Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Neuropharmacology ; 238: 109642, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37392820

ABSTRACT

The involvement of the mGlu5 receptors in the pathophysiology of several forms of monogenic autism has been supported by numerous studies following the seminal observation that mGlu5 receptor-dependent long-term depression was enhanced in the hippocampus of mice modeling the fragile-X syndrome (FXS). Surprisingly, there are no studies examining the canonical signal transduction pathway activated by mGlu5 receptors (i.e. polyphosphoinositide - PI - hydrolysis) in mouse models of autism. We have developed a method for in vivo assessment of PI hydrolysis based on systemic injection of lithium chloride followed by treatment with the selective mGlu5 receptor PAM, VU0360172, and measurement of endogenous inositolmonophosphate (InsP) in brain tissue. Here, we report that mGlu5 receptor-mediated PI hydrolysis was blunted in the cerebral cortex, hippocampus, and corpus striatum of Ube3am-/p+ mice modeling Angelman syndrome (AS), and in the cerebral cortex and hippocampus of Fmr1 knockout mice modeling FXS. In vivo mGlu5 receptor-mediated stimulation of Akt on threonine 308 was also blunted in the hippocampus of FXS mice. These changes were associated with a significant increase in cortical and striatal Homer1 levels and striatal mGlu5 receptor and Gαq levels in AS mice, and with a reduction in cortical mGlu5 receptor and hippocampal Gαq levels, and an increase in cortical phospholipase-Cß and hippocampal Homer1 levels in FXS mice. This is the first evidence that the canonical transduction pathway activated by mGlu5 receptors is down-regulated in brain regions of mice modeling monogenic autism.


Subject(s)
Angelman Syndrome , Autistic Disorder , Fragile X Syndrome , Mice , Animals , Phosphatidylinositol Phosphates/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Hydrolysis , Disease Models, Animal , Mice, Knockout , Fragile X Syndrome/metabolism , Carrier Proteins , Fragile X Mental Retardation Protein/metabolism
2.
Curr Neuropharmacol ; 21(11): 2233-2236, 2023.
Article in English | MEDLINE | ID: mdl-35339181

ABSTRACT

BACKGROUND: Noradrenergic fibers originating from the locus coeruleus densely innervate limbic structures, including the piriform cortex, which is the limbic structure with the lowest seizure threshold. Noradrenaline (NA) modulates limbic seizures while stimulating autophagy through ß2- adrenergic receptors (AR). Since autophagy is related to seizure threshold, this perspective questions whether modulating ß2-AR focally within the anterior piriform cortex affects limbic seizures. OBJECTIVE: In this perspective, we analyzed a potential role for ß2-AR as an anticonvulsant target within the anterior piriform cortex, area tempestas (AT). METHODS: We developed this perspective based on current literature on the role of NA in limbic seizures and autophagy. The perspective is also grounded on preliminary data obtained by microinfusing within AT either a ß2-AR agonist (salbutamol) or a ß2-AR antagonist (butoxamine) 5 minutes before bicuculline. RESULTS: ß2-AR stimulation fully prevents limbic seizures induced by bicuculline micro-infusion in AT. Conversely, antagonism at ß2-AR worsens bicuculline-induced seizure severity and prolongs seizure duration, leading to self-sustaining status epilepticus. These data indicate a specific role for ß2-AR as an anticonvulsant in AT. CONCLUSION: NA counteracts limbic seizures. This relies on various receptors in different brain areas. The anterior piriform cortex plays a key role in patients affected by limbic epilepsy. The anticonvulsant effects of NA through ß2-AR may be related to the stimulation of the autophagy pathway. Recent literature and present data draw a perspective where ß2-AR stimulation while stimulating autophagy mitigates limbic seizures, focally within AT. The mechanism linking ß2-AR to autophagy and seizure modulation should be extensively investigated.


Subject(s)
Anticonvulsants , Norepinephrine , Rats , Animals , Humans , Norepinephrine/adverse effects , Norepinephrine/metabolism , Bicuculline/adverse effects , Rats, Sprague-Dawley , Seizures/drug therapy , Seizures/chemically induced , Receptors, Adrenergic
3.
Curr Neuropharmacol ; 21(1): 105-118, 2023.
Article in English | MEDLINE | ID: mdl-35579153

ABSTRACT

BACKGROUND: Previous studies suggest that different metabotropic glutamate (mGlu) receptor subtypes are potential drug targets for treating absence epilepsy. However, no information is available on mGlu3 receptors. OBJECTIVE: To examine whether (i) changes of mGlu3 receptor expression/signaling are found in the somatosensory cortex and thalamus of WAG/Rij rats developing spontaneous absence seizures; (ii) selective activation of mGlu3 receptors with LY2794193 affects the number and duration of spikewave discharges (SWDs) in WAG/Rij rats; and (iii) a genetic variant of GRM3 (encoding the mGlu3 receptor) is associated with absence epilepsy. METHODS: Animals: immunoblot analysis of mGlu3 receptors, GAT-1, GLAST, and GLT-1; realtime PCR analysis of mGlu3 mRNA levels; assessment of mGlu3 receptor signaling; EEG analysis of SWDs; assessment of depressive-like behavior. Humans: search for GRM3 and GRM5 missense variants in 196 patients with absence epilepsy or other Idiopathic Generalized Epilepsy (IGE)/ Genetic Generalized Epilepsy (GGE) and 125,748 controls. RESULTS: mGlu3 protein levels and mGlu3-mediated inhibition of cAMP formation were reduced in the thalamus and somatosensory cortex of pre-symptomatic (25-27 days old) and symptomatic (6-7 months old) WAG/Rij rats compared to age-matched controls. Treatment with LY2794193 (1 or 10 mg/kg, i.p.) reduced absence seizures and depressive-like behavior in WAG/Rij rats. LY2794193 also enhanced GAT1, GLAST, and GLT-1 protein levels in the thalamus and somatosensory cortex. GRM3 and GRM5 gene variants did not differ between epileptic patients and controls. CONCLUSION: We suggest that mGlu3 receptors modulate the activity of the cortico-thalamo-cortical circuit underlying SWDs and that selective mGlu3 receptor agonists are promising candidate drugs for absence epilepsy treatment.


Subject(s)
Epilepsy, Absence , Receptors, Metabotropic Glutamate , Rats , Humans , Animals , Infant , Epilepsy, Absence/drug therapy , Epilepsy, Absence/genetics , Epilepsy, Absence/metabolism , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Electroencephalography , Seizures , Human Genetics , Disease Models, Animal
4.
Curr Neuropharmacol ; 20(6): 1006-1010, 2022.
Article in English | MEDLINE | ID: mdl-34636300

ABSTRACT

BACKGROUND: The piriform cortex, known as area tempestas, has a high propensity to trigger limbic epileptic seizures. Recent studies on human patients indicate that a resection containing the piriform cortex produces a marked improvement in patients suffering from intractable limbic seizures. This calls for looking back at the pharmacological and anatomical data on area tempestas. Within the piriform cortex, status epilepticus can be induced by impairing the desensitization of AMPA receptors. The mechanistic target of rapamycin complex1 (mTORC1) is a promising candidate. OBJECTIVE: The present perspective aims to link the novel role of the piriform cortex with recent evidence on the modulation of AMPA receptors under the influence of mTORC1. This is based on recent evidence and preliminary data, leading to the formulation of interaction between mTORC1 and AMPA receptors to mitigate the onset of long-lasting, self-sustaining, neurotoxic status epilepticus. METHODS: The perspective grounds its method on recent literature along with the actual experimental procedure to elicit status epilepticus from the piriform cortex and the method to administer the mTORC1 inhibitor rapamycin to mitigate seizure expression and brain damage. RESULTS: The available and present perspectives converge to show that rapamycin may disrupt the seizure circuitry initiated in the piriform cortex to mitigate seizure duration, severity, and brain damage. CONCLUSION: The perspective provides a novel scenario to understand refractory epilepsy and selfsustaining status epilepticus. It is expected to provide a beneficial outcome in patients suffering from temporal lobe epilepsy.


Subject(s)
Receptors, AMPA , Status Epilepticus , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Seizures/chemically induced , Sirolimus/adverse effects , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , TOR Serine-Threonine Kinases/adverse effects , TOR Serine-Threonine Kinases/metabolism
5.
Curr Neuropharmacol ; 20(7): 1254-1260, 2022.
Article in English | MEDLINE | ID: mdl-34911428

ABSTRACT

BACKGROUND: Absence epilepsy is characterized by the presence of spike-and-wave discharges (SWDs) at the EEG generated within the cortico-thalamo-cortical circuit. The molecular mechanisms involved in the pathophysiology of absence epilepsy are only partially known. WAG/Rij rats older than 2-3 months develop spontaneous SWDs, and they are sensitive to anti- absence medications. Hence, WAG/Rij rats are extensively used as a model for absence epilepsy with predictive validity. OBJECTIVE: The aim of the study was to examine the possibility that the orexin system, which supports the wake status in experimental animals and humans, plays a role in the pathophysiology of absence seizures. METHODS: The perspective grounds its method from recent literature along with measurements of orexin receptor type-1 (OX1) protein levels in the thalamus and somatosensory cortex of WAG/Rij rats and non-epileptic Wistar control rats at two ages (25 days and 6-7 months). OX1 protein levels were measured by immunoblotting. RESULTS: The analysis of the current literature suggests that the orexin system might be involved in the pathophysiology of absence epilepsy and might be targeted by therapeutic intervention. Experimental data are in line with this hypothesis, showing that OX1 protein levels were reduced in the thalamus and somatosensory cortex of symptomatic WAG/Rij rats (6-7 months of age) with respect to non-epileptic controls, whereas these differences were not seen in pre-symptomatic, 25 days-old WAG/Rij rats. CONCLUSION: This perspective might pave the way for future studies on the involvement of the orexinergic system in the pathophysiology of SWDs associated with absence epilepsy and its comorbidities.


Subject(s)
Epilepsy, Absence , Animals , Disease Models, Animal , Electroencephalography/methods , Epilepsy, Absence/drug therapy , Humans , Orexins/therapeutic use , Rats , Rats, Wistar
6.
Neuropharmacology ; 196: 108686, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34197893

ABSTRACT

Absence epilepsy is frequently associated with cognitive dysfunction, although the underlying mechanisms are not well understood. Here we report that some forms of hippocampal synaptic plasticity are abnormal in symptomatic Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats. Metabotropic Glu 1/5 receptor-mediated long term depression (LTD) at Schaffer collateral CA1 synapses is significantly reduced in symptomatic, 5-6 months old WAG/Rij rats compared to age-matched non epileptic control rats. There were no significant changes in mGlu1/5-dependent LTD in pre-symptomatic, 4-6 weeks old WAG/Rij rats compared to age matched controls. The changes in LTD found in symptomatic WAG/Rij forms are not indicative of general deficits in all forms of synaptic plasticity as long term potentiation (LTP) was unchanged. Immunoblot analysis of hippocampal tissue showed a significant reduction in mGlu5 receptor expression, a trend to an increase in pan Homer protein levels and a decrease in GluA1 receptor expression in the hippocampus of symptomatic WAG/Rij rats vs non-epileptic control rats. There were no changes in mGlu1α receptor or GluA2 protein levels. These findings suggest that abnormalities in hippocampal mGlu5 receptor-dependent synaptic plasticity are associated with the pathological phenotype of WAG/Rij rats. This lays the groundwork for the study of mGlu5 receptors as a candidate drug target for the treatment of cognitive dysfunction linked to absence epilepsy.


Subject(s)
Epilepsy, Absence/metabolism , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Epilepsy, Absence/physiopathology , Homer Scaffolding Proteins/metabolism , Neuronal Plasticity/physiology , Rats
7.
Prog Neurobiol ; 204: 102112, 2021 09.
Article in English | MEDLINE | ID: mdl-34171442

ABSTRACT

As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.


Subject(s)
Autophagy , Neurodegenerative Diseases , Dopamine , Humans , Methamphetamine/toxicity , Neurodegenerative Diseases/chemically induced , Neuronal Plasticity , Neurotransmitter Agents
8.
Curr Neuropharmacol ; 19(6): 747-765, 2021.
Article in English | MEDLINE | ID: mdl-32867642

ABSTRACT

BACKGROUND: A dysfunction in glutamate neurotransmission is critical for seizure. Glutamate is the major excitatory drive in the cerebral cortex, where seizures occur. Glutamate acts via (i) ionotropic (iGlu) receptors, which are ligand-gated ion channels mediating fast excitatory synaptic transmission; and (ii) G proteins coupled metabotropic (mGlu) receptors. OBJECTIVE: To overview the evidence on the role of iGlu receptors in the onset, duration, and severity of convulsive and non-convulsive seizures to lay the groundwork for novel strategies for drug-resistant epilepsy. METHODS: We used PubMed crossed-search for "glutamate receptor and epilepsy" (sorting 3,170 reports), searched for "ionotropic glutamate receptors", "AMPA receptors", "NMDA receptors", "kainate receptors", "convulsive seizures", "absence epilepsy", and selected those papers focusing this Review's scope. RESULTS: iGlu receptor antagonists inhibit, whereas agonists worsen experimental seizures in various animal species. Clinical development of iGlu receptor antagonists has been limited by the occurrence of adverse effects caused by inhibition of fast excitatory synaptic transmission. To date, only one drug (perampanel) selectively targeting iGlu receptors is marketed for the treatment of focal epilepsy. However, other drugs, such as topiramate and felbamate, inhibit iGlu receptors in addition to other mechanisms. CONCLUSION: This review is expected to help dissect those steps induced by iGlu receptors activation, which may be altered to provide antiepileptic efficacy without altering key physiological brain functions, thus improving the safety and tolerability of iGlu-receptor directed antiepileptic agents. This effort mostly applies to drug resistant seizures, which impact the quality of life and often lead to status epilepticus, which is a medical urgency.


Subject(s)
Epilepsy, Absence , Receptors, Ionotropic Glutamate , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Epilepsy, Absence/drug therapy , Quality of Life , Seizures/drug therapy
9.
Neuropharmacology ; 178: 108240, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32768418

ABSTRACT

Previous studies have shown that injection of the mGlu5 receptor positive allosteric modulator (PAM) VU0360172 into either the thalamus or somatosensory cortex markedly reduces the frequency of spike-and-wave discharges (SWDs) in the WAG/Rij model of absence epilepsy. Here we have investigated the effects of VU0360172 on GABA transport in the thalamus and somatosensory cortex, as possible modes of action underlying the suppression of SWDs. Systemic VU0360172 injections increase GABA uptake in thalamic synaptosomes from epileptic WAG/Rij rats. Consistent with this observation, VU0360172 could also enhance thalamic GAT-1 protein expression, depending on the dosing regimen. This increase in GAT-1 expression was also observed in the thalamus from non-epileptic rats (presymptomatic WAG/Rij and Wistar) and appeared to occur selectively in neurons. The tonic GABAA receptor current present in ventrobasal thalamocortical neurons was significantly reduced by VU0360172 consistent with changes in GAT-1 and GABA uptake. The in vivo effects of VU0360172 (reduction in tonic GABA current and increase in GAT-1 expression) could be reproduced in vitro by treating thalamic slices with VU0360172 for at least 1 h and appeared to be dependent on the activation of PLC. Thus, the effects of VU0360172 do not require an intact thalamocortical circuit. In the somatosensory cortex, VU0360172 reduced GABA uptake but did not cause significant changes in GAT-1 protein levels. These findings reveal a novel mechanism of regulation mediated by mGlu5 receptors, which could underlie the powerful anti-absence effect of mGlu5 receptor enhancers in animal models.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Niacinamide/analogs & derivatives , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/metabolism , Thalamus/metabolism , gamma-Aminobutyric Acid/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Dose-Response Relationship, Drug , Male , Niacinamide/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Receptors, GABA-A/metabolism , Thalamus/drug effects , gamma-Aminobutyric Acid/pharmacology
10.
Expert Opin Ther Targets ; 23(4): 341-351, 2019 04.
Article in English | MEDLINE | ID: mdl-30801204

ABSTRACT

INTRODUCTION: Several drugs targeting the GABAergic system are used in the treatment of epilepsy, but only one drug targeting glutamate receptors is on the market. This is surprising because an imbalance between excitatory and inhibitory neurotransmission lies at the core of the pathophysiology of epilepsy. One possible explanation is that drug development has been directed towards the synthesis of molecules that inhibit the activity of ionotropic glutamate receptors. These receptors mediate fast excitatory synaptic transmission in the central nervous system (CNS) and their blockade may cause severe adverse effects such as sedation, cognitive impairment, and psychotomimetic effects. Metabotropic glutamate (mGlu) receptors are more promising drug targets because these receptors modulate synaptic transmission rather than mediate it. Areas covered: We review the current evidence that links mGlu receptor subtypes to the pathophysiology and experimental treatment of convulsive and absence seizures. Expert opinion: While mGlu5 receptor negative allosteric modulators have the potential to be protective against convulsive seizures and hyperactivity-induced neurodegeneration, drugs that enhance mGlu5 and mGlu7 receptor function may have beneficial effects in the treatment of absence epilepsy. Evidence related to the other mGlu receptor subtypes is more fragmentary; further investigations are required for an improved understanding of their role in the generation and propagation of seizures.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Receptors, Metabotropic Glutamate/drug effects , Animals , Epilepsy/physiopathology , Epilepsy, Absence/drug therapy , Epilepsy, Absence/physiopathology , Humans , Molecular Targeted Therapy , Receptors, Metabotropic Glutamate/metabolism , Synaptic Transmission/drug effects
11.
Int J Mol Sci ; 19(7)2018 Jul 06.
Article in English | MEDLINE | ID: mdl-29986414

ABSTRACT

Absence epilepsy and depression are comorbid disorders, but the molecular link between the two disorders is unknown. Here, we examined the role of the melatoninergic system in the pathophysiology of spike and wave discharges (SWDs) and depression-like behaviour in the Wistar Albino Glaxo from Rijswijk (WAG/Rij) rat model of absence epilepsy. In WAG/Rij rats, SWD incidence was higher during the dark period of the light-dark cycle, in agreement with previous findings. However, neither pinealectomy nor melatonin administration had any effect on SWD incidence, suggesting that the melatoninergic system was not involved in the pathophysiology of absence-like seizures. Endogenous melatonin levels were lower in the hippocampus of WAG/Rij rats as compared to non-epileptic control rats, and this was associated with higher levels of melatonin receptors in the hippocampus, but not in the thalamus. In line with the reduced melatonin levels, cell density was lower in the hippocampus of WAG/Rij rats and was further reduced by pinealectomy. As expected, WAG/Rij rats showed an increased depression-like behaviour in the sucrose preference and forced swim tests, as compared to non-epileptic controls. Pinealectomy abolished the difference between the two strains of rats by enhancing depression-like behaviour in non-epileptic controls. Melatonin replacement displayed a significant antidepressant-like effect in both WAG/Rij and control rats. These findings suggest that a defect of hippocampal melatoninergic system may be one of the mechanisms underlying the depression-like phenotype in WAG/Rij rats and that activation of melatonin receptors might represent a valuable strategy in the treatment of depression associated with absence epilepsy.


Subject(s)
Depression/metabolism , Epilepsy, Absence/metabolism , Hippocampus/metabolism , Melatonin/metabolism , Animals , Behavior Rating Scale , Disease Models, Animal , Male , Melatonin/administration & dosage , Pineal Gland/metabolism , Rats , Rats, Wistar
13.
Epilepsia ; 58(11): 1993-2001, 2017 11.
Article in English | MEDLINE | ID: mdl-28913875

ABSTRACT

OBJECTIVES: Thrombospondins, which are known to interact with the α2 δ subunit of voltage-sensitive calcium channels to stimulate the formation of excitatory synapses, have recently been implicated in the process of epileptogenesis. No studies have been so far performed on thrombospondins in models of absence epilepsy. We examined whether expression of the gene encoding for thrombospondin-1 was altered in the brain of WAG/Rij rats, which model absence epilepsy in humans. In addition, we examined the frequency of genetic variants of THBS1 in a large cohort of children affected by idiopathic/genetic generalized epilepsies (IGE/GGEs). METHODS: We measured the transcripts of thrombospondin-1 and α2 δ subunit, and protein levels of α2 δ, Rab3A, and the vesicular glutamate transporter, VGLUT1, in the somatosensory cortex and ventrobasal thalamus of presymptomatic and symptomatic WAG/Rij rats and in two control strains by real-time polymerase chain reaction (PCR) and immunoblotting. We examined the genetic variants of THBS1 and CACNA2D1 in two independent cohorts of patients affected by IGE/GGE recruited through the Genetic Commission of the Italian League Against Epilepsy (LICE) and the EuroEPINOMICS-CoGIE Consortium. RESULTS: Thrombospondin-1 messenger RNA (mRNA) levels were largely reduced in the ventrobasal thalamus of both presymptomatic and symptomatic WAG/Rij rats, whereas levels in the somatosensory cortex were unchanged. VGLUT1 protein levels were also reduced in the ventrobasal thalamus of WAG/Rij rats. Genetic variants of THBS1 were significantly more frequent in patients affected by IGE/GGE than in nonepileptic controls, whereas the frequency of CACNA2D1 was unchanged. SIGNIFICANCE: These findings suggest that thrombospondin-1 may have a role in the pathogenesis of IGE/GGEs.


Subject(s)
Calcium Channels/genetics , Disease Models, Animal , Epilepsy, Absence/genetics , Epilepsy, Generalized/genetics , Thrombospondin 1/genetics , Animals , Calcium Channels/biosynthesis , Cohort Studies , Epilepsy, Absence/metabolism , Epilepsy, Generalized/metabolism , Humans , Male , Rats , Rats, Wistar , Thrombospondin 1/biosynthesis
14.
Curr Neuropharmacol ; 15(6): 918-925, 2017.
Article in English | MEDLINE | ID: mdl-28290248

ABSTRACT

BACKGROUND: Spike-wave discharges, underlying absence seizures, are generated within a cortico-thalamo-cortical network that involves the somatosensory cortex, the reticular thalamic nucleus, and the ventrobasal thalamic nuclei. Activation of T-type voltage-sensitive calcium channels (VSCCs) contributes to the pathological oscillatory activity of this network, and some of the first-line drugs used in the treatment of absence epilepsy inhibit T-type calcium channels. The α2δ subunit is a component of high voltage-activated VSCCs (i.e., L-, N-, P/Q-, and R channels) and studies carried out in heterologous expression systems suggest that it may also associate with T channels. The α2δ subunit is also targeted by thrombospondins, which regulate synaptogenesis in the central nervous system. OBJECTIVE: To discuss the potential role for the thrombospondin/α2δ axis in the pathophysiology of absence epilepsy. METHODS: We searched PubMed articles for the terms "absence epilepsy", "T-type voltage-sensitive calcium channels", "α2δ subunit", "ducky mice", "pregabalin", "gabapentin", "thrombospondins", and included papers focusing this Review's scope. RESULTS: We moved from the evidence that mice lacking the α2δ-2 subunit show absence seizures and α 2δ ligands (gabapentin and pregabalin) are detrimental in the treatment of absence epilepsy. This suggests that α2δ may be protective against absence epilepsy via a mechanism that does not involve T channels. We discuss the interaction between thrombospondins and α2δ and its potential relevance in the regulation of excitatory synaptic formation in the cortico-thalamo-cortical network. CONCLUSION: We speculate on the possibility that the thrombospondin/α2 δ axis is critical for the correct functioning of the cortico-thalamo-cortical network, and that abnormalities in this axis may play a role in the pathophysiology of absence epilepsy.


Subject(s)
Calcium Channels/metabolism , Epilepsy, Absence/metabolism , Animals , Epilepsy, Absence/drug therapy , Humans , Thrombospondins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...