Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Mol Neurobiol ; 44(1): 60, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39287687

RESUMEN

Microglia are macrophage cells residing in the brain, where they exert a key role in neuronal protection. Through the gut-brain axis, metabolites produced by gut commensal microbes can influence brain functions, including microglial activity. The nuclear factor erythroid 2-related factor 2 (NRF2) is a key regulator of the oxidative stress response in microglia, controlling the expression of cytoprotective genes. Lactobacilli-derived cell-free supernatants (CFSs) are postbiotics that have shown antioxidant and immunomodulatory effects in several in vitro and in vivo studies. This study aimed to explore the effects of lactobacilli CFSs on modulating microglial responses against oxidative stress and inflammation. HMC3 microglia were exposed to lipopolysaccaride (LPS), as an inflammatory trigger, before and after administration of CFSs from three human gut probiotic species. The NRF2 nuclear protein activation and the expression of NRF2-controlled antioxidant genes were investigated by immunoassay and quantitative RT-PCR, respectively. Furthermore, the level of pro- and anti-inflammatory cytokines was evaluated by immunoassay. All CFSs induced a significant increase of NRF2 nuclear activity in basal conditions and upon inflammation. The transcription of antioxidant genes, namely heme oxygenase 1, superoxide dismutase (SOD), glutathione-S transferase, glutathione peroxidase, and catalase also increased, especially after inflammatory stimulus. Besides, higher SOD1 activity was detected relative to inflamed microglia. In addition, CFSs pre-treatment of microglia attenuated pro-inflammatory TNF-α levels while increasing anti-inflammatory IL-10 levels. These findings confirmed that gut microorganisms' metabolites can play a relevant role in adjuvating the microglia cellular response against neuroinflammation and oxidative stress, which are known to cause neurodegenerative diseases.


Asunto(s)
Inflamación , Lactobacillus , Microglía , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Transducción de Señal , Superóxido Dismutasa-1 , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Microglía/metabolismo , Microglía/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa-1/metabolismo , Lipopolisacáridos/farmacología , Citocinas/metabolismo , Antioxidantes/metabolismo , Antioxidantes/farmacología , Línea Celular
2.
Epilepsia ; 64(5): 1175-1189, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36807867

RESUMEN

Animal models of human brain disorders permit researchers to explore disease mechanisms and to test potential therapies. However, therapeutic molecules derived from animal models often translate poorly to the clinic. Although human data may be more relevant, experiments on patients are constrained, and living tissue is unavailable for many disorders. Here, we compare work on animal models and on human tissue for three epileptic syndromes where human tissue is excised therapeutically: (1) acquired temporal lobe epilepsies, (2) inherited epilepsies associated with cortical malformations, and (3) peritumoral epilepsies. Animal models rest on assumed equivalencies between human brains and brains of mice, the most frequently used model animal. We ask how differences between mouse and human brains could influence models. General principles and compromises in model construction and validation are examined for a range of neurological diseases. Models may be judged on how well they predict novel therapeutic molecules or new mechanisms. The efficacy and safety of new molecules are evaluated in clinical trials. We judge new mechanisms by comparing data from work on animal models with data from work on patient tissue. In conclusion, we stress the need to cross-verify findings from animal models and from living human tissue to avoid the assumption that mechanisms are identical.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Síndromes Epilépticos , Humanos , Animales , Ratones , Epilepsia/genética , Epilepsia/terapia , Encéfalo , Modelos Animales
3.
J Neurosci ; 40(7): 1373-1388, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31896671

RESUMEN

Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Glioma/fisiopatología , Microglía/fisiología , Receptores Purinérgicos P2Y12/fisiología , Receptores Purinérgicos P2Y1/fisiología , Receptores Purinérgicos P2/fisiología , Neoplasias Supratentoriales/fisiopatología , Adenosina Difosfato/farmacología , Adulto , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Forma de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/fisiología , Extensiones de la Superficie Celular/ultraestructura , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/patología , Femenino , Glioma/patología , Humanos , Microscopía Intravital , Masculino , Microglía/efectos de los fármacos , Microglía/ultraestructura , Persona de Mediana Edad , Lectinas de Plantas , Agonistas Purinérgicos/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Neoplasias Supratentoriales/patología , Esclerosis Tuberosa/complicaciones
4.
Eur J Neurosci ; 50(1): 1759-1778, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30767299

RESUMEN

Lipid homeostasis is dysregulated in some neurodegenerative diseases and after brain injuries due to excess glutamate or lack of oxygen. However the kinetics and cell specificity of dysregulation in different groups of lipids during excitotoxic neuronal death are not clear. Here we examined the changes during excitotoxic neuronal death induced by injecting kainic acid (KA) into the CA1 region of mouse hippocampus. We compared neuronal loss and glial cell proliferation with changes in lipid-related transcripts and markers for different lipid groups, over 12 days after KA-treatment. As neurons showed initial signs of damage, transcripts and proteins linked to fatty acid oxidation were up-regulated. Cholesterol biosynthesis induced by transcripts controlled by the transcription factor Srebp2 seems to be responsible for a transient increase in neuronal free cholesterol at 1 to 2 days. In microglia, but not in neurons, Perilipin-2 associated lipid droplets were induced and properties of Nile red emissions suggest lipid contents change over time. After microglial expression of phagocytotic markers at 2 days, some neutral lipid deposits co-localized with lysosome markers of microglia and were detected within putative phagocytotic cups. These data delineate distinct lipid signals in neurons and glial cells during excitotoxic processes from initial neuronal damage to engagement of the lysosome-phagosome system.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Perfilación de la Expresión Génica , Ácido Kaínico/farmacología , Gotas Lipídicas/metabolismo , Lípidos de la Membrana/metabolismo , Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Animales , Biomarcadores/metabolismo , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colesterol/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microscopía Electrónica , Microscopía de Fluorescencia por Excitación Multifotónica , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Regulación hacia Arriba
5.
Brain ; 141(12): 3343-3360, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462183

RESUMEN

Microglia, the immune cells of the brain, are highly plastic and possess multiple functional phenotypes. Differences in phenotype in different regions and different states of epileptic human brain have been little studied. Here we use transcriptomics, anatomy, imaging of living cells and ELISA measurements of cytokine release to examine microglia from patients with temporal lobe epilepsies. Two distinct microglial phenotypes were explored. First we asked how microglial phenotype differs between regions of high and low neuronal loss in the same brain. Second, we asked how microglial phenotype is changed by a recent seizure. In sclerotic areas with few neurons, microglia have an amoeboid rather than ramified shape, express activation markers and respond faster to purinergic stimuli. The repairing interleukin, IL-10, regulates the basal phenotype of microglia in the CA1 and CA3 regions with neuronal loss and gliosis. To understand changes in phenotype induced by a seizure, we estimated the delay from the last seizure until tissue collection from changes in reads for immediate early gene transcripts. Pseudotime ordering of these data was validated by comparison with results from kainate-treated mice. It revealed a local and transient phenotype in which microglia secrete the human interleukin CXCL8, IL-1B and other cytokines. This secretory response is mediated in part via the NRLP3 inflammasome.


Asunto(s)
Encéfalo/inmunología , Encéfalo/patología , Epilepsia del Lóbulo Temporal/inmunología , Epilepsia del Lóbulo Temporal/patología , Microglía/patología , Adulto , Anciano , Animales , Epilepsia del Lóbulo Temporal/metabolismo , Femenino , Humanos , Interleucina-10/metabolismo , Masculino , Ratones , Microglía/metabolismo , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fenotipo , Transcriptoma , Adulto Joven
6.
Neural Plast ; 2019: 4651031, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804991

RESUMEN

An increasing number of studies show that selective serotonin reuptake inhibitors (SSRIs) exert their therapeutic action, at least in part, by amplifying the influence of the living environment on mood. As a consequence, when administered in a favorable environment, SSRIs lead to a reduction of symptoms, but in stressful conditions, they show limited efficacy. Therefore, novel therapeutic approaches able to neutralize the influence of the stressful environment on treatment are needed. The aim of our study was to test whether, in a mouse model of depression, the combined administration of SSRI fluoxetine and metformin, a drug able to improve the metabolic profile, counteracts the limited efficacy of fluoxetine alone when administered in stressful conditions. Indeed, metabolic alterations are associated to both the onset of major depression and the antidepressant efficacy. To this goal, adult C57BL/6 male mice were exposed to stress for 6 weeks; the first two weeks was aimed at generating a mouse model of depression. During the remaining 4 weeks, mice received one of the following treatments: vehicle, fluoxetine, metformin, or a combination of fluoxetine and metformin. We measured liking- and wanting-type anhedonia as behavioral phenotypes of depression and assessed the expression levels of selected genes involved in major depressive disorder and antidepressant response in the dorsal and ventral hippocampus, which are differently involved in the depressive symptomatology. The combined treatment was more effective than fluoxetine alone in ameliorating the depressive phenotype after one week of treatment. This was associated to an increase in IGF2 mRNA expression and enhanced long-term potentiation, specifically in the dorsal hippocampus, at the end of treatment. Overall, the present results show that, when administered in stressful conditions, the combined fluoxetine and metformin treatment may represent a more effective approach than fluoxetine alone in a short term. Finally, our findings highlight the relevance of polypharmacological strategy as effective interventions to increase the efficacy of the antidepressant drugs currently available.


Asunto(s)
Anhedonia/efectos de los fármacos , Antidepresivos/uso terapéutico , Trastorno Depresivo/tratamiento farmacológico , Fluoxetina/uso terapéutico , Hipocampo/efectos de los fármacos , Factor II del Crecimiento Similar a la Insulina/metabolismo , Metformina/uso terapéutico , Animales , Antidepresivos/farmacología , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Fluoxetina/farmacología , Hipocampo/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Masculino , Metformina/farmacología , Ratones , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico
7.
Glia ; 64(5): 826-39, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26847266

RESUMEN

The past decade has witnessed a revolution in our understanding of microglia. These immune cells were shown to actively remodel neuronal circuits, leading to propose new pathogenic mechanisms. To study microglial implication in the loss of synapses, the best pathological correlate of cognitive decline across chronic stress, aging, and diseases, we recently conducted ultrastructural analyses. Our work uncovered the existence of a new microglial phenotype that is rarely present under steady state conditions, in hippocampus, cerebral cortex, amygdala, and hypothalamus, but becomes abundant during chronic stress, aging, fractalkine signaling deficiency (CX3 CR1 knockout mice), and Alzheimer's disease pathology (APP-PS1 mice). Even though these cells display ultrastructural features of microglia, they are strikingly distinct from the other phenotypes described so far at the ultrastructural level. They exhibit several signs of oxidative stress, including a condensed, electron-dense cytoplasm and nucleoplasm making them as "dark" as mitochondria, accompanied by a pronounced remodeling of their nuclear chromatin. Dark microglia appear to be much more active than the normal microglia, reaching for synaptic clefts, while extensively encircling axon terminals and dendritic spines with their highly ramified and thin processes. They stain for the myeloid cell markers IBA1 and GFP (in CX3 CR1-GFP mice), and strongly express CD11b and microglia-specific 4D4 in their processes encircling synaptic elements, and TREM2 when they associate with amyloid plaques. Overall, these findings suggest that dark microglia, a new phenotype that we identified based on their unique properties, could play a significant role in the pathological remodeling of neuronal circuits, especially at synapses.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Corteza Cerebral/patología , Microglía/patología , Estrés Psicológico/patología , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antígenos CD/metabolismo , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Fenotipo , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Estrés Psicológico/genética
8.
Brain Behav Immun ; 55: 114-125, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26231972

RESUMEN

Chronic stress is one of the most relevant triggering factors for major depression. Microglial cells are highly sensitive to stress and, more generally, to environmental challenges. However, the role of these brain immune cells in mediating the effects of stress is still unclear. Fractalkine signaling - which comprises the chemokine CX3CL1, mainly expressed by neurons, and its receptor CX3CR1, almost exclusively present on microglia in the healthy brain - has been reported to critically regulate microglial activity. Here, we investigated whether interfering with microglial function by deleting the Cx3cr1 gene affects the brain's response to chronic stress. To this purpose, we housed Cx3cr1 knockout and wild-type adult mice in either control or stressful environments for 2weeks, and investigated the consequences on microglial phenotype and interactions with synapses, synaptic transmission, behavioral response and corticosterone levels. Our results show that hampering neuron-microglia communication via the CX3CR1-CX3CL1 pathway prevents the effects of chronic unpredictable stress on microglial function, short- and long-term neuronal plasticity and depressive-like behavior. Overall, the present findings suggest that microglia-regulated mechanisms may underlie the differential susceptibility to stress and consequently the vulnerability to diseases triggered by the experience of stressful events, such as major depression.


Asunto(s)
Conducta Animal/fisiología , Receptor 1 de Quimiocinas CX3C/deficiencia , Trastorno Depresivo Mayor/fisiopatología , Microglía , Plasticidad Neuronal/fisiología , Neuronas , Transducción de Señal/fisiología , Estrés Psicológico/fisiopatología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Brain Behav Immun ; 58: 261-271, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27474084

RESUMEN

It has been hypothesized that selective serotonin reuptake inhibitors (SSRIs), the most common treatment for major depression, affect mood through changes in immune function. However, the effects of SSRIs on inflammatory response are contradictory since these act either as anti- or pro-inflammatory drugs. Previous experimental and clinical studies showed that the quality of the living environment moderates the outcome of antidepressant treatment. Therefore, we hypothesized that the interplay between SSRIs and the environment may, at least partially, explain the apparent incongruence regarding the effects of SSRI treatment on the inflammatory response. In order to investigate such interplay, we exposed C57BL/6 mice to chronic stress to induce a depression-like phenotype and, subsequently, to fluoxetine treatment or vehicle (21days) while being exposed to either an enriched or a stressful condition. At the end of treatment, we measured the expression levels of several anti- and pro-inflammatory cytokines and inflammatory mediators in the whole hippocampus and in isolated microglia. We also determined microglial density, distribution, and morphology to investigate their surveillance state. Results show that the effects of fluoxetine treatment on inflammation and microglial function, as compared to vehicle, were dependent on the quality of the living environment. In particular, fluoxetine administered in the enriched condition increased the expression of pro-inflammatory markers compared to vehicle, while treatment in a stressful condition produced anti-inflammatory effects. These findings provide new insights regarding the effects of SSRIs on inflammation, which may be crucial to devise pharmacological strategies aimed at enhancing antidepressant efficacy by means of controlling environmental conditions.


Asunto(s)
Encefalitis/metabolismo , Ambiente , Fluoxetina/administración & dosificación , Microglía/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Animales , Citocinas/metabolismo , Depresión , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/fisiología , Estrés Psicológico
10.
Adv Neurobiol ; 37: 569-578, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39207713

RESUMEN

Microglia are macrophages residing in the central nervous system, where they perform immune surveillance, synaptic remodeling, neurogenesis, and monitor signals arising from brain injuries or potential pathogens.Commonly, rodent models are used for studying microglia because of the available transgenic mouse lines in which specific genetic manipulations are successfully accomplished. However, human and rodents microglia showed significant differences, which are reflected in different morphological and functional properties. These differences are in genetic and transcriptomic, but also in the expression of signaling molecules and age-associated changes.Several strategies are available to study human microglia, as using surgical brain resections from epileptic and tumoral tissues and from post mortem brain samples. In addition, the generation of human-induced pluripotent stem cells (hPSCs) and the possibility to differentiate them in microglia-like cells provide unique opportunities to compare microglia functions between rodents' and human brain.The use of human ex vivo and in vitro brain models allows the study of human microglia, mimicking in vivo conditions. This will be useful for a better understanding of the real live behavior and functions of microglia in the human brain. This chapter aims to highlight significant similarities and differences between human and rodent microglia in order to re-evaluate mouse models of different human brain disorders, proposing the use of in vitro and ex vivo human brain models.Studies on living human microglia in the brain may help to define divergences from animal models and to improve clinical interventions to treat brain pathologies, using alternatives targets.


Asunto(s)
Microglía , Animales , Humanos , Ratones , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/metabolismo , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Microglía/inmunología , Microglía/metabolismo
11.
Cell Rep ; 43(7): 114504, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38996064

RESUMEN

Astroglial networks closely interact with neuronal populations, but their functional contribution to neuronal representation of sensory information remains unexplored. The superior colliculus (SC) integrates multi-sensory information by generating distinct spatial patterns of neuronal functional responses to specific sensory stimulation. Here, we report that astrocytes from the mouse SC form extensive networks in the retinorecipient layer compared to visual cortex. This strong astroglial connectivity relies on high expression of gap-junction proteins. Genetic disruption of this connectivity functionally impairs SC retinotopic and orientation preference responses. These alterations are region specific, absent in primary visual cortex, and associated at the circuit level with a specific impairment of collicular neurons synaptic transmission. This has implications for SC-related visually induced innate behavior, as disrupting astroglial networks impairs light-evoked temporary arrest. Our results indicate that astroglial networks shape synaptic circuit activity underlying SC functional visual responses and play a crucial role in integrating visual cues to drive sensory-motor behavior.


Asunto(s)
Astrocitos , Neuronas , Colículos Superiores , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Ratones , Colículos Superiores/fisiología , Neuronas/fisiología , Neuronas/metabolismo , Ratones Endogámicos C57BL , Transmisión Sináptica/fisiología , Estimulación Luminosa , Corteza Visual/fisiología , Red Nerviosa/fisiología , Masculino
12.
Cell Rep ; 43(5): 114158, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38722742

RESUMEN

Throughout the brain, astrocytes form networks mediated by gap junction channels that promote the activity of neuronal ensembles. Although their inputs on neuronal information processing are well established, how molecular gap junction channels shape neuronal network patterns remains unclear. Here, using astroglial connexin-deficient mice, in which astrocytes are disconnected and neuronal bursting patterns are abnormal, we show that astrocyte networks strengthen bursting activity via dynamic regulation of extracellular potassium levels, independently of glutamate homeostasis or metabolic support. Using a facilitation-depression model, we identify neuronal afterhyperpolarization as the key parameter underlying bursting pattern regulation by extracellular potassium in mice with disconnected astrocytes. We confirm this prediction experimentally and reveal that astroglial network control of extracellular potassium sustains neuronal afterhyperpolarization via KCNQ voltage-gated K+ channels. Altogether, these data delineate how astroglial gap junctions mechanistically strengthen neuronal population bursts and point to approaches for controlling aberrant activity in neurological diseases.


Asunto(s)
Astrocitos , Uniones Comunicantes , Hipocampo , Canales de Potasio KCNQ , Potasio , Animales , Ratones , Potenciales de Acción/fisiología , Astrocitos/metabolismo , Conexinas/metabolismo , Conexinas/genética , Uniones Comunicantes/metabolismo , Hipocampo/metabolismo , Canales de Potasio KCNQ/metabolismo , Canales de Potasio KCNQ/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Neuronas/metabolismo , Potasio/metabolismo , Masculino , Femenino
13.
Cell Rep ; 42(5): 112456, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37126448

RESUMEN

The regulation of translation in astrocytes, the main glial cells in the brain, remains poorly characterized. We developed a high-throughput proteomics screen for polysome-associated proteins in astrocytes and focused on ribosomal protein receptor of activated protein C kinase 1 (RACK1), a critical factor in translational regulation. In astrocyte somata and perisynaptic astrocytic processes (PAPs), RACK1 preferentially binds to a number of mRNAs, including Kcnj10, encoding the inward-rectifying potassium (K+) channel Kir4.1. By developing an astrocyte-specific, conditional RACK1 knockout mouse model, we show that RACK1 represses production of Kir4.1 in hippocampal astrocytes and PAPs. Upregulation of Kir4.1 in the absence of RACK1 increases astrocytic Kir4.1-mediated K+ currents and volume. It also modifies neuronal activity attenuating burst frequency and duration. Reporter-based assays reveal that RACK1 controls Kcnj10 translation through the transcript's 5' untranslated region. Hence, translational regulation by RACK1 in astrocytes represses Kir4.1 expression and influences neuronal activity.


Asunto(s)
Astrocitos , Neuroglía , Animales , Ratones , Astrocitos/metabolismo , Ratones Noqueados , Neuroglía/metabolismo , Neuronas , Receptores de Cinasa C Activada/metabolismo , Ribosomas
14.
Methods Mol Biol ; 2034: 111-125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392681

RESUMEN

Although microglial cells are not electrically excitable, they express a large repertoire of ion channels that are activated by voltage, stretch, extracellular ligands, or intracellular pathways (e.g. Ca2+, G-proteins). The patch-clamp technique is the electrophysiological method of choice to study these channels whose expression varies largely in pathological conditions but also during normal development and aging. This chapter focuses on protocols allowing the recording and the analysis of these channels in acute brain slices, with a particular emphasis on the study of channels activated by extracellular ligands.


Asunto(s)
Encéfalo , Potenciales de la Membrana , Microglía , Animales , Encéfalo/citología , Encéfalo/metabolismo , Humanos , Microglía/citología , Microglía/metabolismo , Técnicas de Placa-Clamp
15.
Methods Mol Biol ; 2034: 325-336, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392696

RESUMEN

Human microglia, as those of rodents, may possess multiple functional phenotypes. Here we present protocols to determine elements of these microglial phenotypes obtained after therapeutic excision of brain tissue from patients with epilepsies of the temporal lobe and cortical gliomas. This technique permits to identify microglia, to determine their shape and expression of state-specific markers, to measure resting and induced motilities, to define a human microglial transcriptome, and to determine how it changes after a seizure.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas , Epilepsia del Lóbulo Temporal , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma , Microglía , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/patología , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Microglía/metabolismo , Microglía/patología
16.
J Neurosci Methods ; 298: 33-44, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29427611

RESUMEN

BACKGROUND: Insights into human brain diseases may emerge from tissue obtained after operations on patients. However techniques requiring transduction of transgenes carried by viral vectors cannot be applied to acute human tissue. NEW METHOD: We show that organotypic culture techniques can be used to maintain tissue from patients with three different neurological syndromes for several weeks in vitro. Optimized viral vector techniques and promoters for transgene expression are described. RESULTS: Region-specific differences in neuronal form, firing pattern and organization as well as pathological activities were maintained over 40-50 days in culture. Both adeno-associated virus and lentivirus based vectors were persistently expressed from ∼10 days after application, providing 30-40 days to exploit genetically expressed constructs. Different promoters, including hSyn, e/hSyn, CMV and CaMKII, provided cell-type specific transgene expression. The Ca probe GCaMP let us explore epileptogenic synchrony and a FRET-based probe was used to follow activity of the kinase mTORC1. COMPARISON WITH EXISTING METHODS: The use of a defined culture medium, with low concentrations of amino acids and no growth factors, permitted organotypic culture of tissue from humans aged 3-62 years. Epileptic activity was maintained and excitability changed relatively little until ∼6 weeks in culture. CONCLUSIONS: Characteristic morphology and region-specific neuronal activities are maintained in organotypic culture of tissue from patients diagnosed with mesial temporal lobe epilepsy, cortical dysplasia and cortical glioblastoma. Viral vector techniques permit expression of probes for long-term measurements of multi-cellular activity and intra-cellular signaling.


Asunto(s)
Encefalopatías/metabolismo , Encefalopatías/patología , Encéfalo/metabolismo , Encéfalo/patología , Imagen Óptica , Técnicas de Cultivo de Tejidos/métodos , Adolescente , Adulto , Encefalopatías/cirugía , Niño , Preescolar , Medios de Cultivo , Epilepsia/metabolismo , Epilepsia/patología , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Imagen Óptica/métodos , Técnicas de Cultivo de Órganos/métodos , Imagen de Colorante Sensible al Voltaje/métodos , Adulto Joven
17.
Sci Rep ; 6: 34633, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721466

RESUMEN

Chemokines have several physio-pathological roles in the brain. Among them, the modulation of synaptic contacts and neurotransmission recently emerged as crucial activities during brain development, in adulthood, upon neuroinflammation and neurodegenerative diseases. CXCL16 is a chemokine normally expressed in the brain, where it exerts neuroprotective activity against glutamate-induced damages through cross communication with astrocytes and the involvement of the adenosine receptor type 3 (A3R) and the chemokine CCL2. Here we demonstrated for the first time that CXCL16 exerts a modulatory activity on inhibitory and excitatory synaptic transmission in CA1 area. We found that CXCL16 increases the frequency of the miniature inhibitory synaptic currents (mIPSCs) and the paired-pulse ratio (PPR) of evoked IPSCs (eIPSCs), suggesting a presynaptic modulation of the probability of GABA release. In addition, CXCL16 increases the frequency of the miniature excitatory synaptic currents (mEPSCs) and reduces the PPR of evoked excitatory transmission, indicating that the chemokine also modulates and enhances the release of glutamate. These effects were not present in the A3RKO mice and in WT slices treated with minocycline, confirming the involvement of A3 receptors and introducing microglial cells as key mediators of the modulatory activity of CXCL16 on neurons.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Quimiocina CXCL16/metabolismo , Potenciales Evocados/fisiología , Ácido Glutámico/metabolismo , Neurotransmisores/metabolismo , Transmisión Sináptica/fisiología , Animales , Quimiocina CXCL16/genética , Potenciales Evocados/efectos de los fármacos , Ácido Glutámico/genética , Ratones , Ratones Noqueados , Minociclina/farmacología , Receptor de Adenosina A3/genética , Receptor de Adenosina A3/metabolismo , Transmisión Sináptica/efectos de los fármacos
18.
Sci Rep ; 6: 38242, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27922053

RESUMEN

Evidence for different physiological properties along the hippocampal longitudinal axis is emerging. Here, we examined the electrophysiological features of neurons at different dorso-ventral sites of the mouse CA1 hippocampal region. Cell position was defined with respect to longitudinal coordinates of each slice. We measured variations in neuronal excitability, subthreshold membrane properties and neurotransmitter responses along the longitudinal axis. We found that (i) pyramidal cells of the dorsal hippocampus (DH) were less excitable than those of the ventral hippocampus (VH). Resting Membrane Potential (RMP) was more hyperpolarized and somatic Input Resistance (Ri) was lower in DH compared to VH. (ii) The Paired-pulse ratio (PPR) of focally induced synaptic responses was systematically reduced from the DH to the VH; (iii) Long-term-potentiation was most pronounced in the DH and fell gradually in the intermediate hippocampus and in the VH; (iv) the frequency of miniature GABAergic events was higher in the VH than in the DH; (v) the PPR of evoked inhibitory post-synaptic current (IPSC) was higher in the DH than in the VH. These findings indicate an increased probability of both GABA and glutamate release and a reduced plasticity in the ventral compared to more dorsal regions of the hippocampus.


Asunto(s)
Electrofisiología/métodos , Hipocampo/fisiología , Potenciales de Acción , Animales , Región CA1 Hipocampal/fisiología , Masculino , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Técnicas de Cultivo de Órganos/métodos , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA