Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 14: 66, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32296307

RESUMEN

Microglia are constantly surveying their microenvironment and rapidly react to impairments by changing their morphology, migrating toward stimuli and adopting gene expression profiles characterizing their activated state. The increased expression of the M2-like marker Mannose receptor 1 (Mrc1), which is also referred to as CD206, in microglia has been reported after M2-like activation in vitro and in vivo. Mrc1 is a 175-kDa transmembrane pattern recognition receptor which binds a variety of carbohydrates and is involved in the pinocytosis and the phagocytosis of immune cells, including microglia, and thought to contribute to a neuroprotective microglial phenotype. Here we analyzed the effects of TGFß signaling on Mrc1 expression in microglia in vivo and in vitro. Using C57BL/6 wild type and Cx3cr1 CreERT2 :R26-YFP:Tgfbr2 fl/fl mice-derived microglia, we show that the silencing of TGFß signaling results in the upregulation of Mrc1, whereas recombinant TGFß1 induced the delayed downregulation of Mrc1. Furthermore, chromatin immunoprecipitation experiments provided evidence that Mrc1 is not a direct Smad2/Smad4 target gene in microglia. Altogether our data indicate that the changes in Mrc1 expression after the activation or the silencing of microglial TGFß signaling are likely to be mediated by modifications of the secondary intracellular signaling events influenced by TGFß signaling.

2.
Nat Commun ; 9(1): 4011, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30275444

RESUMEN

TGFß1 has been implicated in regulating functional aspects of several distinct immune cell populations including central nervous system (CNS) resident microglia. Activation and priming of microglia have been demonstrated to contribute to the progression of neurodegenerative diseases and, thus, underlie stringent control by endogenous regulatory factors including TGFß1. Here, we demonstrate that deletion of Tgfbr2 in adult postnatal microglia does neither result in impairment of the microglia-specific gene expression signatures, nor is microglial survival and maintenance affected. Tgfbr2-deficient microglia were characterised by distinct morphological changes and transcriptome analysis using RNAseq revealed that loss of TGFß signalling results in upregulation of microglia activation and priming markers. Moreover, protein arrays demonstrated increased secretion of CXCL10 and CCL2 accompanied by activation of immune cell signalling as evidenced by increased phosphorylation of TAK1. Together, these data underline the importance of microglial TGFß signalling to regulate microglia adaptive changes.


Asunto(s)
Homeostasis , Microglía/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/metabolismo , Perfilación de la Expresión Génica , Silenciador del Gen , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Transgénicos , Microglía/citología , Fosforilación , Análisis por Matrices de Proteínas , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética
3.
Front Immunol ; 9: 1728, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30093905

RESUMEN

Microglia maturation takes place during the postnatal weeks and is characterized by the establishment of a unique microglia-specific gene expression pattern. Tmem119, Fcrls, Hexb, and Olfml3 have been identified among these microglia-specific genes. Transforming growth factor ß1 (TGFß1) has been reported as a critical factor for microglia maturation and maintenance and active TGFß signaling precedes the inductions of microglial gene expression. In this study, we demonstrate Olfml3 expression in adult microglia and further provide evidence that TGFß1 induces upregulation of Olfml3 expression in postnatal microglia. Using chromatin immunoprecipitation and microglia-specific silencing of TGFß signaling in vitro and in vivo, we in clearly show that Olfml3 is a direct TGFß1/Smad2 target gene. Together, our data underline the importance of TGFß1 as a critical regulator of microglia functions and microglia maturation and further broaden our understanding of TGFß1-mediated effects on the resident immune cells of the central nervous system.


Asunto(s)
Regulación de la Expresión Génica , Microglía/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Sitios de Unión , Ratones , Especificidad de Órganos , Unión Proteica , Transporte de Proteínas
4.
Int J Mol Sci ; 19(3)2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29494550

RESUMEN

Microglia are the resident immune cells of the central nervous system (CNS) and participate in physiological and pathological processes. Their unique developmental nature suggests age-dependent structural and functional impairments that might contribute to neurodegenerative diseases. In the present study, we addressed the age-dependent changes in cortical microglia gene expression patterns and the expression of M1- and M2-like activation markers. Iba1 immunohistochemistry, isolation of cortical microglia followed by fluorescence-activated cell sorting and RNA isolation to analyze transcriptional changes in aged cortical microglia was performed. We provide evidence that aging is associated with decreased numbers of cortical microglia and the establishment of a distinct microglia activation profile including upregulation of Ifi204, Lilrb4, Arhgap, Oas1a, Cd244 and Ildr2. Moreover, flow cytometry revealed that aged cortical microglia express increased levels of Cd206 and Cd36. The data presented in the current study indicate that aged mouse cortical microglia adopt a distinct activation profile, which suggests immunosuppressive and immuno-tolerogenic functions.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/inmunología , Tolerancia Inmunológica , Microglía/inmunología , Microglía/metabolismo , Factores de Edad , Envejecimiento/inmunología , Animales , Biomarcadores , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Ratones , Transcriptoma
5.
Glia ; 66(8): 1695-1708, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29575117

RESUMEN

Microglia are involved in a widespread set of physiological and pathological processes and further play important roles during neurodevelopmental events. Postnatal maturation of microglia has been associated with the establishment of microglia-specific gene expression patterns. The mechanisms governing microglia maturation are only partially understood but Tgfß1 has been suggested to be one important mediator. In the present study, we demonstrate that early postnatal microglia maturation is associated with alternative microglia activation, increased engulfment of apoptotic cells as well as activated microglial Tgfß signaling. Interestingly, microglial Tgfß signaling preceded the induction of the microglia-specific gene expression indicating the importance of Tgfß1 for postnatal microglia maturation. Moreover, we provide evidence that Tgfß1 is expressed by neurons in postnatal and adult brains defining neuron-microglia communication via Tgfß1 as an important event. Finally, we introduce the recently identified microglia marker Tmem119 as a direct Tgfß1-Smad2 target gene. Taken together, the data presented here further increase the understanding of Tgfß1-mediated effects in microglia and place emphasis on the importance of Tgfß1 for microglia maturation and maintenance.


Asunto(s)
Encéfalo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Animales Recién Nacidos , Macrófagos/metabolismo , Ratones Noqueados , Transducción de Señal/efectos de los fármacos
6.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096319

RESUMEN

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Asunto(s)
Encéfalo/citología , Quimiocina CXCL10/inmunología , Trastornos del Conocimiento/genética , Células Endoteliales/inmunología , Células Epiteliales/inmunología , Conducta de Enfermedad/fisiología , Receptor de Interferón alfa y beta/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Encéfalo/inmunología , Comunicación Celular/inmunología , Células Cultivadas , Trastornos del Conocimiento/psicología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Endotelio/citología , Endotelio/inmunología , Epitelio/inmunología , Interferón Tipo I/uso terapéutico , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , ARN Bicatenario/genética , Receptor de Interferón alfa y beta/inmunología , Receptores CXCR3/inmunología , Transducción de Señal/inmunología , Virosis/inmunología
7.
Int J Mol Sci ; 17(2)2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26821015

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease characterised by histopathological and biochemical manifestations such as loss of midbrain dopaminergic (DA) neurons and decrease in dopamine levels accompanied by a concomitant neuroinflammatory response in the affected brain regions. Over the past decades, the use of toxin-based animal models has been crucial to elucidate disease pathophysiology, and to develop therapeutic approaches aimed to alleviate its motor symptoms. Analyses of transgenic mice deficient for cytokines, chemokine as well as neurotrophic factors and their respective receptors in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD have broadened the current knowledge of neuroinflammation and neurotrophic support. Here, we provide a comprehensive review that summarises the contribution of microglia-mediated neuroinflammation in MPTP-induced neurodegeneration. Moreover, we highlight the contribution of neurotrophic factors as endogenous and/or exogenous molecules to slow the progression of midbrain dopaminergic (mDA) neurons and further discuss the potential of combined therapeutic approaches employing neuroinflammation modifying agents and neurotrophic factors.


Asunto(s)
Modelos Animales de Enfermedad , Intoxicación por MPTP/inmunología , Microglía/patología , Factores de Crecimiento Nervioso/metabolismo , Enfermedad de Parkinson/inmunología , Animales , Humanos , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/patología , Ratones , Ratones Transgénicos , Microglía/inmunología , Factores de Crecimiento Nervioso/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Receptores de Factor de Crecimiento Nervioso/metabolismo
8.
J Neurochem ; 134(1): 125-34, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25827682

RESUMEN

Microglia-mediated neuroinflammation has been reported as a common feature of familial and sporadic forms of Parkinson's disease (PD), and a growing body of evidence indicates that onset and progression of PD correlates with the extent of neuroinflammatory responses involving Interferon γ (IFNγ). Transforming growth factor ß1 (TGFß1) has been shown to be a major player in the regulation of microglia activation states and functions and, thus, might be a potential therapeutic agent by shaping microglial activation phenotypes during the course of neurodegenerative diseases such as PD. In this study, we demonstrate that TGFß1 is able to block IFNγ-induced microglia activation by attenuating STAT1 phosphorylation and IFNγRα expression. Moreover, we identified a set of genes involved in microglial IFNγ signaling transduction that were significantly down-regulated upon TGFß1 treatment, resulting in decreased sensitivity of microglia toward IFNγ stimuli. Interestingly, genes mediating negative regulation of IFNγ signaling, such as SOCS2 and SOCS6, were up-regulated after TGFß1 treatment. Finally, we demonstrate that TGFß1 is capable of protecting midbrain dopaminergic (mDA) neurons from IFNγ-driven neurotoxicity in mixed neuron-glia cultures derived from embryonic day 14 (E14) midbrain tissue. Together, these data underline the importance of TGFß1 as a key immunoregulatory factor for microglia by silencing IFNγ-mediated microglia activation and, thereby, rescuing mDA neurons from IFNγ-induced neurotoxicity. Interferon γ (IFNγ) is a potent pro-inflammatory factor that triggers the activation of microglia and the subsequent release of neurotoxic factors. Transforming growth factor ß1 (TGFß1) is able to inhibit the IFNγ-mediated activation of microglia, which is characterized by the release of nitric oxide (NO) and tumor necrosis factor α (TNFα). By decreasing the expression of IFNγ-induced genes as well as the signaling receptor IFNγR1, TGFß1 reduces the responsiveness of microglia towards IFNγ. In mixed neuron-glia cultures, TGFß1 protects midbrain dopaminergic (mDA) neurons from IFNγ-induced neurotoxicity.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Interferón gamma/antagonistas & inhibidores , Interferón gamma/toxicidad , Microglía/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Crecimiento Transformador beta1/farmacología , Animales , Animales Recién Nacidos , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología
9.
Glia ; 63(1): 142-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25130376

RESUMEN

Milk fat globule-epidermal growth factor-factor 8 (Mfge8) has been described as an essential molecule during microglia-mediated clearance of apoptotic cells via binding to phosphatidylserine residues and subsequent phagocytosis. Impaired uptake of apoptotic cells by microglia results in prolonged inflammatory responses and damage of healthy cells. Although the mechanisms of Mfge8-mediated engulfment of apoptotic cells are well understood, endogenous or exogenous factors that regulate Mfge8 expression remain elusive. Here, we describe that TGFß1 increases the expression of Mfge8 and enhances the engulfment of apoptotic cells by primary mouse microglia in a Mfge8-dependent manner. Further, apoptotic cells are capable of increasing microglial TGFß expression and release and shift the microglia phenotype toward alternative activation. Moreover, we provide evidence that Mfge8 expression is differentially regulated in microglia after classical and alternative activation and that Mfge8 is not able to exert direct antiinflammatory effects on LPS-treated primary microglia. Together, these results underline the importance of TGFß1 as a regulatory factor for microglia and suggest that increased TGFß1 expression in models of neurodegeneration might be involved in clearance of apoptotic cells via regulation of Mfge8 expression.


Asunto(s)
Antígenos de Superficie/metabolismo , Apoptosis/fisiología , Glucolípidos/metabolismo , Glicoproteínas/metabolismo , Microglía/metabolismo , Proteínas de la Leche/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Gotas Lipídicas , Ratones Endogámicos C57BL , Fagocitosis/genética , Activación Transcripcional/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...