Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1205112, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37293130

RESUMEN

Tuberous sclerosis complex (TSC) is a rare monogenic disorder co-diagnosed with high rates of autism and is caused by loss of function mutations in the TSC1 or TSC2 genes. A key pathway hyperactivated in TSC is the mammalian/mechanistic target of rapamycin complex 1 (mTORC1), which regulates cap-dependent mRNA translation. We previously demonstrated that exaggerated cap-dependent translation leads to autism-related phenotypes and increased mRNA translation and protein expression of Neuroligin 1 (Nlgn1) in mice. Inhibition of Nlgn1 expression reversed social behavior deficits in mice with increased cap-dependent translation. Herein, we report elevated translation of Nlgn1 mRNA and an increase in its protein expression. Genetic or pharmacological inhibition of Nlgn1 expression in Tsc2 +/- mice rescued impaired hippocampal mGluR-LTD, contextual discrimination and social behavior deficits in Tsc2 +/- mice, without correcting mTORC1 hyperactivation. Thus, we demonstrate that reduction of Nlgn1 expression in Tsc2 +/- mice is a new therapeutic strategy for TSC and potentially other neurodevelopmental disorders.

2.
Mol Autism ; 11(1): 29, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375878

RESUMEN

BACKGROUND: Mutations in TSC1 or TSC2 genes cause tuberous sclerosis complex (TSC), a disorder associated with epilepsy, autism, and intellectual disability. TSC1 and TSC2 are repressors of the mechanistic target of rapamycin complex 1 (mTORC1), a key regulator of protein synthesis. Dysregulation of mTORC1 in TSC mouse models leads to impairments in excitation-inhibition balance, synaptic plasticity, and hippocampus-dependent learning and memory deficits. However, synaptic inhibition arises from multiple types of inhibitory interneurons and how changes in specific interneurons contribute to TSC remains largely unknown. In the present work, we determined the effect of conditional Tsc1 haploinsufficiency in a specific subgroup of inhibitory cells on hippocampal function in mice. METHODS: We investigated the consequences of conditional heterozygous knockout of Tsc1 in MGE-derived inhibitory cells by crossing Nkx2.1Cre/wt;Tsc1f/f mice. We examined the changes in mTORC1 activity and synaptic transmission in hippocampal cells, as well as hippocampus-related cognitive tasks. RESULTS: We detected selective increases in phosphorylation of ribosomal protein S6 in interneurons, indicating cell-specific-upregulated mTORC1 signaling. At the behavioral level, Nkx2.1Cre/wt;Tsc1f/wt mice exhibited intact contextual fear memory, but impaired contextual fear discrimination. They displayed intact spatial learning and reference memory but impairment in spatial working memory. Whole-cell recordings in hippocampal slices of Nkx2.1Cre/wt;Tsc1f/wt mice showed intact basic membrane properties, as well as miniature excitatory and inhibitory synaptic transmission, in pyramidal and Nkx2.1-expressing inhibitory cells. Using optogenetic activation of Nkx2.1 interneurons in slices of Nkx2.1Cre/wt;Tsc1f/wt mice, we found a decrease in synaptic inhibition of pyramidal cells. Chronic, but not acute treatment, with the mTORC1 inhibitor rapamycin reversed the impairment in synaptic inhibition. CONCLUSIONS: Our results indicate that Tsc1 haploinsufficiency in MGE-derived inhibitory cells upregulates mTORC1 activity in these interneurons, reduces their synaptic inhibition of pyramidal cells, and alters contextual fear discrimination and spatial working memory. Thus, selective dysregulation of mTORC1 function in Nkx2.1-expressing inhibitory cells appears sufficient to impair synaptic inhibition and contributes to cognitive deficits in the Tsc1 mouse model of TSC.


Asunto(s)
Miedo , Haploinsuficiencia , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Memoria a Corto Plazo , Células Piramidales/metabolismo , Transmisión Sináptica/genética , Factor Nuclear Tiroideo 1/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Heterocigoto , Interneuronas , Masculino , Ratones , Ratones Noqueados , Factor Nuclear Tiroideo 1/metabolismo
3.
Nat Commun ; 9(1): 2459, 2018 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-29941989

RESUMEN

Translation of mRNA into protein has a fundamental role in neurodevelopment, plasticity, and memory formation; however, its contribution in the pathophysiology of depressive disorders is not fully understood. We investigated the involvement of MNK1/2 (MAPK-interacting serine/threonine-protein kinase 1 and 2) and their target, eIF4E (eukaryotic initiation factor 4E), in depression-like behavior in mice. Mice carrying a mutation in eIF4E for the MNK1/2 phosphorylation site (Ser209Ala, Eif4e ki/ki), the Mnk1/2 double knockout mice (Mnk1/2-/-), or mice treated with the MNK1/2 inhibitor, cercosporamide, displayed anxiety- and depression-like behaviors, impaired serotonin-induced excitatory synaptic activity in the prefrontal cortex, and diminished firing of the dorsal raphe neurons. In Eif4e ki/ki mice, brain IκBα, was decreased, while the NF-κB target, TNFα was elevated. TNFα inhibition in Eif4e ki/ki mice rescued, whereas TNFα administration to wild-type mice mimicked the depression-like behaviors and 5-HT synaptic deficits. We conclude that eIF4E phosphorylation modulates depression-like behavior through regulation of inflammatory responses.


Asunto(s)
Ansiedad/patología , Depresión/patología , Factor 4E Eucariótico de Iniciación/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Animales , Antidepresivos/farmacología , Ansiedad/inducido químicamente , Ansiedad/genética , Conducta Animal/fisiología , Benzofuranos/farmacología , Citalopram/farmacología , Depresión/inducido químicamente , Depresión/genética , Trastorno Depresivo Mayor/patología , Femenino , Fluoxetina/farmacología , Inflamación/patología , Ketamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor NF-kappaB alfa/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Inhibidores de Captación de Serotonina y Norepinefrina/farmacología , Transmisión Sináptica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Neurosci ; 37(3): 546-561, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28100738

RESUMEN

MicroRNAs (miRNA) play an important role in post-transcriptional gene regulation of several physiological and pathological processes. In multiple sclerosis (MS), a chronic inflammatory and degenerative disease of the CNS, and in its mouse model, the experimental autoimmune encephalomyelitis (EAE), miRNA dysregulation has been mainly related to immune system dysfunction and white matter (WM) pathology. However, little is known about their role in gray matter pathology. Here, we explored miRNA involvement in the inflammation-driven alterations of synaptic structure and function, collectively known as synaptopathy, a neuropathological process contributing to excitotoxic neurodegeneration in MS/EAE. Particularly, we observed that miR-142-3p is increased in the CSF of patients with active MS and in EAE brains. We propose miR-142-3p as a molecular mediator of the IL-1ß-dependent downregulation of the glial glutamate-aspartate transporter (GLAST), which causes an enhancement of the glutamatergic transmission in the EAE cerebellum. The synaptic abnormalities mediated by IL-1ß and the clinical and neuropathological manifestations of EAE disappeared in miR-142 knock-out mice. Furthermore, we observed that in vivo miR-142-3p inhibition, either by a preventive and local treatment or by a therapeutic and systemic strategy, abolished IL-1ß- and GLAST-dependent synaptopathy in EAE wild-type mice. Consistently, miR-142-3p was responsible for the glutamatergic synaptic alterations caused by CSF of patients with MS, and CSF levels of miR-142-3p correlated with prospective MS disease progression. Our findings highlight miR-142-3p as key molecular player in IL-1ß-mediated synaptic dysfunction, possibly leading to excitotoxic damage in both EAE and MS diseases. Inhibition of miR-142-3p could be neuroprotective in MS. SIGNIFICANCE STATEMENT: Current studies suggest the role of glutamate excitotoxicity in the development and progression of multiple sclerosis (MS) and of its mouse model experimental autoimmune encephalomyelitis (EAE). The molecular mechanisms linking inflammation and synaptic alterations in MS/EAE are still unknown. Here, we identified miR-142-3p as a determinant molecular actor in inflammation-dependent synaptopathy typical of both MS and EAE. miR-142-3p was upregulated in the CSF of MS patients and in EAE cerebellum. Inhibition of miR-142-3p, locally in EAE brain and in a MS chimeric ex vivo model, recovered glutamatergic synaptic enhancement typical of EAE/MS. We proved that miR-142-3p promoted the IL-1ß-dependent glutamate dysfunction by targeting glutamate-aspartate transporter (GLAST), a crucial glial transporter involved in glutamate homeostasis. Finally, we suggest miR-142-3p as a negative prognostic factor in patients with relapsing-remitting multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-1beta/biosíntesis , MicroARNs/biosíntesis , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Sinapsis/metabolismo , Adulto , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/patología , Femenino , Técnicas de Sustitución del Gen , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/líquido cefalorraquídeo , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Sinapsis/patología
5.
Neurobiol Dis ; 74: 347-58, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25511803

RESUMEN

Mood disturbances are frequent in patients with multiple sclerosis (MS), even in non-disabled patients and in the remitting stages of the disease. It is still largely unknown how the pathophysiological process on MS causes anxiety and depression, but the dopaminergic system is likely involved. Aim of the present study was to investigate depressive-like behavior in mice with experimental autoimmune encephalomyelitis (EAE), a model of MS, and its possible link to dopaminergic neurotransmission. Behavioral, amperometric and biochemical experiments were performed to determine the role of inflammation in mood control in EAE. First, we assessed the independence of mood alterations from motor disability during the acute phase of the disease, by showing a depressive-like behavior in EAE mice with mild clinical score and preserved motor skills (mild-EAE). Second, we linked such behavioral changes to the selective increased striatal expression of interleukin-1beta (IL-1ß) in a context of mild inflammation and to dopaminergic system alterations. Indeed, in the striatum of EAE mice, we observed an impairment of dopamine (DA) neurotransmission, since DA release was reduced and signaling through DA D1- and D2-like receptors was unbalanced. In conclusion, the present study provides first evidence of the link between the depressive-like behavior and the alteration of dopaminergic system in EAE mice, raising the possibility that IL-1ß driven dysfunction of dopaminergic signaling might play a role in mood disturbances also in MS patients.


Asunto(s)
Cuerpo Estriado/inmunología , Depresión/metabolismo , Dopamina/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/psicología , Interleucina-1beta/metabolismo , Enfermedad Aguda , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/inmunología , Amígdala del Cerebelo/patología , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Depresión/tratamiento farmacológico , Depresión/patología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/patología , Interleucina-1beta/antagonistas & inhibidores , Ratones Endogámicos C57BL , Destreza Motora , ARN Mensajero/metabolismo , Distribución Aleatoria , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Índice de Severidad de la Enfermedad , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
6.
Neuropharmacology ; 79: 567-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24440366

RESUMEN

Type-1 cannabinoid receptors (CB1R) are important regulators of the neurodegenerative damage in multiple sclerosis (MS) and in experimental autoimmune encephalomyelitis (EAE). In GABAergic striatal neurons, CB1R stimulation exerts protective effects by limiting inflammation-induced potentiation of glutamate-mediated spontaneous excitatory postsynaptic currents (sEPSCs). Here we show that CB1R located on GABAergic or on glutamatergic neurons are differentially involved in the pre- and postsynaptic alterations of sEPSCs caused by EAE in the striatum. After induction of EAE, mice selectively lacking CB1R on GABAergic neurons (GABA-CB1R-KO) showed exacerbated alterations of sEPSC duration in GABAergic medium spiny neurons (MSN). On the other hand, EAE-induced alterations of corticostriatal sEPSC frequency were exacerbated only in mice lacking CB1R on glutamatergic neurons (Glu-CB1R-KO), indicating that this subset of receptors controls the effects of inflammation on glutamate release. While EAE severity was enhanced in whole CB1R-KO mice, GABA-CB1R-KO and Glu-CB1R-KO mice had similar motor deficits as the respective wild-type (WT) counterparts. Our results provide further evidence that CB1R are involved in EAE pathophysiology, and suggest that both pre- and postsynaptic alterations of glutamate transmission are important to drive excitotoxic neurodegeneration typical of this disorder.


Asunto(s)
Cuerpo Estriado/fisiopatología , Encefalomielitis Autoinmune Experimental/fisiopatología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Receptor Cannabinoide CB1/metabolismo , Transmisión Sináptica/fisiología , Animales , Progresión de la Enfermedad , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terminales Presinápticos/fisiología , Factores de Tiempo , Ácido gamma-Aminobutírico/metabolismo
7.
J Neuroimmunol ; 264(1-2): 8-13, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24045165

RESUMEN

Alterations of glutamate-mediated synaptic transmission occur in both multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the animal model of MS. Here we investigated whether intracerebroventricular (Icv) administration of cladribine has effects on EAE. Icv infusion of cladribine reduced the clinical deficits of EAE mice and reversed EAE-induced enhancement of excitatory postsynaptic current (sEPSC) frequency, a neurophysiological measure of glutamatergic synaptopathy associated with central inflammation. Cladribine failed to interfere with EAE-induced microglial and astroglial activation, but blocked EAE synaptic alterations by interfering with interleukin-1ß effects. Cladribine possesses neuroprotective properties in experimental MS that are independent of its peripheral immunosuppressant action.


Asunto(s)
Cladribina/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Interleucina-1beta/metabolismo , Sinapsis/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/citología , Quelantes/farmacología , Cuerpo Estriado/citología , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/farmacología , Inmunosupresores/farmacología , Técnicas In Vitro , Interleucina-1beta/farmacología , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Técnicas de Placa-Clamp
8.
J Neurosci ; 33(29): 12105-21, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864696

RESUMEN

Cerebellar deficit contributes significantly to disability in multiple sclerosis (MS). Several clinical and experimental studies have investigated the pathophysiology of cerebellar dysfunction in this neuroinflammatory disorder, but the cellular and molecular mechanisms are still unclear. In experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, proinflammatory cytokines, together with a degeneration of inhibitory neurons, contribute to impair GABAergic transmission at Purkinje cells (PCs). Here, we investigated glutamatergic transmission to gain insight into the pathophysiology of cerebellar dysfunction in EAE. Electrophysiological recordings from PCs showed increased duration of spontaneous excitatory postsynaptic currents (EPSCs) during the symptomatic phase of EAE, suggesting an alteration of glutamate uptake played by Bergmann glia. We indeed observed an impaired functioning of the glutamate-aspartate transporter/excitatory amino acid transporter 1 (GLAST/EAAT1) in EAE cerebellum caused by protein downregulation and in correlation with prominent astroglia activation. We have also demonstrated that the proinflammatory cytokine interleukin-1ß (IL-1ß), released by a subset of activated microglia/macrophages and infiltrating lymphocytes, was involved directly in such synaptic alteration. In fact, brief incubation of IL-1ß in normal cerebellar slices replicated EAE modifications through a rapid GLAST/EAAT1 downregulation, whereas incubation of an IL-1 receptor antagonist (IL-1ra) in EAE slices reduced spontaneous EPSC alterations. Finally, EAE mice treated with intracerebroventricular IL-1ra showed normal glutamatergic and GABAergic transmissions, along with GLAST/EAAT1 normalization, milder inflammation, and reduced motor deficits. These results highlight the crucial role played by the proinflammatory IL-1ß in triggering molecular and synaptic events involved in neurodegenerative processes that characterize neuroinflammatory diseases such as MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Ácido Glutámico/metabolismo , Interleucina-1beta/farmacología , Células de Purkinje/efectos de los fármacos , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Encefalomielitis Autoinmune Experimental/fisiopatología , Transportador 1 de Aminoácidos Excitadores/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ratones , Células de Purkinje/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología
9.
J Neuroimmune Pharmacol ; 8(3): 651-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23370991

RESUMEN

Glutamate-mediated excitotoxicity is supposed to induce neurodegeneration in multiple sclerosis (MS). Glatiramer acetate (GA) is an immunomodulatory agent used in MS treatment with potential neuroprotective action. Aim of the present study was to investigate whether GA has effects on glutamate transmission alterations occurring in experimental autoimmune encephalomyelitis (EAE), to disclose a possible mechanism of GA-induced neuroprotection in this mouse model of MS. Single neuron electrophysiological recordings and immunofluorescence analysis of microglia activation were performed in the striatum of EAE mice, treated or not with GA, at different stages of the disease. GA treatment was able to reverse the tumor necrosis factor-α (TNF-α)-induced alterations of striatal glutamate-mediated excitatory postsynaptic currents (EPSCs) of EAE mice. Incubation of striatal slices of control animals with lymphocytes taken from EAE mice treated with GA failed to replicate such an anti-glutamatergic effect, while activated microglial cells stimulated with GA in vitro mimicked the effect of GA treatment of EAE mice. Consistently, EAE mice treated with GA had less microglial activation and less TNF-α expression than untreated EAE animals. Furthermore, direct application of GA to EAE slices replicated the in vivo protective activity of GA. Our results show that GA is neuroprotective against glutamate toxicity independently of its peripheral immunodulatory action, and through direct modulation of microglial activation and TNF-α release in the grey matter of EAE and possibly of MS brains.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Inmunosupresores/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Péptidos/uso terapéutico , Sinapsis/patología , Animales , Células Cultivadas , Femenino , Acetato de Glatiramer , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Sinapsis/efectos de los fármacos
10.
Exp Neurol ; 237(2): 296-303, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22836148

RESUMEN

Multiple sclerosis (MS) causes a variety of motor and sensory deficits and it is also associated with mood disturbances. It is unclear if anxiety and depression in MS entirely reflect a subjective reaction to a chronic disease causing motor disability or rather depend on specific effects of neuroinflammation in neuronal circuits. To answer this question, behavioral, electrophysiological, and immunofluorescence experiments were performed in mice with experimental autoimmune encephalomyelitis (EAE), which models MS in mice. First, we observed high anxiety indexes in EAE mice, preceding the appearance of motor defects. Then, we demonstrated that tumor necrosis factor α (TNF-α) has a crucial role in anxiety associated with neuroinflammation. In fact, intracerebroventricular (icv) administration of etanercept, an inhibitor of TNF-α signaling, resulted in anxiolytic-like effects in EAE-mice. Accordingly, icv injection of TNF-α induced per se overt anxious behavior in control mice. Moreover, we propose the striatum as one of the brain regions potentially involved in EAE anxious behavior. We observed that before disease onset EAE striatum presents elevated TNF-α levels and strong activated microglia, early signs of inflammation associated with alterations of striatal excitatory postsynaptic currents (EPSCs). Interestingly, etanercept corrected the synaptic defects of pre-symptomatic EAE mice while icv injection of TNF-α in non-EAE mice altered EPSCs, thus mimicking the synaptic effects of EAE. In conclusion, anxiety characterizes EAE course since the very early phases of the disease. TNF-α released from activated microglia mediates this effect likely through the modulation of striatal excitatory synaptic transmission.


Asunto(s)
Ansiedad/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/psicología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ansiedad/metabolismo , Encefalomielitis Autoinmune Experimental/fisiopatología , Ensayo de Inmunoadsorción Enzimática , Etanercept , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoglobulina G/farmacología , Factores Inmunológicos/farmacología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/psicología , Técnicas de Placa-Clamp , Receptores del Factor de Necrosis Tumoral
11.
Neurobiol Dis ; 46(2): 414-24, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22349452

RESUMEN

A significant proportion of multiple sclerosis (MS) patients have functionally relevant cerebellar deficits, which significantly contribute to disability. Although clinical and experimental studies have been conducted to understand the pathophysiology of cerebellar dysfunction in MS, no electrophysiological and morphological studies have investigated potential alterations of synaptic connections of cerebellar Purkinje cells (PC). For this reason we analyzed cerebellar PC GABAergic connectivity in mice with MOG((35-55))-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We observed a strong reduction in the frequency of the spontaneous inhibitory post-synaptic currents (IPSCs) recorded from PCs during the symptomatic phase of the disease, and in presence of prominent microglia activation not only in the white matter (WM) but also in the molecular layer (ML). The massive GABAergic innervation on PCs from basket and stellate cells was reduced and associated to a decrease of the number of these inhibitory interneurons. On the contrary no significant loss of the PCs could be detected. Incubation of interleukin-1beta (IL-1ß) was sufficient to mimic the electrophysiological alterations observed in EAE mice. We thus suggest that microglia and pro-inflammatory cytokines, together with a degeneration of basket and stellate cells and their synaptic terminals, contribute to impair GABAergic transmission on PCs during EAE. Our results support a growing body of evidence that GABAergic signaling is compromised in EAE and in MS, and show a selective susceptibility to neuronal and synaptic degeneration of cerebellar inhibitory interneurons.


Asunto(s)
Encefalomielitis Autoinmune Experimental/fisiopatología , Neuronas GABAérgicas/patología , Células de Purkinje/fisiología , Transducción de Señal/fisiología , Animales , Encefalomielitis Autoinmune Experimental/patología , Femenino , Neuronas GABAérgicas/fisiología , Ratones , Ratones Endogámicos C57BL , Terminales Presinápticos/patología , Terminales Presinápticos/fisiología , Células de Purkinje/patología
12.
Neurobiol Dis ; 43(3): 669-77, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21672630

RESUMEN

Transient receptor potential vanilloid 1 (TRPV1) channels are involved in several inflammatory diseases. However, their action is still controversial, and both pro-inflammatory and anti-inflammatory roles have been described. We used a strain of TRPV1-KO mice to characterize the role of these channels in experimental autoimmune encephalomyelitis (EAE), which models multiple sclerosis (MS) in mice. EAE mice showed higher lethality in the peak phase of the disease and a better recovery of the surviving animals in the chronic stages, compared to their wild-type (WT) counterparts. By means of whole-cell patch clamp experiments in corticostriatal brain slices, we found that the absence of TRPV1 channels exacerbated the defect of glutamate transmission occurring in the peak phase of EAE, and attenuated the alterations of GABA synapses in the chronic phase of EAE, thus paralleling the dual effects of TRPV1-KO on the motor deficits of EAE mice. Furthermore, in slices from non-EAE mice, we found that genetic or pharmacological blockade of TRPV1 channels enhanced the synaptic effects of tumor necrosis factor α (TNF-α) on glutamate-mediated excitatory postsynaptic currents, and prevented the action of interleukin 1ß (IL-1ß) on GABAergic inhibitory postsynaptic currents. Together, our results suggest that TRPV1 channels contrast TNF-α-mediated synaptic deficits in the peak phase of EAE and, in the chronic stages, enhance IL-1ß-induced GABAergic defects. The opposing interplay with the synaptic actions of the two major pro-inflammatory cytokines might explain the bimodal effects of TRPV1 ablation on the motor deficits of EAE, and suggests that the inflammatory milieu determines whether TRPV1 channels exert preferentially aversive or protective effects on neurons during neuroinflammatory diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/prevención & control , Interleucina-1beta/fisiología , Sinapsis/fisiología , Canales Catiónicos TRPV/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Encefalomielitis Autoinmune Experimental/patología , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Inflamación/genética , Inflamación/metabolismo , Inflamación/fisiopatología , Potenciales Postsinápticos Inhibidores/genética , Potenciales Postsinápticos Inhibidores/fisiología , Interleucina-1beta/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Técnicas de Cultivo de Órganos , Sinapsis/genética , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...