Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38986723

RESUMEN

Microglia are increasingly recognized to contribute to brain health and disease. Preclinical studies using laboratory rodents are essential to advance our understanding of the physiological and pathophysiological functions of these cells in the central nervous system. Rodents are nocturnal animals, and they are mostly maintained in a defined light-dark cycle within animal facilities, with many laboratories investigating microglial molecular and functional profiles during the animals' light (sleep) phase. However, only a few studies have considered possible differences in microglial functions between the active and sleep phases. Based on initial evidence suggesting that microglial intrinsic clock genes can affect their phenotypes, we sought to investigate differences in transcriptional, proteotype and functional profiles of microglia between light (sleep) and dark (active) phases, and how these changes are affected in pathological models. We found marked transcriptional and proteotype differences between microglia harvested from male mice during the light or dark phase. Amongst others, these differences related to genes and proteins associated with immune responses, motility, and phagocytosis, which were reflected by functional alterations in microglial synaptic pruning and response to bacterial stimuli. Possibly accounting for such changes, we found RNA and protein regulation in SWI/SNF and NuRD chromatin remodeling complexes between light and dark phases. Importantly, we also show that the time of microglial sample collection influences the nature of microglial transcriptomic changes in a model of immune-mediated neurodevelopmental disorders. Our findings emphasize the importance of considering diurnal factors in studying microglial cells and indicate that implementing a circadian perspective is pivotal for advancing our understanding of their physiological and pathophysiological roles in brain health and disease.

2.
PLoS One ; 19(7): e0302376, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38990806

RESUMEN

We applied the patch-seq technique to harvest transcripts from individual microglial cells from cortex, hippocampus and corpus callosum of acute brain slices from adult mice. After recording membrane currents with the patch-clamp technique, the cytoplasm was collected via the pipette and underwent adapted SMART-seq2 preparation with subsequent sequencing. On average, 4138 genes were detected in 113 cells from hippocampus, corpus callosum and cortex, including microglia markers such as Tmem119, P2ry12 and Siglec-H. Comparing our dataset to previously published single cell mRNA sequencing data from FACS-isolated microglia indicated that two clusters of cells were absent in our patch-seq dataset. Pathway analysis of marker genes in FACS-specific clusters revealed association with microglial activation and stress response. This indicates that under normal conditions microglia in situ lack transcripts associated with a stress-response, and that the microglia-isolation procedure by mechanical dissociation and FACS triggers the expression of genes related to activation and stress.


Asunto(s)
Microglía , Microglía/metabolismo , Animales , Ratones , Citometría de Flujo/métodos , Estrés Fisiológico/genética , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Masculino , Hipocampo/metabolismo , Hipocampo/citología , Análisis de la Célula Individual/métodos
3.
Acta Neuropathol Commun ; 12(1): 50, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566120

RESUMEN

Tumor-associated microglia and blood-derived macrophages (TAMs) play a central role in modulating the immune suppressive microenvironment in glioma. Here, we show that GPNMB is predominantly expressed by TAMs in human glioblastoma multiforme and the murine RCAS-PDGFb high grade glioma model. Loss of GPNMB in the in vivo tumor microenvironment results in significantly smaller tumor volumes and generates a pro-inflammatory innate and adaptive immune cell microenvironment. The impact of host-derived GPNMB on tumor growth was confirmed in two distinct murine glioma cell lines in organotypic brain slices from GPNMB-KO and control mice. Using published data bases of human glioma, the elevated levels in TAMs could be confirmed and the GPNMB expression correlated with a poorer survival.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Humanos , Ratones , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/patología , Glioma/patología , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Microambiente Tumoral
4.
Brain Behav Immun ; 119: 465-481, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38552926

RESUMEN

Microglia modulate synaptic refinement in the central nervous system (CNS). We have previously shown that a mouse model with innate high anxiety-related behavior (HAB) displays higher CD68+ microglia density in the key regions of anxiety circuits compared to mice with normal anxiety-related behavior (NAB) in males, and that minocycline treatment attenuated the enhanced anxiety of HAB male. Given that a higher prevalence of anxiety is widely reported in females compared to males, little is known concerning sex differences at the cellular level. Herein, we address this by analyzing microglia heterogeneity and function in the HAB and NAB brains of both sexes. Single-cell RNA sequencing revealed ten distinct microglia clusters varied by their frequency and gene expression profile. We report striking sex differences, especially in the major microglia clusters of HABs, indicating a higher expression of genes associated with phagocytosis and synaptic engulfment in the female compared to the male. On a functional level, we show that female HAB microglia engulfed a greater amount of hippocampal vGLUT1+ excitatory synapses compared to the male. We moreover show that female HAB microglia engulfed more synaptosomes compared to the male HAB in vitro. Due to previously reported effects of minocycline on microglia, we finally administered oral minocycline to HABs of both sexes and showed a significant reduction in the engulfment of synapses by female HAB microglia. In parallel to our microglia-specific findings, we further showed an anxiolytic effect of minocycline on female HABs, which is complementary to our previous findings in the male HABs. Our study, therefore, identifies the altered function of synaptic engulfment by microglia as a potential avenue to target and resolve microglia heterogeneity in mice with innate high anxiety.


Asunto(s)
Ansiedad , Microglía , Minociclina , Caracteres Sexuales , Animales , Minociclina/farmacología , Microglía/metabolismo , Microglía/efectos de los fármacos , Femenino , Ansiedad/metabolismo , Ansiedad/tratamiento farmacológico , Masculino , Ratones , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Ratones Endogámicos C57BL , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Modelos Animales de Enfermedad , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Fagocitosis/efectos de los fármacos
5.
eNeuro ; 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38388443

RESUMEN

The type I transmembrane protein BT-IgSF is predominantly localized in the brain and testes. It belongs to the CAR subgroup of Ig cell adhesion proteins, that are hypothesized to regulate connexin expression or localization. Here, we studied the putative link between BT-IgSF and connexins in astrocytes, ependymal cells and neurons of the mouse. Global knockout of BT-IgSF caused an increase in the clustering of connexin43 (Gja1), but not of connexin30 (Gjb6), on astrocytes and ependymal cells. Additionally, knockout animals displayed reduced expression levels of connexin43 protein in the cortex and hippocampus. Importantly, analysis of biocytin spread in hippocampal or cortical slices from mature mice of either sex revealed a decrease in astrocytic cell-cell coupling in the absence of BT-IgSF. Blocking either protein biosynthesis or proteolysis showed that the lysosomal pathway increased connexin43 degradation in astrocytes. Localization of connexin43 in subcellular compartments was not impaired in astrocytes of BT-IgSF mutants. In contrast to connexin43 the localization and expression of connexin36 (Gjd2) on neurons was not affected by the absence of BT-IgSF. Overall, our data indicate that the IgCAM BT-IgSF is essential for correct gap junction-mediated astrocyte-to-astrocyte cell communication.Significance Statement Astrocytes regulate a variety of physiological processes in the developing and adult brain that are essential for proper brain function. Astrocytes form extensive networks in the brain and communicate via gap junctions. Disruptions of gap junction coupling are found in several diseases such as neurodegeneration or epilepsy. Here, we demonstrate that the cell adhesion protein BT-IgSF is essential for gap junction mediated coupling between astrocytes in the cortex and hippocampus.

6.
Dis Model Mech ; 16(12)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37990867

RESUMEN

Neurofibromatosis type 1 (NF1) is an autosomal dominant condition caused by germline mutations in the neurofibromin 1 (NF1) gene. Children with NF1 are prone to the development of multiple nervous system abnormalities, including autism and brain tumors, which could reflect the effect of NF1 mutation on microglia function. Using heterozygous Nf1-mutant mice, we previously demonstrated that impaired purinergic signaling underlies deficits in microglia process extension and phagocytosis in situ. To determine whether these abnormalities are also observed in human microglia in the setting of NF1, we leveraged an engineered isogenic series of human induced pluripotent stem cells to generate human microglia-like (hiMGL) cells heterozygous for three different NF1 gene mutations found in patients with NF1. Whereas all NF1-mutant and isogenic control hiMGL cells expressed classical microglia markers and exhibited similar transcriptomes and cytokine/chemokine release profiles, only NF1-mutant hiMGL cells had defects in P2X receptor activation, phagocytosis and motility. Taken together, these findings indicate that heterozygous NF1 mutations impair a subset of the functional properties of human microglia, which could contribute to the neurological abnormalities seen in children with NF1.


Asunto(s)
Células Madre Pluripotentes Inducidas , Neurofibromatosis 1 , Animales , Humanos , Ratones , Genes de Neurofibromatosis 1 , Microglía/patología , Mutación/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética
7.
Glia ; 71(11): 2679-2695, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37641212

RESUMEN

Triggering receptor expressed on myeloid cell 2 (TREM2), a myeloid cell-specific signaling molecule, controls essential functions of microglia and impacts on the pathogenesis of Alzheimer's disease and other neurodegenerative disorders. TREM2 is also highly expressed in tumor-associated macrophages in different types of cancer. Here, we studied whether TREM2 influences glioma progression. We found a gender-dependent effect of glioma growth in wild-type (WT) animals injected with GL261-EGFP glioma cells. Most importantly, TREM2 promotes glioma progression in male but not female animals. The accumulation of glioma-associated microglia/macrophages (GAMs) and CD31+ blood vessel density is reduced in male TREM2-deficient mice. A transcriptomic analysis of glioma tissue revealed that TREM2 deficiency suppresses immune-related genes. In an organotypic slice model devoid of functional vascularization and immune components from periphery, the tumor size was not affected by TREM2-deficiency. In human resection samples from glioblastoma, TREM2 is upregulated in GAMs. Based on the Cancer Genome Atlas Program (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases, the TREM2 expression levels were negatively correlated with survival. Thus, the TREM2-dependent crosstalk between GAMs and the vasculature formation promotes glioma growth.


Asunto(s)
Glioblastoma , Glioma , Humanos , Masculino , Animales , Ratones , Microglía , Macrófagos , Encéfalo , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
8.
Brain Behav Immun ; 111: 61-75, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37001827

RESUMEN

Neuroligin-4 (NLGN4) loss-of-function mutations are associated with monogenic heritable autism spectrum disorder (ASD) and cause alterations in both synaptic and behavioral phenotypes. Microglia, the resident CNS macrophages, are implicated in ASD development and progression. Here we studied the impact of NLGN4 loss in a mouse model, focusing on microglia phenotype and function in both male and female mice. NLGN4 depletion caused lower microglia density, less ramified morphology, reduced response to injury and purinergic signaling specifically in the hippocampal CA3 region predominantly in male mice. Proteomic analysis revealed disrupted energy metabolism in male microglia and provided further evidence for sexual dimorphism in the ASD associated microglial phenotype. In addition, we observed impaired gamma oscillations in a sex-dependent manner. Lastly, estradiol application in male NLGN4-/- mice restored the altered microglial phenotype and function. Together, these results indicate that loss of NLGN4 affects not only neuronal network activity, but also changes the microglia state in a sex-dependent manner that could be targeted by estradiol treatment.


Asunto(s)
Trastorno del Espectro Autista , Masculino , Femenino , Animales , Ratones , Trastorno del Espectro Autista/genética , Microglía , Ratones Noqueados , Proteómica , Neuronas/fisiología
9.
Acta Neuropathol Commun ; 11(1): 36, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36890585

RESUMEN

We previously discovered a sex-by-genotype defect in microglia function using a heterozygous germline knockout mouse model of Neurofibromatosis type 1 (Nf1 ± mice), in which only microglia from male Nf1 ± mice exhibited defects in purinergic signaling. Herein, we leveraged an unbiased proteomic approach to demonstrate that male, but not female, heterozygous Nf1 ± microglia exhibit differences in protein expression, which largely reflect pathways involved in cytoskeletal organization. In keeping with these predicted defects in cytoskeletal function, only male Nf1 ± microglia had reduced process arborization and surveillance capacity. To determine whether these microglial defects were cell autonomous or reflected adaptive responses to Nf1 heterozygosity in other cells in the brain, we generated conditional microglia Nf1-mutant knockout mice by intercrossing Nf1flox/flox with Cx3cr1-CreER mice (Nf1flox/wt; Cx3cr1-CreER mice, Nf1MG ± mice). Surprisingly, neither male nor female Nf1MG ± mouse microglia had impaired process arborization or surveillance capacity. In contrast, when Nf1 heterozygosity was generated in neurons, astrocytes and oligodendrocytes by intercrossing Nf1flox/flox with hGFAP-Cre mice (Nf1flox/wt; hGFAP-Cre mice, Nf1GFAP ± mice), the microglia defects found in Nf1 ± mice were recapitulated. Collectively, these data reveal that Nf1 ± sexually dimorphic microglia abnormalities are likely not cell-intrinsic properties, but rather reflect a response to Nf1 heterozygosity in other brain cells.


Asunto(s)
Neurofibromatosis 1 , Ratones , Masculino , Animales , Neurofibromatosis 1/genética , Microglía/metabolismo , Proteómica , Ratones Noqueados , Encéfalo/metabolismo
10.
PLoS One ; 18(2): e0278325, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36745631

RESUMEN

Microglia are the immune effector cells of the central nervous system (CNS) and react to pathologic events with a complex process including the release of nitric oxide (NO). NO is a free radical, which is toxic for all cells at high concentrations. To target an exaggerated NO release, we tested a library of 16 544 chemical compounds for their effect on lipopolysaccharide (LPS)-induced NO release in cell line and primary neonatal microglia. We identified a compound (C1) which significantly reduced NO release in a dose-dependent manner, with a low IC50 (252 nM) and no toxic side effects in vitro or in vivo. Target finding strategies such as in silico modelling and mass spectroscopy hint towards a direct interaction between C1 and the nitric oxide synthase making C1 a great candidate for specific intra-cellular interaction with the NO producing machinery.


Asunto(s)
Microglía , Óxido Nítrico , Recién Nacido , Humanos , Microglía/metabolismo , Óxido Nítrico/metabolismo , Enfermedades Neuroinflamatorias , Óxido Nítrico Sintasa de Tipo II/metabolismo , Línea Celular , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo
11.
Mol Syst Biol ; 19(2): e11147, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36573354

RESUMEN

Tissue dissociation, a crucial step in single-cell sample preparation, can alter the transcriptional state of a sample through the intrinsic cellular stress response. Here we demonstrate a general approach for measuring transcriptional response during sample preparation. In our method, transcripts made during dissociation are labeled for later identification upon sequencing. We found general as well as cell-type-specific dissociation response programs in zebrafish larvae, and we observed sample-to-sample variation in the dissociation response of mouse cardiomyocytes despite well-controlled experimental conditions. Finally, we showed that dissociation of the mouse hippocampus can lead to the artificial activation of microglia. In summary, our approach facilitates experimental optimization of dissociation procedures as well as computational removal of transcriptional perturbation response.


Asunto(s)
ARN , Transcriptoma , Ratones , Animales , Pez Cebra/genética , Análisis de Secuencia de ARN/métodos , Microglía , Análisis de la Célula Individual , Perfilación de la Expresión Génica/métodos
12.
Cell Rep ; 39(2): 110670, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417708

RESUMEN

Factors released from glioma-associated microglia/macrophages (GAMs) play a crucial role in glioblastoma multiforme (GBM) progression. Here, we study the importance of CCL18, a cytokine expressed in human but not in rodent GAMs, as a modulator of glioma growth. Since CCL18 signaling could not be studied in classical mouse glioma models, we developed an approach by transplanting induced pluripotent stem cell-derived human microglia and human glioma cells into mouse brain slices depleted of their intrinsic microglia. We observe that CCL18 promotes glioma cell growth and invasion. Chemokine (C-C motif) receptor 8 (CCR8) is identified as a functional receptor for CCL18 on glioma cells, and ACP5 (acid phosphatase 5) is revealed as an important part of the downstream signaling cascade for mediating glioma growth. We conclude, based on the results from an in vitro, ex vivo humanized glioma model and an in vivo GBM model that microglia/macrophage-derived CCL18 promotes glioma growth.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Línea Celular Tumoral , Quimiocinas CC , Humanos , Macrófagos , Ratones , Microglía , Receptores CCR8 , Fosfatasa Ácida Tartratorresistente
13.
J Neurosci Res ; 100(4): 1105-1122, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35213755

RESUMEN

Microglia are the innate immune cells of the central nervous system (CNS). In the adult uncompromised CNS, they have a highly ramified morphology and continuously extend and retract their processes. A subpopulation of microglial cells forms close soma-to-soma contacts with neurons and have been termed satellite microglia, yet the role of such interaction is largely unknown. Here, we analyzed the distribution of satellite microglia in different areas of the CNS of adult male mice applying transgenic- and immunolabeling of neuronal subtypes and microglia followed by three-dimensional imaging analysis. We quantified satellite microglia associated with GABAergic and glutamatergic neurons in the somatosensory cortex, striatum, and thalamus; with dopaminergic and serotonergic neurons in the basal forebrain and raphe nucleus, respectively; and with cerebellar Purkinje cell neurons. Satellite microglia in the retina were assessed qualitatively. Microglia form satellites with all neuronal subtypes studied, whereas a preference for a specific neuron subtype was not found. The occurrence and frequency of satellite microglia is determined by the histo-architectural organization of the brain area and the densities of neuronal somata therein.


Asunto(s)
Microglía , Neuronas , Animales , Encéfalo , Masculino , Ratones , Microglía/fisiología , Neuronas/fisiología
14.
Front Immunol ; 13: 1066456, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713399

RESUMEN

Introduction: The pandemic coronavirus disease 19 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is marked by thromboembolic events and an inflammatory response throughout the body, including the brain. Methods: Employing the machine learning approach BrainDead we systematically screened for SARS-CoV-2 genome-derived single-stranded (ss) RNA fragments with high potential to activate the viral RNA-sensing innate immune receptors Toll-like receptor (TLR)7 and/or TLR8. Analyzing HEK TLR7/8 reporter cells we tested such RNA fragments with respect to their potential to induce activation of human TLR7 and TLR8 and to activate human macrophages, as well as iPSC-derived human microglia, the resident immune cells in the brain. Results: We experimentally validated several sequence-specific RNA fragment candidates out of the SARS-CoV-2 RNA fragments predicted in silico as activators of human TLR7 and TLR8. Moreover, these SARS-CoV-2 ssRNAs induced cytokine release from human macrophages and iPSC-derived human microglia in a sequence- and species-specific fashion. Discussion: Our findings determine TLR7 and TLR8 as key sensors of SARS-CoV-2-derived ssRNAs and may deepen our understanding of the mechanisms how this virus triggers, but also modulates an inflammatory response through innate immune signaling.


Asunto(s)
COVID-19 , Citocinas , Humanos , SARS-CoV-2/genética , ARN Viral , Receptor Toll-Like 7 , Microglía , Receptor Toll-Like 8 , Macrófagos
15.
Cell Rep ; 37(13): 110128, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965412

RESUMEN

Microglia, the resident macrophages in the central nervous system, express receptors for classical neurotransmitters, such as γ-aminobutyric acid (GABA) and glutamate, suggesting that they sense synaptic activity. To detect microglial Ca2+ responses to neuronal activity, we generate transgenic mouse lines expressing the fluorescent Ca2+ indicator GCaMP6m, specifically in microglia and demonstrate that electrical stimulation of the Schaffer collateral pathway results in microglial Ca2+ responses in early postnatal but not adult hippocampus. Preceding the microglial responses, we also observe similar Ca2+ responses in astrocytes, and both are sensitive to tetrodotoxin. Blocking astrocytic glutamate uptake or GABA transport abolishes stimulation-induced microglial responses as well as antagonizing the microglial GABAB receptor. Our data, therefore, suggest that the neuronal activity-induced glutamate uptake and the release of GABA by astrocytes trigger the activation of GABAB receptors in microglia. This neuron, astrocyte, and microglia communication pathway might modulate microglial activity in developing neuronal networks.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptores de GABA/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Estimulación Eléctrica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
Front Immunol ; 12: 715774, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34589086

RESUMEN

The chaperone protein Unc-93 homolog B1 (UNC93B1) regulates internalization, trafficking, and stabilization of nucleic acid-sensing Toll-like receptors (TLR) in peripheral immune cells. We sought to determine UNC93B1 expression and its functional relevance in inflammatory and injurious processes in the central nervous system (CNS). We found that UNC93B1 is expressed in various CNS cells including microglia, astrocytes, oligodendrocytes, and neurons, as assessed by PCR, immunocyto-/histochemistry, and flow cytometry. UNC93B1 expression in the murine brain increased during development. Exposure to the microRNA let-7b, a recently discovered endogenous TLR7 activator, but also to TLR3 and TLR4 agonists, led to increased UNC93B1 expression in microglia and neurons. Microglial activation by extracellular let-7b required functional UNC93B1, as assessed by TNF ELISA. Neuronal injury induced by extracellular let-7b was dependent on UNC93B1, as UNC93B1-deficient neurons were unaffected by the microRNA's neurotoxicity in vitro. Intrathecal application of let-7b triggered neurodegeneration in wild-type mice, whereas mice deficient for UNC93B1 were protected against injurious effects on neurons and axons. In summary, our data demonstrate broad UNC93B1 expression in the murine brain and establish this chaperone as a modulator of neuroinflammation and neuronal injury triggered by extracellular microRNA and subsequent induction of TLR signaling.


Asunto(s)
Sistema Nervioso Central/metabolismo , Regulación de la Expresión Génica , Proteínas de Transporte de Membrana/genética , MicroARNs/genética , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/metabolismo , Neuronas/metabolismo , Animales , Biomarcadores , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Enfermedades Neuroinflamatorias/patología , Neuronas/efectos de los fármacos , Organogénesis/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
17.
Glia ; 69(9): 2291-2304, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34080730

RESUMEN

Histamine is a monoaminergic neurotransmitter which is released within the entire brain from ascending axons originating in the tuberomammillary nucleus in a sleep state-dependent fashion. Besides the modulation of neuronal firing patterns, brain histamine levels are also thought to modulate functions of glial cells. Microglia are the innate immune cells and professional phagocytes of the central nervous system, and histamine was previously shown to have multiple effects on microglial functions in health and disease. Isolated microglia respond only to agonists of the Hrh2 subtype of histamine receptors (Hrh), and the expression of that isoform is confirmed by a metadata analysis of microglia transcriptomes. When we studied the effect of the histamine receptor isoforms in cortical and thalamic microglia by in situ live cell Ca2+ imaging using a novel, microglia-specific indicator mouse line, microglial cells respond to external histamine application mainly in a Hrh1-, and to a lower extent also in a Hrh2-dependent manner. The Hrh1 response was sensitive to blockers of purinergic P2ry12 receptors, and since Hrh1 expression was predominantly found in astrocytes, we suggest that the Hrh1 response in microglia is mediated by astrocyte ATP release and activation of P2ry12 receptors in microglia. Histamine also stimulates microglial phagocytic activity via Hrh1- and P2ry12-mediated signaling. Taken together, we provide evidence that histamine acts indirectly on microglial Ca2+ levels and phagocytic activity via astrocyte histamine receptor-controlled purinergic signaling.


Asunto(s)
Histamina , Microglía , Animales , Astrocitos/metabolismo , Histamina/metabolismo , Histamina/farmacología , Ratones , Microglía/metabolismo , Receptores Purinérgicos/metabolismo , Transducción de Señal/fisiología
18.
Nat Neurosci ; 24(3): 312-325, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589835

RESUMEN

Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.


Asunto(s)
Envejecimiento/patología , Astrocitos/patología , Encéfalo/patología , Médula Espinal/patología , Animales , Encefalopatías/patología , Lesiones Encefálicas/patología , Humanos , Traumatismos de la Médula Espinal/patología
19.
Cell Rep ; 34(3): 108642, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33472059

RESUMEN

Thalamic astrocytes and oligodendrocytes are coupled via gap junctions and form panglial networks. Here, we show that these networks have a key role in energy supply of neurons. Filling an astrocyte or an oligodendrocyte in acute slices with glucose or lactate is sufficient to rescue the decline of stimulation-induced field post-synaptic potential (fPSP) amplitudes during extracellular glucose deprivation (EGD). In mice lacking oligodendroglial coupling, loading an astrocyte with glucose does not rescue the EGD-mediated loss of fPSPs. Monocarboxylate and glucose transporters are required for rescuing synaptic activity during EGD. In mice deficient in astrocyte coupling, filling of an oligodendrocyte with glucose does not rescue fPSPs during EGD. Our results demonstrate that, in the thalamus, astrocytes and oligodendrocytes are jointly engaged in delivering energy substrates for sustaining neuronal activity and suggest that oligodendrocytes exert their effect mainly by assisting astrocytes in metabolite transfer to the postsynapse.


Asunto(s)
Astrocitos/metabolismo , Oligodendroglía/metabolismo , Tálamo/fisiología , Animales , Emparejamiento Cromosómico , Ratones
20.
J Neurosci Res ; 99(4): 1048-1063, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33404121

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is released by glioma cells and promotes tumor growth. We have previously found that GDNF released from the tumor cells is a chemoattractant for microglial cells, the immune cells of the central nervous system. Here we show that GDNF increases matrix metalloproteinase (MMP) 9 and MMP14 expression in cultured microglial cells from mixed sexes of neonatal mice. The GDNF-induced microglial MMP9 and MMP14 upregulation is mediated by GDNF family receptor alpha 1 receptors and dependent on p38 mitogen-activated protein kinase signaling. In organotypic brain slices, GDNF promotes the growth of glioma and this effect depends on the presence of microglia. We also previously found that MMP9 and MMP14 upregulation can be mediated by Toll-like receptor (TLR) 2 signaling and here we demonstrate that GDNF increases the expression of TLR1 and TLR2. In conclusion, GDNF promotes the pro-tumorigenic phenotype of microglia.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Glioma/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Línea Celular Tumoral , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Imidazoles/farmacología , Masculino , Metaanálisis como Asunto , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Cultivo Primario de Células , Piridinas/farmacología , Transducción de Señal , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...