Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 61(4): 2367-2389, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37874479

ABSTRACT

Structural epilepsies display complex immune activation signatures. However, it is unclear which neuroinflammatory pathways drive pathobiology. Transcriptome studies of brain resections from mesial temporal lobe epilepsy (mTLE) patients revealed a dysregulation of transforming growth factor ß, interferon α/ß, and nuclear factor erythroid 2-related factor 2 pathways. Since these pathways are regulated by ubiquitin-specific proteases (USP), in particular USP15, we hypothesized that USP15 blockade may provide therapeutic relief in treatment-resistant epilepsies. For validation, transgenic mice which either constitutively or inducibly lack Usp15 gene expression underwent intrahippocampal kainate injections to induce mTLE. We show that the severity of status epilepticus is unaltered in mice constitutively lacking Usp15 compared to wild types. Cell death, reactive gliosis, and changes in the inflammatory transcriptome were pronounced at 4 days after kainate injection. However, these brain inflammation signatures did not differ between genotypes. Likewise, induced deletion of Usp15 in chronic epilepsy did not affect seizure generation, cell death, gliosis, or the transcriptome. Concordantly, siRNA-mediated knockdown of Usp15 in a microglial cell line did not impact inflammatory responses in the form of cytokine release. Our data show that a lack of USP15 is insufficient to modulate the expression of relevant neuroinflammatory pathways in an mTLE mouse model and do not support targeting USP15 as a therapeutic approach for pharmacoresistant epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Animals , Humans , Mice , Down-Regulation , Gliosis , Hippocampus/metabolism , Kainic Acid , Mice, Transgenic , Ubiquitin-Specific Proteases/metabolism
2.
Biochim Biophys Acta Biomembr ; 1862(3): 183152, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31843475

ABSTRACT

Dopamine receptors (DRs) are class A G-Protein Coupled Receptors (GPCRs) prevalent in the central nervous system (CNS). These receptors mediate physiological functions ranging from voluntary movement and reward recognition to hormonal regulation and hypertension. Drugs targeting dopaminergic neurotransmission have been employed to treat several neurological and psychiatric disorders, including Parkinson's disease, schizophrenia, Huntington's disease, attention deficit hyperactivity disorder (ADHD), and Tourette's syndrome. In vivo, incorporation of GPCRs into lipid membranes is known to be key to their biological function and, by inference, maintenance of their tertiary structure. A further significant challenge in the structural and biochemical characterization of human DRs is their low levels of expression in mammalian cells. Thus, the purification and enrichment of DRs whilst retaining their structural integrity and function is highly desirable for biophysical studies. A promising new approach is the use of styrene-maleic acid (SMA) copolymer to solubilize GPCRs directly in their native environment, to produce polymer-assembled Lipodisqs (LQs). We have developed a novel methodology to yield detergent-free D1-containing Lipodisqs directly from HEK293f cells expressing wild-type human dopamine receptor 1 (D1). We demonstrate that D1 in the Lipodisq retains activity comparable to that in the native environment and report, for the first time, the affinity constant for the interaction of the peptide neurotransmitter neurotensin (NT) with D1, in the native state.


Subject(s)
Lipid Bilayers/chemistry , Receptors, Dopamine D1/isolation & purification , Receptors, Dopamine/isolation & purification , Cell Line , Detergents , HEK293 Cells , Humans , Maleates/chemistry , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Receptors, Dopamine/metabolism , Receptors, Dopamine D1/metabolism , Receptors, G-Protein-Coupled/isolation & purification , Styrenes/chemistry
3.
Epilepsia ; 60(7): 1412-1423, 2019 07.
Article in English | MEDLINE | ID: mdl-31179549

ABSTRACT

OBJECTIVE: The cystine/glutamate antiporter system xc- could represent a new target for antiepileptogenic treatments due to its crucial roles in glutamate homeostasis and neuroinflammation. To demonstrate this, we compared epilepsy development and seizure susceptibility in xCT knockout mice (xCT-/- ) and in littermate controls (xCT+/+ ) in different chronic models of epilepsy. METHODS: Mice were surgically implanted with electrodes in the basolateral amygdala and chronically stimulated to develop self-sustained status epilepticus (SSSE); continuous video-electroencephalography monitoring was performed for 28 days after SE and hippocampal histopathology was assessed. Corneal kindling was induced by twice daily electrical stimulation at 6 Hz and maintenance of the fully kindled state was evaluated. Next, messenger RNA (mRNA) and protein levels of xCT and of the proteins involved in the phosphoinositide 3-kinase (PI3K)/Akt/glycogen synthase kinase 3ß (GSK-3ß)/eukaryotic initiation factor 2α (eIF2α)/activating transcription factor 4 (ATF4) signaling pathway were measured at different time points during epileptogenesis in NMRI mice treated with pilocarpine. Finally, the anticonvulsant effect of sulfasalazine (SAS), a nonselective system xc- inhibitor, was assessed against 6 Hz-evoked seizures in pilocarpine-treated mice. RESULTS: In the SSSE model, xCT-/- mice displayed a significant delayed epileptogenesis, a reduced number of spontaneous recurrent seizures, and less pronounced astrocytic and microglial activation. Moreover, xCT-/- mice showed reduced seizure severity during 6 Hz kindling development and a lower incidence of generalized seizures during the maintenance of the fully kindled state. In pilocarpine-treated mice, protein levels of the PI3K/Akt/GSK-3ß/eIF2α/ATF4 pathway were increased during the chronic phase of the model, consistent with previous findings in the hippocampus of patients with epilepsy. Finally, repeated administration of SAS protected pilocarpine-treated mice against acute 6 Hz seizure induction, in contrast to sham controls, in which system xc- is not activated. SIGNIFICANCE: Inhibition of system xc- could be an attractive target for the development of new therapies with a potential for disease modification in epilepsy.


Subject(s)
Amino Acid Transport System y+/drug effects , Anticonvulsants/pharmacology , Epilepsy/drug therapy , Amino Acid Transport System y+/metabolism , Animals , Anticonvulsants/therapeutic use , Disease Models, Animal , Electroencephalography , Epilepsy/etiology , Epilepsy/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pilocarpine/pharmacology , Status Epilepticus/drug therapy , Status Epilepticus/etiology , Status Epilepticus/metabolism
4.
Brain ; 141(11): 3130-3143, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30307467

ABSTRACT

Epilepsy therapy is based on drugs that treat the symptoms rather than the underlying mechanisms of the disease (epileptogenesis). There are no treatments for preventing seizures or improving disease prognosis, including neurological comorbidities. The search of pathogenic mechanisms of epileptogenesis highlighted that neuroinflammatory cytokines [i.e. interleukin-1ß (IL-1ß), tumour necrosis factor-α (Tnf-α)] are induced in human and experimental epilepsies, and contribute to seizure generation in animal models. A major role in controlling the inflammatory response is played by specialized pro-resolving lipid mediators acting on specific G-protein coupled receptors. Of note, the role that these pathways have in epileptogenic tissue remains largely unexplored. Using a murine model of epilepsy, we show that specialized pro-resolving mechanisms are activated by status epilepticus before the onset of spontaneous seizures, but with a marked delay as compared to the neuroinflammatory response. This was assessed by measuring the time course of mRNA levels of 5-lipoxygenase (Alox5) and 15-lipoxygenase (Alox15), the key biosynthetic enzymes of pro-resolving lipid mediators, versus Il1b and Tnfa transcripts and proteins. In the same hippocampal tissue, we found a similar delayed expression of two main pro-resolving receptors, the lipoxin A4 receptor/formyl peptide receptor 2 and the chemerin receptor. These receptors were also induced in the human hippocampus after status epilepticus and in patients with temporal lobe epilepsy. This evidence supports the hypothesis that the neuroinflammatory response is sustained by a failure to engage pro-resolving mechanisms during epileptogenesis. Lipidomic LC-MS/MS analysis showed that lipid mediator levels apt to resolve the neuroinflammatory response were also significantly altered in the hippocampus during epileptogenesis with a shift in the biosynthesis of several pro-resolving mediator families including the n-3 docosapentaenoic acid (DPA)-derived protectin D1. Of note, intracerebroventricular injection of this mediator during epileptogenesis in mice dose-dependently reduced the hippocampal expression of both Il1b and Tnfa mRNAs. This effect was associated with marked improvement in mouse weight recovery and rescue of cognitive deficit in the novel object recognition test. Notably, the frequency of spontaneous seizures was drastically reduced by 2-fold on average and the average seizure duration was shortened by 40% after treatment discontinuation. As a result, the total time spent in seizures was reduced by 3-fold in mice treated with n-3 DPA-derived protectin D1. Taken together, the present findings demonstrate that epilepsy is characterized by an inadequate engagement of resolution pathways. Boosting endogenous resolution responses significantly improved disease outcomes, providing novel treatment avenues.


Subject(s)
Anticonvulsants/therapeutic use , Docosahexaenoic Acids/therapeutic use , Encephalitis/drug therapy , Epilepsy/drug therapy , Animals , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , CD11b Antigen/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Encephalitis/chemically induced , Epilepsy/chemically induced , Epilepsy/complications , Epilepsy/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/pathology , Kainic Acid/toxicity , Leukotriene B4/therapeutic use , Lipid Metabolism/drug effects , Lipoxins/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Nat Commun ; 9(1): 3561, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30177815

ABSTRACT

The identification of drug targets is highly challenging, particularly for diseases of the brain. To address this problem, we developed and experimentally validated a general computational framework for drug target discovery that combines gene regulatory information with causal reasoning ("Causal Reasoning Analytical Framework for Target discovery"-CRAFT). Using a systems genetics approach and starting from gene expression data from the target tissue, CRAFT provides a predictive framework for identifying cell membrane receptors with a direction-specified influence over disease-related gene expression profiles. As proof of concept, we applied CRAFT to epilepsy and predicted the tyrosine kinase receptor Csf1R as a potential therapeutic target. The predicted effect of Csf1R blockade in attenuating epilepsy seizures was validated in three pre-clinical models of epilepsy. These results highlight CRAFT as a systems-level framework for target discovery and suggest Csf1R blockade as a novel therapeutic strategy in epilepsy. CRAFT is applicable to disease settings other than epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy, Temporal Lobe/genetics , Epilepsy/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Computer Simulation , Disease Models, Animal , Drug Discovery , Epilepsy/chemically induced , Epilepsy/drug therapy , Gene Expression Profiling , Gene Expression Regulation , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Mice , Molecular Targeted Therapy , Muscarinic Agonists/toxicity , Pilocarpine/toxicity , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Sequence Analysis, RNA , Systems Biology
6.
Epilepsia ; 59(9): e147-e151, 2018 09.
Article in English | MEDLINE | ID: mdl-30144048

ABSTRACT

Brivaracetam (BRV) and levetiracetam (LEV) are effective antiepileptic drugs that bind selectively to the synaptic vesicle 2A (SV2A) protein. BRV differs from LEV in preclinical studies in that it exhibits a more potent and complete seizure protection across animal models. We reported previously that an allosteric modulator of the SV2A protein had differential effects on BRV compared with LEV, suggesting that they act at different sites or with different conformations of the SV2A protein. If this is the case, then we hypothesized that mutations of specific amino acids in the SV2A protein may have differential effects on BRV and LEV binding by the modulator. Mutation of some amino acids identified previously in the binding site of racetams to the SV2A protein had marked effects on binding of both [3 H]BRV and [3 H]LEV (eg, W300F, F277A, G303A, F658A, Y462A, W666A, I663A, D670A, and V661A). However, 3 amino acids were identified (K694, I273, and S294) in which mutation lost the effect of the modulator on [3 H]LEV binding with no effect on the modulation of [3 H]BRV binding. These results confirm that BRV and LEV bind to the human synaptic vesicle 2A protein at closely related sites but interact with these sites in a different way.


Subject(s)
Anticonvulsants/pharmacology , Levetiracetam/pharmacology , Membrane Glycoproteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Pyrrolidinones/pharmacology , Anilides/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Protein Binding/drug effects , Protein Binding/genetics , Radioligand Assay , Transfection , Tritium/pharmacokinetics
7.
Neurotherapeutics ; 15(2): 470-488, 2018 04.
Article in English | MEDLINE | ID: mdl-29464573

ABSTRACT

Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1ß, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.


Subject(s)
Brain Injuries, Traumatic/metabolism , Encephalitis/metabolism , Epilepsy/metabolism , Hippocampus/metabolism , Animals , Astrocytes/metabolism , Brain Injuries, Traumatic/complications , Cell Death , Cytokines/metabolism , Disease Models, Animal , Encephalitis/complications , Epilepsy/etiology , Female , Male , Microglia/metabolism , Neurons/metabolism , Rats, Sprague-Dawley , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
8.
BMC Neurosci ; 14: 87, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23937191

ABSTRACT

BACKGROUND: SV2A, SV2B and SV2C are synaptic vesicle proteins that are structurally related to members of the major facilitator superfamily (MFS). The function and transported substrate of the SV2 proteins is not clearly defined although they are linked to neurotransmitters release in a presynaptic calcium concentration-dependent manner. SV2A and SV2B exhibit broad expression in the central nervous system while SV2C appears to be more restricted in defined areas such as striatum. SV2A knockout mice start to display generalized seizures at a late developmental stage, around post-natal day 7 (P7), and die around P15. More recently, SV2A was demonstrated to be the molecular target of levetiracetam, an approved anti-epileptic drug (AED). The purpose of this work was to precisely analyze and quantify the SV2A, SV2B and SV2C expression during brain development to understand the contribution of these proteins in brain development and their impact on epileptic seizures. RESULTS: First, we systematically analyzed by immunohistofluorescence, the SV2A, SV2B and SV2C expression during mouse brain development, from embryonic day 12 (E12) to P30. This semi-quantitative approach suggests a modulation of SV2A and SV2B expression in hippocampus around P7. This is the reason why we used various quantitative approaches (laser microdissection of whole hippocampus followed by qRT-PCR and western blot analysis) indicating that SV2A and SV2B expression increased between P5 and P7 and remained stable between P7 and P10. Moreover, the increase of SV2A expression in the hippocampus at P7 was mainly observed in the CA1 region while SV2B expression in this region remains stable. CONCLUSIONS: The observed alterations of SV2A expression in hippocampus are consistent with the appearance of seizures in SV2A-/- animals at early postnatal age and the hypothesis that SV2A absence favors epileptic seizures around P7.


Subject(s)
Brain/embryology , Brain/growth & development , Brain/metabolism , Membrane Glycoproteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Seizures/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Membrane Glycoproteins/analysis , Mice , Mice, Inbred BALB C , Mice, Knockout , Microdissection , Nerve Tissue Proteins/analysis , Protein Isoforms/analysis , Protein Isoforms/biosynthesis
9.
J Neurosci Res ; 91(3): 436-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239147

ABSTRACT

Many antiepileptic drugs (AEDs) exert their therapeutic activity by modifying the inactivation properties of voltage-gated sodium (Na(v) ) channels. Lacosamide is unique among AEDs in that it selectively enhances the slow inactivation component. Although numerous studies have investigated the effects of AEDs on Na(v) channel inactivation, a direct comparison of results cannot be made because of varying experimental conditions. In this study, the effects of different AEDs on Na(v) channel steady-state slow inactivation were investigated under identical experimental conditions using whole-cell patch-clamp in N1E-115 mouse neuroblastoma cells. All drugs were tested at 100 µM, and results were compared with those from time-matched control groups. Lacosamide significantly shifted the voltage dependence of Na(v) current (I(Na) ) slow inactivation toward more hyperpolarized potentials (by -33 ± 7 mV), whereas the maximal fraction of slow inactivated channels and the curve slope did not differ significantly. Neither SPM6953 (lacosamide inactive enantiomer), nor carbamazepine, nor zonisamide affected the voltage dependence of I(Na) slow inactivation, the maximal fraction of slow inactivated channels, or the curve slope. Phenytoin significantly increased the maximal fraction of slow inactivated channels (by 28% ± 9%) in a voltage-independent manner but did not affect the curve slope. Lamotrigine slightly increased the fraction of inactivated currents (by 15% ± 4%) and widened the range of the slow inactivation voltage dependence. Lamotrigine and rufinamide induced weak, but significant, shifts of I(Na) slow inactivation toward more depolarized potentials. The effects of lacosamide on Na(v) channel slow inactivation corroborate previous observations that lacosamide has a unique mode of action among AEDs that act on Na(v) channels.


Subject(s)
Acetamides/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Anticonvulsants/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Animals , Cell Line, Tumor , Lacosamide , Mice , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...