptose/efeitos dos fármacos , Indóis/farmacologia , Inibidores da Monoaminoxidase/toxicidade , Monoaminoxidase/metabolismo , Fármacos Neuroprotetores/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Apoptose/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Indóis/uso terapêutico , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/fisiologia
16.
Dev Neurobiol ; 68(2): 195-208, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18000830

RESUMO

Interleukin-6 (IL-6) is one of the key players in the response of the brain cortex to injury. We have described previously that astrocyte-driven production of IL-6 (GFAP-IL6) in transgenic mice, although causing spontaneous neuroinflammation and long term damage, is beneficial after an acute (freeze) injury in the cortex, increasing healing and decreasing oxidative stress and apoptosis. To determine the transcriptional basis for these responses here we analyzed the global gene expression profile of the cortex, at 0 (unlesioned), 1 or 4 days post lesion (dpl), in both GFAP-IL6 mice and their control littermates. GFAP-IL6 mice showed an increase in genes associated with the inflammatory response both at 1 dpl (Iftm1, Endod1) and 4 dpl (Gfap, C4b), decreased expression of proapoptotic genes (i.e. Gadd45b, Clic4, p21) as well as reduced expression of genes involved in the control of oxidative stress (Atf4). Furthermore, the presence of IL-6 altered the expression of genes involved in hemostasis (Vwf), cell migration and proliferation (Cap2), and synaptic activity (Vamp2). All these changes in gene expression could underlie the phenotype of the GFAP-IL6 mice after injury, but many other possible factors were also identified in this study, highlighting the utility of this approach for deciphering new pathways orchestrated by IL-6.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/genética , Encefalite/genética , Regulação da Expressão Gênica/genética , Interleucina-6/genética , Animais , Apoptose/genética , Astrócitos/imunologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/fisiopatologia , Movimento Celular/genética , Proliferação de Células , Encefalite/imunologia , Encefalite/fisiopatologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Proteína Glial Fibrilar Ácida/genética , Hemostasia/genética , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transmissão Sináptica/genética , Regulação para Cima/genética
17.
Neuroimmunomodulation ; 14(3-4): 139-43, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18073505

RESUMO

Traumatic brain injury is one of the leading causes of incapacity and death among young people. Injury to the brain elicits a potent inflammatory response, comprising recruitment of inflammatory cells, reactive astrogliosis and activation of brain macrophages. Under the influence of presumably several cytokines and growth factors, a cascade of events is activated that result ultimately in increased oxidative stress and tissue damage, but also in activation of counterregulatory factors and tissue regeneration. The complexity of this response is being unraveled by high-throughput methodologies such as microarrays. The combination of these modern techniques with the comparison of normal and genetically modified mice boosts the significance of the results obtained. With this approach, we have demonstrated that a cytokine such as interleukin-6 is one of the key players in the response of the brain to injury.


Assuntos
Lesões Encefálicas/complicações , Lesões Encefálicas/imunologia , Encefalite/genética , Encefalite/imunologia , Regulação da Expressão Gênica/imunologia , Interleucina-6/genética , Animais , Astrócitos/imunologia , Lesões Encefálicas/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Encefalite/fisiopatologia , Feminino , Perfilação da Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Gliose/genética , Gliose/imunologia , Gliose/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
18.
J Neurosci Res ; 85(12): 2668-85, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17131423

RESUMO

Cytokines, such as tumour necrosis factor (TNF)-alpha and lymphotoxin-alpha, have been described widely to play important roles in the brain in physiologic conditions and after traumatic injury. However, the exact mechanisms involved in their function have not been fully elucidated. We give some insight on their role by using animals lacking either Type 1 receptor (TNFR1KO) or Type 2 (TNFR2KO) and their controls (C57Bl/6). Both TNFR1KO and to a greater extent TNFR2KO mice showed increased exploration/activity neurobehavioral traits in the hole board test, such as rearings, head dippings, and ambulations, compared with wild-type mice, suggesting an inhibitory role of TNFR1/TNFR2 signaling. In contrast, no significant differences were observed in the elevated plus maze test, ruling out a major role of these receptors in the control of anxiety. We next evaluated the response to a freeze injury to the somatosensorial cortex. The effect of the cryolesion on motor function was evaluated with the horizontal ladder beam test, and the results showed that both TNFR1KO and TNFR2KO mice made fewer errors, suggesting a detrimental role for TNFR1/TNFR2 signaling for coping with brain damage. Expression of approximately 22600 genes was analyzed using an Affymetrix chip (MOE430A) at 0 (unlesioned), 1, or 4 days post-lesion in the three strains. The results show a unique and major role of both TNF receptors on the pattern of gene expression elicited by the injury but also in normal conditions, and suggest that blocking of TNFR1/TNFR2 receptors may be beneficial after a traumatic brain injury.


Assuntos
Comportamento Animal/fisiologia , Lesões Encefálicas/patologia , Encéfalo/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Recuperação de Função Fisiológica/genética , Análise de Variância , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/genética , Hibridização In Situ/métodos , Aprendizagem em Labirinto/fisiologia , Metalotioneína/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Desempenho Psicomotor/fisiologia , Receptores do Fator de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência
19.
J Neurosci Res ; 83(6): 974-84, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493670

RESUMO

Brain injury and neuroinflammation are pathophysiologic contributors to acute and chronic neurologic disorders, which are progressive diseases not fully understood. Mammalian metallothioneins I and II (MT-I&II) have significant neuroprotective functions, but the precise mechanisms underlying these effects are still unknown. To gain insight in this regard, we have evaluated whether a distant, most likely single-domain MT (Drosophila MTN) functions similarly to mammalian MT-I&II (recombinant mouse MT-I and human MT-IIa and native rabbit MT-II) after cryogenic injury to the cortex in Mt1&2 KO mice. All the recombinant proteins showed similar neuroprotective properties to native MT-II, significantly reducing brain inflammation (macrophages, T cells, and pro-inflammatory cytokines), oxidative stress, neurodegeneration, and apoptosis. These results in principle do not support specific protein-protein interactions as the mechanism underlying the neuroprotective effects of these proteins because a non-homologous and structurally unrelated MT such as Drosophila MTN functions similarly to mammalian MTs. We have also evaluated for the first time the neurobiologic effects of exogenous MT-III, a major CNS MT isoform. Human rMT-III, in contrast to human nMT-IIa, did not affect inflammation, oxidative stress, and apoptosis, and showed opposite effects on several growth factors, neurotrophins, and markers of synaptic growth and plasticity. Our data thus highlight specific and divergent roles of exogenous MT-III vs. the MT-I&II isoforms that are consistent with those attributed to the endogenous proteins, and confirm the suitability of recombinant synthesis for future therapeutic use that may become relevant to clinical neurology.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Metalotioneína/metabolismo , Neurobiologia/métodos , Análise de Variância , Animais , Antígenos CD/metabolismo , Apoptose/fisiologia , Lesões Encefálicas/complicações , Lesões Encefálicas/tratamento farmacológico , Contagem de Células/métodos , Interações Medicamentosas , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Inflamação/etiologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Metalotioneína/classificação , Metalotioneína/deficiência , Metalotioneína/uso terapêutico , Camundongos , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Coelhos , Proteínas Recombinantes/metabolismo , Sensibilidade e Especificidade
20.
J Neurosci Res ; 82(5): 701-16, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16267827

RESUMO

Tumor necrosis factor-alpha (TNF-alpha) is one of the mediators dramatically increased after traumatic brain injury that leads to the activation, proliferation, and hypertrophy of mononuclear, phagocytic cells and gliosis. Eventually, TNF-alpha can induce both apoptosis and necrosis via intracellular signaling. This cytokine exerts its functions via interaction with two receptors: type-1 receptor (TNFR1) and type-2 receptor (TNFR2). In this work, the inflammatory response after a freeze injury (cryolesion) in the cortex was studied in wild-type (WT) animals and in mice lacking TNFR1 (TNFR1 KO) or TNFR2 (TNFR2 KO). Lack of TNFR1, but not of TNFR2, significantly decreased the inflammatory response and tissue damage elicited by the cryolesion at both 3 and 7 days postlesion, with decreased gliosis, lower IL-1beta immunostaining, and a reduction of apoptosis markers. Cryolesion produced a clear induction of the proinflammatory cytokines interleukin (IL)-1alpha, IL-1beta, IL-6, and TNF-alpha; this induction was significantly lower in the TNFR1 KO mice. Host response genes (ICAM-1, A20, EB22/5, and GFAP) were also induced by the cryolesion, but to a lesser extent in TNFR1 KO mice. Lack of TNFR1 signaling also affected the expression of apoptosis/cell death-related genes (Fas, Rip, p53), matrix metalloproteinases (MMP3, MMP9, MMP12), and their inhibitors (TIMP1), suggesting a role of TNFR1 in extracellular matrix remodeling after injury. However, GDNF, NGF, and BDNF expression were not affected by TNFR1 deficiency. Overall, these results suggest that TNFR1 is involved in the early establishment of the inflammatory response and that its deficiency causes a decreased inflammatory response and tissue damage following brain injury.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Encefalite/metabolismo , Degeneração Neural/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Animais , Encéfalo/fisiopatologia , Lesões Encefálicas/genética , Lesões Encefálicas/fisiopatologia , Temperatura Baixa/efeitos adversos , Citocinas/metabolismo , Denervação , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Encefalite/genética , Encefalite/fisiopatologia , Feminino , Regulação da Expressão Gênica/genética , Genes Precoces/fisiologia , Gliose/genética , Gliose/metabolismo , Gliose/fisiopatologia , Substâncias de Crescimento/genética , Camundongos , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA