Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Glia ; 71(1): 71-90, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36222019

RESUMEN

Microglia, the innate immune cells of the central nervous system (CNS), execute their sentinel, housekeeping and defense functions through a panoply of genes, receptors and released cytokines, chemokines and neurotrophic factors. Moreover, microglia functions are closely linked to the constant communication with other cell types, among them neurons. Depending on the signaling pathway and type of stimuli involved, the outcome of microglia operation can be neuroprotective or neurodegenerative. Accordingly, microglia are increasingly becoming considered cellular targets for therapeutic intervention. Among signals controlling microglia activity, the endocannabinoid (EC) system has been shown to exert a neuroprotective role in many neurological diseases. Like neurons, microglia express functional EC receptors and can produce and degrade ECs. Interestingly, boosting EC signaling leads to an anti-inflammatory and neuroprotective microglia phenotype. Nonetheless, little evidence is available on the microglia-mediated therapeutic effects of EC compounds. This review focuses on the EC signals acting on the CNS microglia in physiological and pathological conditions, namely on the CB1R, CB2R and TRPV1-mediated regulation of microglia properties. It also provides new evidence, which strengthens the understanding of mechanisms underlying the control of microglia functions by ECs. Given the broad expression of the EC system in glial and neuronal cells, the resulting picture is the need for in vivo studies in transgenic mouse models to dissect the contribution of EC microglia signaling in the neuroprotective effects of EC-derived compounds.


Asunto(s)
Microglía , Fármacos Neuroprotectores , Animales , Ratones , Microglía/metabolismo , Endocannabinoides/farmacología , Endocannabinoides/metabolismo , Transducción de Señal , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología
2.
Semin Cell Dev Biol ; 94: 138-151, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31112798

RESUMEN

The continuous crosstalk between microglia and neurons is required for microglia housekeeping functions and contributes to brain homeostasis. Through these exchanges, microglia take part in crucial brain functions, including development and plasticity. The alteration of neuron-microglia communication contributes to brain disease states with consequences, ranging from synaptic function to neuronal survival. This review focuses on the signaling pathways responsible for neuron-microglia crosstalk, highlighting their physiological roles and their alteration or specific involvement in disease. In particular, we discuss studies, establishing how these signaling allow microglial cells to control relevant physiological functions during brain development, including synaptic formation and circuit refinement. In addition, we highlight how microglia and neurons interact functionally to regulate highly dynamical synaptic functions. Microglia are able to release several signaling molecules involved in the regulation of synaptic activity and plasticity. On the other side, molecules of neuronal origin control microglial processes motility in an activity-dependent manner. Indeed, the continuous crosstalk between microglia and neurons is required for the sensing and housekeeping functions of microglia and contributes to the maintenance of brain homeostasis and, particularly, to the sculpting of neuronal connections during development. These interactions lay on the delicate edge between physiological processes and homeostasis alteration in pathology and are themselves altered during neuroinflammation. The full description of these processes could be fundamental for understanding brain functioning in health and disease.


Asunto(s)
Microglía/metabolismo , Neuronas/metabolismo , Transducción de Señal , Animales , Humanos
3.
Glia ; 66(7): 1395-1416, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29473218

RESUMEN

Microglia are the sentinels of the brain but a clear understanding of the factors that modulate their activation in physiological and pathological conditions is still lacking. Here we demonstrate that Nerve Growth Factor (NGF) acts on microglia by steering them toward a neuroprotective and anti-inflammatory phenotype. We show that microglial cells express functional NGF receptors in vitro and ex vivo. Our transcriptomic analysis reveals how, in primary microglia, NGF treatment leads to a modulation of motility, phagocytosis and degradation pathways. At the functional level, NGF induces an increase in membrane dynamics and macropinocytosis and, in vivo, it activates an outward rectifying current that appears to modulate glutamatergic neurotransmission in nearby neurons. Since microglia are supposed to be a major player in Aß peptide clearance in the brain, we tested the effects of NGF on its phagocytosis. NGF was shown to promote TrkA-mediated engulfment of Aß by microglia, and to enhance its degradation. Additionally, the proinflammatory activation induced by Aß treatment is counteracted by the concomitant administration of NGF. Moreover, by acting specifically on microglia, NGF protects neurons from the Aß-induced loss of dendritic spines and inhibition of long term potentiation. Finally, in an ex-vivo setup of acute brain slices, we observed a similar increase in Aß engulfment by microglial cells under the influence of NGF. Our work substantiates a role for NGF in the regulation of microglial homeostatic activities and points toward this neurotrophin as a neuroprotective agent in Aß accumulation pathologies, via its anti-inflammatory activity on microglia.


Asunto(s)
Microglía/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuroprotección/fisiología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Factor de Crecimiento Nervioso/administración & dosificación , Neuronas/citología , Neuronas/metabolismo , Fagocitosis/fisiología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos , Transcriptoma
4.
Front Pharmacol ; 13: 869606, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35721203

RESUMEN

The latest years have witnessed a growing interest towards the relationship between neuropsychiatric disease in children with autism spectrum disorders (ASD) and severe alterations in gut microbiota composition. In parallel, an increasing literature has focused the attention towards the association between derangement of the endocannabinoids machinery and some mechanisms and symptoms identified in ASD pathophysiology, such as alteration of neural development, immune system dysfunction, defective social interaction and stereotypic behavior. In this narrative review, we put together the vast ground of endocannabinoids and their partnership with gut microbiota, pursuing the hypothesis that the crosstalk between these two complex homeostatic systems (bioactive lipid mediators, receptors, biosynthetic and hydrolytic enzymes and the entire bacterial gut ecosystem, signaling molecules, metabolites and short chain fatty acids) may disclose new ideas and functional connections for the development of synergic treatments combining "gut-therapy," nutritional intervention and pharmacological approaches. The two separate domains of the literature have been examined looking for all the plausible (and so far known) overlapping points, describing the mutual changes induced by acting either on the endocannabinoid system or on gut bacteria population and their relevance for the understanding of ASD pathophysiology. Both human pathology and symptoms relief in ASD subjects, as well as multiple ASD-like animal models, have been taken into consideration in order to provide evidence of the relevance of the endocannabinoids-microbiota crosstalk in this major neurodevelopmental disorder.

5.
Biomolecules ; 10(1)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861745

RESUMEN

The gut-brain axis is a multimodal communication system along which immune, metabolic, autonomic, endocrine and enteric nervous signals can shape host physiology and determine liability, development and progression of a vast number of human diseases. Here, we broadly discussed the current knowledge about the either beneficial or deleterious impact of dietary fatty acids on microbiota-brain communication (MBC), and the multiple mechanisms by which different types of lipids can modify gut microbial ecosystem and contribute to the pathophysiology of major neuropsychiatric diseases (NPDs), such as schizophrenia (SCZ), depression and autism spectrum disorders (ASD).


Asunto(s)
Trastorno del Espectro Autista/microbiología , Encéfalo/fisiología , Depresión/microbiología , Ácidos Grasos/metabolismo , Microbioma Gastrointestinal , Esquizofrenia/microbiología , Animales , Trastorno del Espectro Autista/metabolismo , Depresión/metabolismo , Depresión/fisiopatología , Humanos , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología
6.
Nat Commun ; 8: 15292, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28489079

RESUMEN

The capsaicin receptor TRPV1 has been widely characterized in the sensory system as a key component of pain and inflammation. A large amount of evidence shows that TRPV1 is also functional in the brain although its role is still debated. Here we report that TRPV1 is highly expressed in microglial cells rather than neurons of the anterior cingulate cortex and other brain areas. We found that stimulation of microglial TRPV1 controls cortical microglia activation per se and indirectly enhances glutamatergic transmission in neurons by promoting extracellular microglial microvesicles shedding. Conversely, in the cortex of mice suffering from neuropathic pain, TRPV1 is also present in neurons affecting their intrinsic electrical properties and synaptic strength. Altogether, these findings identify brain TRPV1 as potential detector of harmful stimuli and a key player of microglia to neuron communication.


Asunto(s)
Biomarcadores/metabolismo , Encefalitis/metabolismo , Neuralgia/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Micropartículas Derivadas de Células , Potenciales Postsinápticos Excitadores , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Transmisión Sináptica , Canales Catiónicos TRPV/genética
7.
Int Rev Neurobiol ; 85: 363-74, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19607981

RESUMEN

Reactive oxygen species (ROS) accumulation has been described in the brain following an ischemic insult. Superoxide anion is converted by superoxide dismutase into hydrogen peroxide (H2O2), and the latter is then transformed into the toxic hydroxyl radical, through the Haber-Weiss reaction, converted to water by glutathione peroxidase (GPx) or dismuted to water and oxygen through catalase. Accumulation of H2O2 has been suggested to exert neurotoxic effects, although recent in vitro studies have demonstrated either physiological or protective roles of this molecule in the brain. In particular, oxidative stress is critically involved in brain damage induced by transient cerebral ischemia. Here, we demonstrate that inhibition of GPx by systemic (i.p.) administration of mercaptosuccinate (MS, 1.5-150 mg/kg) dose-dependently reduces brain infarct damage produced by transient (2 h) middle cerebral artery occlusion (MCAo) in rat. Neuroprotection was observed when the drug was administered 15 min before the ischemic insult, whereas no effect was detected when the drug was injected 1h before MCAo or upon reperfusion. Furthermore, application of MS (1 mM) to corticostriatal slices limited the irreversible functional derangement of field potentials caused by a prolonged (12 min) oxygen-glucose deprivation. This effect was reverted by concomitant bath application of the catalase inhibitor 3-aminotriazole (20mM), suggesting the involvement of catalase in mediating the neuroprotective effects of MS. Thus, our findings demonstrate that MS is neuroprotective in both in vivo and in vitro ischemic conditions, through a mechanism which may involve increased endogenous levels of H2O2 and its consequent conversion to molecular oxygen by catalase.


Asunto(s)
Glutatión Peroxidasa/antagonistas & inhibidores , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Tiomalatos/uso terapéutico , Amitrol (Herbicida)/farmacología , Animales , Catalasa/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/farmacología , Peróxido de Hidrógeno/metabolismo , Técnicas In Vitro , Masculino , Neocórtex/efectos de los fármacos , Neocórtex/fisiopatología , Neostriado/efectos de los fármacos , Neostriado/fisiopatología , Ratas , Ratas Wistar
8.
Exp Neurol ; 212(1): 230-3, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18501350

RESUMEN

It is not yet clear how L-dopa, that is the most effective drug for the treatment of Parkinson's disease, enters into the dopaminergic neurons to be transformed into dopamine. It is suggested that L-dopa is mainly transported into cells by a group of L-amino acid carriers named "System L". Since these carriers are selectively inhibited by 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid (BCH), we have applied this compound to electrophysiologically recorded dopaminergic neurons of the rat substantia nigra pars compacta to examine the possible modulation of the effects of L-dopa by System L. We have observed that BCH reduced, in a concentration-dependent manner, the membrane hyperpolarization/outward current caused by L-dopa. Interestingly, the actions of dopamine were not changed by this System L inhibitor, suggesting that the reducing effects on L-dopa are not due to a BCH-induced unspecific block of dopamine-mediated events. Therefore, our electrophysiological data that an l-type amino acid carrier, possibly System L, is involved in the transport of L-dopa into dopaminergic neurons.


Asunto(s)
Sistema de Transporte de Aminoácidos L/antagonistas & inhibidores , Aminoácidos Cíclicos/farmacología , Dopamina/metabolismo , Levodopa/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Sistema de Transporte de Aminoácidos L/metabolismo , Aminoácidos/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Femenino , Levodopa/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuronas/metabolismo , Ratas , Ratas Wistar , Sustancia Negra/metabolismo
9.
Eur J Neurosci ; 24(7): 2061-70, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17067303

RESUMEN

Reelin-deficient mice have been used to investigate the role of this extracellular protein in cortico-striatal plasticity and striatum-related behaviours. Here we show that a repetitive electrical stimulation of the cortico-striatal pathway elicited long-term potentiation (LTP) in homozygous reeler (rl/rl) mice, while causing long-term depression in their wild-type (+/+) littermates. The N-methyl-D-aspartic acid (NMDA) receptor antagonist D-(-)-2 amino-5-phosphonopentanoic acid prevented the induction of LTP in (rl/rl) mice, thus confirming that this form of synaptic plasticity was NMDA receptor-dependent. Interestingly, in the presence of tiagabine, a blocker of gamma-aminobutyric acid (GABA) re-uptake system, the probability that (rl/rl) mice showed LTP decreased significantly, thus suggesting an impaired GABAergic transmission in reeler mutants. Consistent with this view, a decreased density of parvalbumin-positive GABAergic striatal interneurons was found in (rl/rl) mice in comparison to (+/+) mice. Finally, compatible with their abnormal striatal function (rl/rl) mice exhibited procedural learning deficits. Our data, showing alterations in cortico-striatal plasticity largely depending on a depressed GABAergic tone, delineate a mechanism whereby the lack of reelin may affect cognitive functions.


Asunto(s)
Reacción de Prevención/fisiología , Conducta Animal/fisiología , Corteza Cerebral/citología , Cuerpo Estriado/citología , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , 2-Amino-5-fosfonovalerato/farmacología , Animales , Dopamina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de la radiación , Agonistas del GABA/farmacología , Inmunohistoquímica/normas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos/fisiología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Ácidos Nipecóticos/farmacología , Parvalbúminas/metabolismo , Probabilidad , Proteína Reelina , Sinapsis/efectos de los fármacos , Tiagabina , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA