Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Neurosci ; 33(29): 12105-21, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864696

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Glutamic Acid/metabolism , Interleukin-1beta/pharmacology , Purkinje Cells/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Animals , Cerebellum/drug effects , Cerebellum/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Mice , Purkinje Cells/metabolism , Synapses/physiology , Synaptic Transmission/physiology
2.
Neurobiol Dis ; 46(2): 414-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22349452

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , GABAergic Neurons/pathology , Purkinje Cells/physiology , Signal Transduction/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , GABAergic Neurons/physiology , Mice , Mice, Inbred C57BL , Presynaptic Terminals/pathology , Presynaptic Terminals/physiology , Purkinje Cells/pathology
3.
Neurobiol Dis ; 43(3): 669-77, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21672630

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-1beta/physiology , Synapses/physiology , TRPV Cation Channels/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Disease Models, Animal , Down-Regulation/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Female , Inflammation/genetics , Inflammation/metabolism , Inflammation/physiopathology , Inhibitory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/physiology , Interleukin-1beta/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Synapses/genetics , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation/genetics
4.
Brain Behav Immun ; 25(5): 947-56, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20940040

ABSTRACT

Synaptic dysfunction triggers neuronal damage in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). While excessive glutamate signaling has been reported in the striatum of EAE, it is still uncertain whether GABA synapses are altered. Electrophysiological recordings showed a reduction of spontaneous GABAergic synaptic currents (sIPSCs) recorded from striatal projection neurons of mice with MOG((35-55))-induced EAE. GABAergic sIPSC deficits started in the acute phase of the disease (20-25days post immunization, dpi), and were exacerbated at later time-points (35, 50, 70 and 90dpi). Of note, in slices they were independent of microglial activation and of release of TNF-α. Indeed, sIPSC inhibition likely involved synaptic inputs arising from GABAergic interneurons, because EAE preferentially reduced sIPSCs of high amplitude, and was associated with a selective loss of striatal parvalbumin (PV)-positive GABAergic interneurons, which contact striatal projection neurons in their somatic region, giving rise to more efficient synaptic inhibition. Furthermore, we found also that the chronic persistence of pro-inflammatory cytokines were able, per se, to produce profound alterations of electrophysiological network properties, that were reverted by GABA administration. The results of the present investigation indicate defective GABA transmission in MS models depending from alteration of PV cells number and, in part, deriving from the effects of a chronic inflammation, and suggest that pharmacological agents potentiating GABA signaling might be considered to limit neuronal damage in MS patients.


Subject(s)
Corpus Striatum/physiopathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/physiology , Animals , Blotting, Western , Cells, Cultured , Corpus Striatum/metabolism , Cytokines/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/physiology , Neurons/metabolism , Neurons/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/physiology
5.
J Neurosci ; 29(32): 10131-43, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19675247

ABSTRACT

Understanding the modulation of the neural circuitry of fear is clearly one of the most important aims in neurobiology. Protein phosphorylation in response to external stimuli is considered a major mechanism underlying dynamic changes in neural circuitry. TrkB (Ntrk2) neurotrophin receptor tyrosine kinase potently modulates synaptic plasticity and activates signal transduction pathways mainly through two phosphorylation sites [Y515/Shc site; Y816/PLCgamma (phospholipase Cgamma) site]. To identify the molecular pathways required for fear learning and amygdalar synaptic plasticity downstream of TrkB, we used highly defined genetic mouse models carrying single point mutations at one of these two sites (Y515F or Y816F) to examine the physiological relevance of pathways activated through these sites for pavlovian fear conditioning (FC), as well as for synaptic plasticity as measured by field recordings obtained from neurons of different amygdala nuclei. We show that a Y816F point mutation impairs acquisition of FC, amygdalar synaptic plasticity, and CaMKII signaling at synapses. In contrast, a Y515F point mutation affects consolidation but not acquisition of FC to tone, and also alters AKT signaling. Thus, TrkB receptors modulate specific phases of fear learning and amygdalar synaptic plasticity through two main phosphorylation docking sites.


Subject(s)
Amygdala/physiology , Fear , Learning/physiology , Membrane Glycoproteins/metabolism , Neuronal Plasticity/physiology , Protein-Tyrosine Kinases/metabolism , Synapses/physiology , Animals , Binding Sites/genetics , Binding Sites/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Classical/physiology , Hippocampus/physiology , In Vitro Techniques , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Glycoproteins/genetics , Memory/physiology , Mice , Mice, Mutant Strains , Phosphorylation/physiology , Point Mutation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Synaptic Transmission/physiology
6.
Sci Rep ; 6: 24701, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27099182

ABSTRACT

Cell assemblies manipulation by optogenetics is pivotal to advance neuroscience and neuroengineering. In in vivo applications, photostimulation often broadly addresses a population of cells simultaneously, leading to feed-forward and to reverberating responses in recurrent microcircuits. The former arise from direct activation of targets downstream, and are straightforward to interpret. The latter are consequence of feedback connectivity and may reflect a variety of time-scales and complex dynamical properties. We investigated wide-field photostimulation in cortical networks in vitro, employing substrate-integrated microelectrode arrays and long-term cultured neuronal networks. We characterized the effect of brief light pulses, while restricting the expression of channelrhodopsin to principal neurons. We evoked robust reverberating responses, oscillating in the physiological gamma frequency range, and found that such a frequency could be reliably manipulated varying the light pulse duration, not its intensity. By pharmacology, mathematical modelling, and intracellular recordings, we conclude that gamma oscillations likely emerge as in vivo from the excitatory-inhibitory interplay and that, unexpectedly, the light stimuli transiently facilitate excitatory synaptic transmission. Of relevance for in vitro models of (dys)functional cortical microcircuitry and in vivo manipulations of cell assemblies, we give for the first time evidence of network-level consequences of the alteration of synaptic physiology by optogenetics.


Subject(s)
Cerebral Cortex/physiology , Evoked Potentials , Neural Pathways , Neurons/physiology , Photic Stimulation , Animals , Cells, Cultured , Gamma Rhythm , Gene Expression , In Vitro Techniques , Light , Models, Neurological , Opsins/genetics , Opsins/metabolism , Rats , Receptors, GABA-A/metabolism , Synaptic Transmission
7.
J Neuroimmune Pharmacol ; 8(3): 651-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23370991

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunosuppressive Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , Synapses/pathology , Animals , Cells, Cultured , Female , Glatiramer Acetate , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Synapses/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL