Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 12167, 2024 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806485

RESUMO

During neuroinflammation, monocytes that infiltrate the central nervous system (CNS) may contribute to regenerative processes depending on their activation status. However, the extent and mechanisms of monocyte-induced CNS repair in patients with neuroinflammatory diseases remain largely unknown, partly due to the lack of a fully human assay platform that can recapitulate monocyte-neural stem cell interactions within the CNS microenvironment. We therefore developed a human model system to assess the impact of monocytic factors on neural stem cells, establishing a high-content compatible assay for screening monocyte-induced neural stem cell proliferation and differentiation. The model combined monocytes isolated from healthy donors and human embryonic stem cell derived neural stem cells and integrated both cell-intrinsic and -extrinsic properties. We identified CNS-mimicking culture media options that induced a monocytic phenotype resembling CNS infiltrating monocytes, while allowing adequate monocyte survival. Monocyte-induced proliferation, gliogenic fate and neurogenic fate of neural stem cells were affected by the conditions of monocytic priming and basal neural stem cell culture as extrinsic factors as well as the neural stem cell passage number as an intrinsic neural stem cell property. We developed a high-content compatible human in vitro assay for the integrated analysis of monocyte-derived factors on CNS repair.


Assuntos
Diferenciação Celular , Proliferação de Células , Monócitos , Células-Tronco Neurais , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Monócitos/citologia , Monócitos/metabolismo , Monócitos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas
2.
Front Cell Neurosci ; 17: 1156802, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37663126

RESUMO

A central issue in regenerative medicine is understanding the mechanisms that regulate the self-renewal of endogenous stem cells in response to injury and disease. Interferons increase hematopoietic stem cells during infection by activating STAT1, but the mechanisms by which STAT1 regulates intrinsic programs in neural stem cells (NSCs) during neuroinflammation is less known. Here we explored the role of STAT1 on NSC self-renewal. We show that overexpressing Stat1 in NSCs derived from the subventricular zone (SVZ) decreases NSC self-renewal capacity while Stat1 deletion increases NSC self-renewal, neurogenesis, and oligodendrogenesis in isolated NSCs. Importantly, we find upregulation of STAT1 in NSCs in a mouse model of multiple sclerosis (MS) and an increase in pathological T cells expressing IFN-γ rather than interleukin 17 (IL-17) in the cerebrospinal fluid of affected mice. We find IFN-γ is superior to IL-17 in reducing proliferation and precipitating an abnormal NSC phenotype featuring increased STAT1 phosphorylation and Stat1 and p16ink4a gene expression. Notably, Stat1-/- NSCs were resistant to the effect of IFN-γ. Lastly, we identified a Stat1-dependent gene expression profile associated with an increase in the Sox9 transcription factor, a regulator of self-renewal. Stat1 binds and transcriptionally represses Sox9 in a transcriptional luciferase assay. We conclude that Stat1 serves as an inducible checkpoint for NSC self-renewal that is upregulated during chronic brain inflammation leading to decreased self-renewal. As such, Stat1 may be a potential target to modulate for next generation therapies to prevent progression and loss of repair function in NSCs/neural progenitors in MS.

3.
Nat Commun ; 10(1): 217, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30644388

RESUMO

In demyelinating diseases including multiple sclerosis (MS), neural stem cells (NSCs) can replace damaged oligodendrocytes if the local microenvironment supports the required differentiation process. Although chitinase-like proteins (CLPs) form part of this microenvironment, their function in this differentiation process is unknown. Here, we demonstrate that murine Chitinase 3-like-3 (Chi3l3/Ym1), human Chi3L1 and Chit1 induce oligodendrogenesis. In mice, Chi3l3 is highly expressed in the subventricular zone, a stem cell niche of the adult brain, and in inflammatory brain lesions during experimental autoimmune encephalomyelitis (EAE). We find that silencing Chi3l3 increases severity of EAE. We present evidence that in NSCs Chi3l3 activates the epidermal growth factor receptor (EGFR), thereby inducing Pyk2-and Erk1/2- dependent expression of a pro-oligodendrogenic transcription factor signature. Our results implicate CLP-EGFR-Pyk2-MEK-ERK as a key intrinsic pathway controlling oligodendrogenesis.


Assuntos
Encefalomielite Autoimune Experimental/etiologia , Receptores ErbB/metabolismo , Lectinas/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Animais , Proteína 1 Semelhante à Quitinase-3/metabolismo , Feminino , Células HEK293 , Hexosaminidases/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos
4.
Circ Res ; 117(9): 779-92, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26294656

RESUMO

RATIONALE: Platelets are known to participate in vascular pathologies; however, their role in neuroinflammatory diseases, such as multiple sclerosis (MS), is unknown. Autoimmune CD4 T cells have been the main focus of studies of MS, although the factors that regulate T-cell differentiation toward pathogenic T helper-1/T helper-17 phenotypes are not completely understood. OBJECTIVE: We investigated the role of platelets in the modulation of CD4 T-cell functions in patients with MS and in mice with experimental autoimmune encephalitis, an animal model for MS. METHODS AND RESULTS: We found that early in MS and experimental autoimmune encephalitis, platelets degranulated and produced soluble factors serotonin (5-hydroxytryptamine), platelet factor 4, and platelet-activating factor, which specifically stimulated differentiation of T cells toward pathogenic T helper-1, T helper-17, and interferon-γ/interleukin-17-producing CD4 T cells. At the later stages of MS and experimental autoimmune encephalitis, platelets became exhausted in their ability to produce proinflammatory factors and stimulate CD4 T cells but substantially increased their ability to form aggregates with CD4 T cells. Formation of platelet-CD4 T-cell aggregates involved the interaction of CD62P on activated platelets with adhesion molecule CD166 on activated CD4 T cells, contributing to downmodulation of CD4 T-cell activation, proliferation, and production of interferon-γ. Blocking of formation of platelet-CD4 T-cell aggregates during progression of experimental autoimmune encephalitis substantially enhanced proliferation of CD4 T cells in the central nervous system and the periphery leading to exacerbation of the disease. CONCLUSION: Our study indicates differential roles for platelets in the regulation of functions of pathogenic CD4 T cells during initiation and progression of central nervous system autoimmune inflammation.


Assuntos
Plaquetas/imunologia , Linfócitos T CD4-Positivos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Adulto , Animais , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/ultraestrutura , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Progressão da Doença , Feminino , Citometria de Fluxo , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Fator de Ativação de Plaquetas/imunologia , Fator de Ativação de Plaquetas/metabolismo , Fator Plaquetário 4/imunologia , Fator Plaquetário 4/metabolismo , Serotonina/imunologia , Serotonina/metabolismo
5.
Glycobiology ; 25(12): 1392-409, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26153105

RESUMO

Neural stem cell (NSC)-based therapies offer potential for neural repair in central nervous system (CNS) inflammatory and degenerative disorders. Typically, these conditions present with multifocal CNS lesions making it impractical to inject NSCs locally, thus mandating optimization of vascular delivery of the cells to involved sites. Here, we analyzed NSCs for expression of molecular effectors of cell migration and found that these cells are natively devoid of E-selectin ligands. Using glycosyltransferase-programmed stereosubstitution (GPS), we glycan engineered the cell surface of NSCs ("GPS-NSCs") with resultant enforced expression of the potent E-selectin ligand HCELL (hematopoietic cell E-/L-selectin ligand) and of an E-selectin-binding glycoform of neural cell adhesion molecule ("NCAM-E"). Following intravenous (i.v.) injection, short-term homing studies demonstrated that, compared with buffer-treated (control) NSCs, GPS-NSCs showed greater neurotropism. Administration of GPS-NSC significantly attenuated the clinical course of experimental autoimmune encephalomyelitis (EAE), with markedly decreased inflammation and improved oligodendroglial and axonal integrity, but without evidence of long-term stem cell engraftment. Notably, this effect of NSC is not a universal property of adult stem cells, as administration of GPS-engineered mouse hematopoietic stem/progenitor cells did not improve EAE clinical course. These findings highlight the utility of cell surface glycan engineering to boost stem cell delivery in neuroinflammatory conditions and indicate that, despite the use of a neural tissue-specific progenitor cell population, neural repair in EAE results from endogenous repair and not from direct, NSC-derived cell replacement.


Assuntos
Movimento Celular , Encefalomielite Autoimune Experimental/terapia , Células-Tronco Neurais/metabolismo , Polissacarídeos/metabolismo , Animais , Terapia Genética , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regeneração Nervosa , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/transplante , Selectinas/metabolismo
6.
PLoS One ; 9(5): e96256, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24788965

RESUMO

BACKGROUND: Glatiramer acetate (GA, Copaxone, Copolymer-1) is an FDA approved drug for the treatment of MS and it is very effective in suppressing neuroinflammation in experimental autoimmune encephalitis (EAE), an animal model of MS. Although this drug was designed to inhibit pathogenic T cells, the exact mechanism of EAE/MS suppression by GA is still not well understood. Previously we presented evidence that platelets become activated and promote neuroinflammation in EAE, suggesting a possible pathogenic role of platelets in MS and EAE. We hypothesized that GA could inhibit neuroinflammation by affecting not only immune cells but also platelets. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the effect of GA on the activation of human platelets in vitro: calcium influx, platelet aggregation and expression of activation markers. Our results in human platelets were confirmed by in-vitro and in-vivo studies of modulation of functions of platelets in mouse model. We found that GA inhibited thrombin-induced calcium influx in human and mouse platelets. GA also decreased thrombin-induced CD31, CD62P, CD63, and active form of αIIbß3 integrin surface expression and formation of platelet aggregates for both mouse and human platelets, and prolonged the bleeding time in mice by 2.7-fold. In addition, we found that GA decreased the extent of macrophage activation induced by co-culture of macrophages with platelets. CONCLUSIONS: GA inhibited the activation of platelets, which suggests a new mechanism of GA action in suppression of EAE/MS by targeting platelets and possibly preventing their interaction with immune cells such as macrophages. Furthermore, the reduction in platelet activation by GA may have additional cardiovascular benefits to prevent thrombosis.


Assuntos
Cálcio/metabolismo , Peptídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Trombina/farmacologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígeno B7-2/metabolismo , Tempo de Sangramento , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Células Cultivadas , Técnicas de Cocultura , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/prevenção & controle , Acetato de Glatiramer , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Transporte de Íons/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/prevenção & controle , Selectina-P/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo
8.
PLoS One ; 8(3): e58979, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555611

RESUMO

Platelets respond to vascular damage and contribute to inflammation, but their role in the neurodegenerative diseases is unknown. We found that the systemic administration of brain lipid rafts induced a massive platelet activation and degranulation resulting in a life-threatening anaphylactic-like response in mice. Platelets were engaged by the sialated glycosphingolipids (gangliosides) integrated in the rigid structures of astroglial and neuronal lipid rafts. The brain-abundant gangliosides GT1b and GQ1b were specifically recognized by the platelets and this recognition involved multiple receptors with P-selectin (CD62P) playing the central role. During the neuroinflammation, platelets accumulated in the central nervous system parenchyma, acquired an activated phenotype and secreted proinflammatory factors, thereby triggering immune response cascades. This study determines a new role of platelets which directly recognize a neuronal damage and communicate with the cells of the immune system in the pathogenesis of neurodegenerative diseases.


Assuntos
Plaquetas/metabolismo , Encéfalo/metabolismo , Glicolipídeos/metabolismo , Microdomínios da Membrana/metabolismo , Anafilaxia/imunologia , Anafilaxia/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Transporte Biológico , Plaquetas/imunologia , Barreira Hematoencefálica/metabolismo , Encéfalo/imunologia , Degranulação Celular , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Gangliosídeos/imunologia , Glicolipídeos/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/imunologia , Camundongos , Neurônios/imunologia , Neurônios/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo
9.
J Vis Exp ; (65)2012 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-22872097

RESUMO

Microglia are cells of the myeloid lineage that reside in the central nervous system (CNS)(1). These cells play an important role in pathologies of many diseases associated with neuroinflammation such as multiple sclerosis (MS)(2). Microglia in a normal CNS express macrophage marker CD11b and exhibit a resting phenotype by expressing low levels of activation markers such as CD45. During pathological events in the CNS, microglia become activated as determined by upregulation of CD45 and other markers(3). The factors that affect microglia phenotype and functions in the CNS are not well studied. MicroRNAs (miRNAs) are a growing family of conserved molecules (~22 nucleotides long) that are involved in many normal physiological processes such as cell growth and differentiation(4) and pathologies such as inflammation(5). MiRNAs downregulate the expression of certain target genes by binding complementary sequences of their mRNAs and play an important role in the activation of innate immune cells including macrophages(6) and microglia(7). In order to investigate miRNA-mediated pathways that define the microglial phenotype, biological function, and to distinguish microglia from other types of macrophages, it is important to quantitatively assess the expression of particular microRNAs in distinct subsets of CNS-resident microglia. Common methods for measuring the expression of miRNAs in the CNS include quantitative PCR from whole neuronal tissue and in situ hybridization. However, quantitative PCR from whole tissue homogenate does not allow the assessment of the expression of miRNA in microglia, which represent only 5-15% of the cells of neuronal tissue. Hybridization in situ allows the assessment of the expression of microRNA in specific cell types in the tissue sections, but this method is not entirely quantitative. In this report we describe a quantitative and sensitive method for the detection of miRNA by real-time PCR in microglia isolated from normal CNS or during neuroinflammation using experimental autoimmune encephalomyelitis (EAE), a mouse model for MS. The described method will be useful to measure the level of expression of microRNAs in microglia in normal CNS or during neuroinflammation associated with various pathologies including MS, stroke, traumatic injury, Alzheimer's disease and brain tumors.


Assuntos
Sistema Nervoso Central/química , Encefalomielite Autoimune Experimental/genética , MicroRNAs/análise , Microglia/química , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Hibridização In Situ/métodos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , MicroRNAs/genética , Microglia/metabolismo , Microglia/patologia , Microglia/fisiologia
10.
Immunity ; 37(2): 249-63, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22884314

RESUMO

Inflammation-mediated neurodegeneration occurs in the acute and the chronic phases of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Classically activated (M1) microglia are key players mediating this process. Here, we identified Galectin-1 (Gal1), an endogenous glycan-binding protein, as a pivotal regulator of M1 microglial activation that targets the activation of p38MAPK-, CREB-, and NF-κB-dependent signaling pathways and hierarchically suppresses downstream proinflammatory mediators, such as iNOS, TNF, and CCL2. Gal1 bound to core 2 O-glycans on CD45, favoring retention of this glycoprotein on the microglial cell surface and augmenting its phosphatase activity and inhibitory function. Gal1 was highly expressed in the acute phase of EAE, and its targeted deletion resulted in pronounced inflammation-induced neurodegeneration. Adoptive transfer of Gal1-secreting astrocytes or administration of recombinant Gal1 suppressed EAE through mechanisms involving microglial deactivation. Thus, Gal1-glycan interactions are essential in tempering microglial activation, brain inflammation, and neurodegeneration, with critical therapeutic implications for MS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Galectina 1/imunologia , Antígenos Comuns de Leucócito/metabolismo , Microglia/imunologia , Animais , Astrócitos/metabolismo , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Quimiocina CCL2/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/terapia , Feminino , Galectina 1/metabolismo , Galectina 1/uso terapêutico , Humanos , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/citologia , Microglia/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
PLoS One ; 6(8): e23618, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21886804

RESUMO

BACKGROUND: Type 1 regulatory T (Tr1) cells, characterized by the secretion of high levels of the anti-inflammatory cytokine interleukin-10 (IL-10), play an important role in the regulation of autoimmune diseases and transplantation. However, effective strategies that specifically induce Tr1 cells in vivo are limited. Furthermore, the pathways controlling the induction of these cells in vivo are not well understood. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that nasal administration of anti-CD3 antibody induces suppressive Tr1 cells in mice. The in vivo induction of Tr1 cells by nasal anti-CD3 is dependent on IL-27 produced by upper airway resident dendritic cells (DCs), and is controlled by the transcription factors aryl hydrocarbon receptor (AHR) and c-Maf. Subsequently, IL-21 acts in an autocrine fashion to expand and maintain the Tr1 cells induced in vivo by nasally administered anti-CD3. CONCLUSIONS/SIGNIFICANCE: Our findings identify a unique approach to generate Tr1 cells in vivo and provide insights into the mechanisms by which these cells are induced.


Assuntos
Células Dendríticas/imunologia , Mucosa Nasal/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Administração Intranasal , Animais , Anticorpos/administração & dosagem , Anticorpos/farmacologia , Autoimunidade/efeitos dos fármacos , Complexo CD3/imunologia , Células Dendríticas/efeitos dos fármacos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Camundongos , Modelos Imunológicos , Mucosa Nasal/efeitos dos fármacos , Receptores de Interleucina-21/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
12.
Ann Neurol ; 69(5): 878-91, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21391234

RESUMO

OBJECTIVE: The subventricular zone (SVZ) of the brain constitutes a niche for neural stem and progenitor cells that can initiate repair after central nervous system (CNS) injury. In a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE), the neural stem cells (NSCs) become activated and initiate regeneration during acute disease, but lose this ability during the chronic phases of disease. We hypothesized that chronic microglia activation contributes to the failure of the NSC repair potential in the SVZ. METHODS: Using bromodeoxyuridine injections at different time points during EAE, we quantified the number of proliferating and differentiating progenitors, and evaluated the structure of the SVZ by electron microscopy. In vivo minocycline treatment during EAE was used to address the effect of microglia inactivation on SVZ dysfunction. RESULTS: In vivo treatment with minocycline, an inhibitor of microglia activation, increases stem cell proliferation in both naive and EAE animals. Minocycline treatment decreases cortical and periventricular pathology in the chronic phase of EAE, improving the proliferation of Sox2 stem cells and NG2 oligodendrocyte precursors cells originating in the SVZ and their differentiation into mature oligodendrocytes. INTERPRETATION: These data suggest that failure of repair observed during chronic EAE correlates with microglia activation and that treatments targeting chronic microglial activation have the potential for enhancing repair in the CNS.


Assuntos
Encefalomielite Autoimune Experimental/patologia , Células-Tronco Neurais/fisiologia , Nicho de Células-Tronco/patologia , Animais , Antibacterianos/farmacologia , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/fisiopatologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Microglia/ultraestrutura , Microscopia Eletrônica de Transmissão , Minociclina/farmacologia , Esclerose Múltipla , Proteína Proteolipídica de Mielina/efeitos adversos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/ultraestrutura , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Fragmentos de Peptídeos/efeitos adversos , Prevenção Secundária , Nicho de Células-Tronco/efeitos dos fármacos , Fatores de Tempo
13.
Brain Behav Immun ; 25(5): 991-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21074605

RESUMO

Microglia play an important role in inflammatory diseases of the central nervous system. There is evidence of microglial diversity with distinct phenotypes exhibiting either neuroprotection and repair or neurotoxicity. However the precise molecular mechanisms underlying this diversity are still unknown. Using a model of experimental autoimmune encephalomyelitis (EAE) we performed transcriptional profiling of isolated subventricular zone microglia from the acute and chronic disease phases of EAE. We found that microglia exhibit disease phase specific gene expression signatures, that correspond to unique gene ontology functions and genomic networks. Our data demonstrate for the first time, distinct transcriptional networks of microglia activation in vivo, that suggests a role as mediators of injury or repair.


Assuntos
Redes Reguladoras de Genes/fisiologia , Microglia/fisiologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/genética , Camundongos , Microglia/metabolismo , Microglia/patologia , Microscopia Confocal , Análise de Sequência com Séries de Oligonucleotídeos , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA