Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Neurobiol Dis ; 181: 106106, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37001613

ABSTRACT

Epilepsy is a comorbidity associated with Alzheimer's disease (AD), often starting many years earlier than memory decline. Investigating this association in the early pre-symptomatic stages of AD can unveil new mechanisms of the pathology as well as guide the use of antiepileptic drugs to prevent or delay hyperexcitability-related pathological effects of AD. We investigated the impact of repeated seizures on hippocampal memory and amyloid-ß (Aß) load in pre-symptomatic Tg2576 mice, a transgenic model of AD. Seizure induction caused memory deficits and an increase in oligomeric Aß42 and fibrillary species selectively in pre-symptomatic transgenic mice, and not in their wildtype littermates. Electrophysiological patch-clamp recordings in ex vivo CA1 pyramidal neurons and immunoblots were carried out to investigate the neuronal alterations associated with the behavioral outcomes of Tg2576 mice. CA1 pyramidal neurons exhibited increased intrinsic excitability and lower hyperpolarization-activated Ih current. CA1 also displayed lower expression of the hyperpolarization-activated cyclic nucleotide-gated HCN1 subunit, a protein already identified as downregulated in the AD human proteome. The antiepileptic drug lamotrigine restored electrophysiological alterations and prevented both memory deficits and the increase in extracellular Aß induced by seizures. Thus our study provides evidence of pre-symptomatic hippocampal neuronal alterations leading to hyperexcitability and associated with both higher susceptibility to seizures and to AD-specific seizure-induced memory impairment. Our findings also provide a basis for the use of the antiepileptic drug lamotrigine as a way to counteract acceleration of AD induced by seizures in the early phases of the pathology.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Anticonvulsants/pharmacology , Lamotrigine/adverse effects , Hippocampus/metabolism , Amyloid beta-Peptides/metabolism , Seizures/pathology , Mice, Transgenic , Disease Models, Animal , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/prevention & control
2.
Eur J Neurol ; 30(9): 2884-2898, 2023 09.
Article in English | MEDLINE | ID: mdl-37326003

ABSTRACT

BACKGROUND AND PURPOSE: Hormonal replacement therapy (HRT) is used for symptomatic treatment of menopause. Some evidence suggests a proconvulsant effect of estrogen and an anticonvulsant role of progesterone. Thus, the use of exogenous sex steroid hormones might influence the course of epilepsy in peri- and postmenopausal women with epilepsy (WWE). We conducted a systematic review on the impact of HRT on the frequency of seizures of WWE. METHODS: PubMed and Scopus were searched for articles published from inception until August 2022. Abstracts from the past 5 years from the European Academy of Neurology and European Epilepsy Congresses were also reviewed. Article reference lists were screened, and relevant articles were retrieved for consultation. Interventional and observational studies on WWE and animal models of estrogen deficiency were included. Critical appraisal was performed using the revised Cochrane risk-of-bias tool for randomized trials and ROBINS-E tool. RESULTS: Of 497 articles screened, 13 studies were included, including three human studies. One cross-sectional study showed a decrease in seizure frequency in WWE using combined HRT, a case-control study showed an increase in comparison with controls, and a randomized clinical trial found a dose-dependent increase in seizure frequency in women with focal epilepsy taking combined HRT. Ten studies addressing the impact of HRT in rat models were also included, which showed conflicting results. CONCLUSIONS: There is scarce evidence of the impact of HRT in WWE. Further studies should evaluate the harmful potential, and prospective registries are needed for monitoring this population.


Subject(s)
Epilepsy , Postmenopause , Female , Humans , Animals , Rats , Case-Control Studies , Prospective Studies , Cross-Sectional Studies , Epilepsy/drug therapy , Seizures/drug therapy , Estrogens/pharmacology , Estrogens/therapeutic use , Gonadal Steroid Hormones/pharmacology , Gonadal Steroid Hormones/therapeutic use , Randomized Controlled Trials as Topic
3.
Cereb Cortex ; 30(4): 2229-2249, 2020 04 14.
Article in English | MEDLINE | ID: mdl-33676371

ABSTRACT

Genetically distinct GABAergic interneuron subtypes play diverse roles in cortical circuits. Previous studies revealed that microRNAs (miRNAs) are differentially expressed in cortical interneuron subtypes, and are essential for the normal migration, maturation, and survival of medial ganglionic eminence-derived interneuron subtypes. How miRNAs function in vasoactive intestinal peptide expressing (VIP+) interneurons derived from the caudal ganglionic eminence remains elusive. Here, we conditionally removed Dicer in postmitotic VIP+ interneurons to block miRNA biogenesis. We found that the intrinsic and synaptic properties of VIP+ interneurons and pyramidal neurons were concordantly affected prior to a progressive loss of VIP+ interneurons. In vivo recording further revealed elevated cortical local field potential power. Mutant mice had a shorter life span but exhibited better spatial working memory and motor coordination. Our results demonstrate that miRNAs are indispensable for the function and survival of VIP+ interneurons, and highlight a key role of VIP+ interneurons in cortical circuits.


Subject(s)
Cerebral Cortex/metabolism , Interneurons/metabolism , MicroRNAs/antagonists & inhibitors , Nerve Net/metabolism , Vasoactive Intestinal Peptide/deficiency , Animals , Cerebral Cortex/growth & development , Male , Maze Learning/physiology , Mice , Mice, Transgenic , MicroRNAs/genetics , Nerve Net/growth & development , Vasoactive Intestinal Peptide/genetics
4.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917911

ABSTRACT

Temporal lobe epilepsy (TLE) is one of the most common types of focal epilepsy, characterized by recurrent spontaneous seizures originating in the temporal lobe(s), with mesial TLE (mTLE) as the worst form of TLE, often associated with hippocampal sclerosis. Abnormal epileptiform discharges are the result, among others, of altered cell-to-cell communication in both chemical and electrical transmissions. Current knowledge about the neurobiology of TLE in human patients emerges from pathological studies of biopsy specimens isolated from the epileptogenic zone or, in a few more recent investigations, from living subjects using positron emission tomography (PET). To overcome limitations related to the use of human tissue, animal models are of great help as they allow the selection of homogeneous samples still presenting a more various scenario of the epileptic syndrome, the presence of a comparable control group, and the availability of a greater amount of tissue for in vitro/ex vivo investigations. This review provides an overview of the structural and functional alterations of synaptic connections in the brain of TLE/mTLE patients and animal models.


Subject(s)
Disease Susceptibility , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Astrocytes/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Oligodendroglia/metabolism , Receptors, GABA/metabolism , Receptors, Ionotropic Glutamate/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
6.
Glia ; 64(8): 1350-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27246930

ABSTRACT

Activated microglia, astrogliosis, expression of pro-inflammatory cytokines, blood brain barrier (BBB) leakage and peripheral immune cell infiltration are features of mesial temporal lobe epilepsy. Numerous studies correlated the expression of pro-inflammatory cytokines with the activated morphology of microglia, attributing them a pro-epileptogenic role. However, microglia and myeloid cells such as macrophages have always been difficult to distinguish due to an overlap in expressed cell surface molecules. Thus, the detrimental role in epilepsy that is attributed to microglia might be shared with myeloid infiltrates. Here, we used a FACS-based approach to discriminate between microglia and myeloid infiltrates isolated from the hippocampus 24 h and 96 h after status epilepticus (SE) in pilocarpine-treated CD1 mice. We observed that microglia do not express MHCII whereas myeloid infiltrates express high levels of MHCII and CD40 96 h after SE. This antigen-presenting cell phenotype correlated with the presence of CD4(pos) T cells. Moreover, microglia only expressed TNFα 24 h after SE while myeloid infiltrates expressed high levels of IL-1ß and TNFα. Immunofluorescence showed that astrocytes but not microglia expressed IL-1ß. Myeloid infiltrates also expressed matrix metalloproteinase (MMP)-9 and 12 while microglia only expressed MMP-12, suggesting the involvement of both cell types in the BBB leakage that follows SE. Finally, both cell types expressed the phagocytosis receptor Axl, pointing to phagocytosis of apoptotic cells as one of the main functions of microglia. Our data suggests that, during early epileptogenesis, microglia from the hippocampus remain rather immune supressed whereas myeloid infiltrates display a strong inflammatory profile. GLIA 2016 GLIA 2016;64:1350-1362.


Subject(s)
Hippocampus/immunology , Microglia/immunology , Myeloid Cells/immunology , Status Epilepticus/immunology , Animals , Astrocytes/immunology , Astrocytes/pathology , CD40 Antigens/metabolism , Disease Models, Animal , Hippocampus/pathology , Interleukin-1beta/metabolism , Male , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 9 , Mice , Microglia/pathology , Myeloid Cells/pathology , Pilocarpine , Piriform Cortex/immunology , Piriform Cortex/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Status Epilepticus/pathology , Axl Receptor Tyrosine Kinase
7.
Cell Rep Med ; 4(5): 101047, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37196628

ABSTRACT

AMPA receptors' synaptic plasticity is involved in epileptogenesis. In this issue, Eiro et al.1 demonstrate that Hebbian plasticity is responsible for increased AMPAR in focal seizures, while homeostatic plasticity induces the reduction of AMPAR in generalized onset seizures.


Subject(s)
Receptors, AMPA , Synapses , Neuronal Plasticity , Homeostasis
8.
Cereb Cortex ; 21(7): 1574-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21112931

ABSTRACT

Human posttraumatic epilepsy (PTE) is highly heterogeneous, ranging from mild remitting to progressive disabling forms. PTE results in simple partial, complex partial, and secondarily generalized seizures with a wide spectrum of durations and semiologies. PTE variability is thought to depend on the heterogeneity of head injury and patient's age, gender, and genetic background. To better understand the role of these factors, we investigated the seizures resulting from calibrated fluid percussion injury (FPI) to adolescent male Sprague-Dawley rats with video electrocorticography. We show that PTE incidence and the frequency and severity of chronic seizures depend on the location and severity of FPI. The frontal neocortex was more prone to epileptogenesis than the parietal and occipital, generating earlier, longer, and more frequent partial seizures. A prominent limbic focus developed in most animals, regardless of parameters of injury. Remarkably, even with carefully controlled injury parameters, including type, severity, and location, the duration of posttraumatic apnea and the age and gender of outbred rats, there was great subject-to-subject variability in frequency, duration, and rate of progression of seizures, indicating that other factors, likely the subjects' genetic background and physiological states, have critical roles in determining the characteristics of PTE.


Subject(s)
Craniocerebral Trauma/pathology , Epilepsy, Post-Traumatic/pathology , Neocortex/pathology , Severity of Illness Index , Animals , Craniocerebral Trauma/complications , Craniocerebral Trauma/physiopathology , Electroencephalography/methods , Epilepsy, Post-Traumatic/etiology , Epilepsy, Post-Traumatic/physiopathology , Male , Neocortex/physiopathology , Rats , Rats, Sprague-Dawley
9.
J Pharmacol Exp Ther ; 336(3): 779-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21123672

ABSTRACT

Carisbamate (CRS) exhibits broad acute anticonvulsant activity in conventional anticonvulsant screens, genetic models of absence epilepsy and audiogenic seizures, and chronic spontaneous motor seizures arising after chemoconvulsant-induced status epilepticus. In add-on phase III trials with pharmacoresistant patients CRS induced < 30% average decreases in partial-onset seizure frequency. We assessed the antiepileptogenic and antiepileptic performance of subchronic CRS administration on posttraumatic epilepsy (PTE) induced by rostral parasaggital fluid percussion injury (rpFPI), which closely replicates human contusive closed head injury. Studies were blind and randomized, and treatment effects were assessed on the basis of sensitive electrocorticography (ECoG) recordings. Antiepileptogenic effects were assessed in independent groups of control and CRS-treated rats, at 1 and 3 months postinjury, after completion of a 2-week prophylactic treatment initiated 15 min after injury. The antiepileptic effects of 1-week CRS treatments were assessed in repeated measures experiments at 1 and 4 months postinjury. The studies were powered to detect ~50 and ~40% decreases in epilepsy incidence and frequency of seizures, respectively. Drug/vehicle treatment, ECoG analysis, and [CRS](plasma) determination all were performed blind. We detected no antiepileptogenic and an equivocal transient antiepileptic effects of CRS despite [CRS](plasma) comparable with or higher than levels attained in previous preclinical and clinical studies. These findings contrast with previous preclinical data demonstrating large efficacy of CRS, but agree with the average effect of CRS seen in clinical trials. The data support the use of rpFPI-induced PTE in the adolescent rat as a model of pharmacoresistant epilepsy for preclinical development.


Subject(s)
Anticonvulsants/therapeutic use , Carbamates/therapeutic use , Craniocerebral Trauma/drug therapy , Epilepsy, Post-Traumatic/drug therapy , Animals , Craniocerebral Trauma/complications , Craniocerebral Trauma/physiopathology , Epilepsy, Post-Traumatic/etiology , Epilepsy, Post-Traumatic/physiopathology , Male , Pilot Projects , Random Allocation , Rats , Rats, Sprague-Dawley , Single-Blind Method
10.
Adv Sci (Weinh) ; 8(10): 2002693, 2021 05.
Article in English | MEDLINE | ID: mdl-34026431

ABSTRACT

Neurological diseases are a prevalent cause of global mortality and are of growing concern when considering an ageing global population. Traditional treatments are accompanied by serious side effects including repeated treatment sessions, invasive surgeries, or infections. For example, in the case of deep brain stimulation, large, stiff, and battery powered neural probes recruit thousands of neurons with each pulse, and can invoke a vigorous immune response. This paper presents challenges in engineering and neuroscience in developing miniaturized and biointegrated alternatives, in the form of microelectrode probes. Progress in design and topology of neural implants has shifted the goal post toward highly specific recording and stimulation, targeting small groups of neurons and reducing the foreign body response with biomimetic design principles. Implantable device design recommendations, fabrication techniques, and clinical evaluation of the impact flexible, integrated probes will have on the treatment of neurological disorders are provided in this report. The choice of biocompatible material dictates fabrication techniques as novel methods reduce the complexity of manufacture. Wireless power, the final hurdle to truly implantable neural interfaces, is discussed. These aspects are the driving force behind continued research: significant breakthroughs in any one of these areas will revolutionize the treatment of neurological disorders.


Subject(s)
Brain/physiology , Deep Brain Stimulation/methods , Equipment Design/methods , Microelectrodes , Nervous System Diseases/therapy , Wireless Technology/instrumentation , Animals , Humans , Neurosciences/methods , Neurosciences/trends
11.
iScience ; 24(12): 103438, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34901791

ABSTRACT

Brain organoids are in vitro three-dimensional (3D) self-organized neural structures, which can enable disease modeling and drug screening. However, their use for standardized large-scale drug screening studies is limited by their high batch-to-batch variability, long differentiation time (10-20 weeks), and high production costs. This is particularly relevant when brain organoids are obtained from human induced pluripotent stem cells (iPSCs). Here, we developed, for the first time, a highly standardized, reproducible, and fast (5 weeks) murine brain organoid model starting from embryonic neural stem cells. We obtained brain organoids, which progressively differentiated and self-organized into 3D networks of functional neurons with dorsal forebrain phenotype. Furthermore, by adding the morphogen WNT3a, we generated brain organoids with specific hippocampal region identity. Overall, our results showed the establishment of a fast, robust and reproducible murine 3D in vitro brain model that may represent a useful tool for high-throughput drug screening and disease modeling.

12.
Cereb Cortex ; 19(7): 1515-20, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18787232

ABSTRACT

The absence of fragile X mental retardation protein results in the fragile X syndrome (FXS), a common form of mental retardation associated with attention deficit, autistic behavior, and epileptic seizures. The phenotype of FXS is reproduced in fragile X mental retardation 1 (fmr1) knockout (KO) mice that have region-specific altered expression of some gamma-aminobutyric acid (GABA(A)) receptor subunits. However, little is known about the characteristics of GABAergic inhibition in the subiculum of these animals. We employed patch-clamp recordings from subicular pyramidal cells in an in vitro slice preparation. In addition, semiquantitative polymerase chain reaction and western blot experiments were performed on subiculum obtained from wild-type (WT) and KO mice. We found that tonic GABA(A) currents were downregulated in fmr1 KO compared with WT neurons, whereas no significant differences were observed in phasic GABA(A) currents. Molecular biology analysis revealed that the tonic GABA(A) receptor subunits alpha5 and delta were underexpressed in the fmr1 KO mouse subiculum compared with WT. Because the subiculum plays a role in both cognitive functions and epileptic disorders, we propose that altered tonic inhibition in this structure contributes to the behavioral deficits and epileptic activity seen in FXS patients. This conclusion is in line with evidence implicating tonic GABA(A) inhibition in learning and memory.


Subject(s)
Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Neural Inhibition , Receptors, GABA-A/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism , Animals , Down-Regulation , Fragile X Mental Retardation Protein/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
CNS Drugs ; 23(7): 555-68, 2009.
Article in English | MEDLINE | ID: mdl-19552484

ABSTRACT

The mechanism of action of several antiepileptic drugs (AEDs) rests on their ability to modulate the activity of voltage-gated sodium currents that are responsible for fast action potential generation. Recent data indicate that lacosamide (a compound with analgesic and anticonvulsant effects in animal models) shares a similar mechanism. When compared with other AEDs, lacosamide has the unique ability to interact with sodium channel slow inactivation without affecting fast inactivation. This article reviews these findings and discusses their relevance within the context of neuronal activity seen during epileptiform discharges generated by limbic neuronal networks in the presence of chemical convulsants. These seizure-like events are characterized by sustained discharges of sodium-dependent action potentials supported by robust depolarizations, thus providing synchronization within neuronal networks. Generally, AEDs such as phenytoin, carbamazepine and lamotrigine block sodium channels when activated. In contrast, lacosamide facilitates slow inactivation of sodium channels both in terms of kinetics and voltage dependency. This effect may be relatively selective for repeatedly depolarized neurons, such as those participating in seizure activity in which the persistence of sodium currents is more pronounced and promotes neuronal excitation. The clinical effectiveness of lacosamide has been demonstrated in randomized, double-blind, parallel-group, placebo-controlled, adjunctive-therapy trials in patients with refractory partial seizures. Further studies should determine whether the effects of lacosamide in animal models and in clinical settings are fully explained by its selective action on sodium current slow inactivation or whether other effects (e.g. interactions with the collapsin-response mediator protein-2) play a contributory role.


Subject(s)
Acetamides/pharmacology , Acetamides/therapeutic use , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Ion Channel Gating/drug effects , Sodium Channels/drug effects , Animals , Clinical Trials as Topic , Disease Models, Animal , Epilepsy/metabolism , Humans , Lacosamide , Models, Molecular
14.
Epilepsia ; 50(3): 521-36, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19178556

ABSTRACT

PURPOSE: The anterior cingulate cortex (ACC)--which plays a role in pain, emotions and behavior--can generate epileptic seizures. To date, little is known on the neuronal mechanisms leading to epileptiform synchronization in this structure. Therefore, we investigated the role of excitatory and inhibitory synaptic transmission in epileptiform activity in this cortical area. In addition, since the ACC presents with a high density of opioid receptors, we studied the effect of opioid agonism on epileptiform synchronization in this brain region. METHODS: We used field and intracellular recordings in conjunction with pharmacological manipulations to characterize the epileptiform activity generated by the rat ACC in a brain slice preparation. RESULTS: Bath-application of the convulsant 4-aminopyridine (4AP, 50 microM) induced both brief and prolonged periods of epileptiform synchronization resembling interictal- and ictal-like discharges, respectively. Interictal events could occur more frequently before the onset of ictal activity that was contributed by N-methyl-D-aspartate (NMDA) receptors. Mu-opioid receptor activation abolished 4AP-induced ictal events and markedly reduced the occurrence of the pharmacologically isolated GABAergic synchronous potentials. Ictal discharges were replaced by interictal events during GABAergic antagonism; this GABA-independent activity was influenced by subsequent mu-opioid agonist application. CONCLUSIONS: Our results indicate that both glutamatergic and GABAergic signaling contribute to epileptiform synchronization leading to the generation of electrographic ictal events in the ACC. In addition, mu-opioid receptors appear to modulate both excitatory and inhibitory mechanisms, thus influencing epileptiform synchronization in the ACC.


Subject(s)
Cortical Synchronization , Electroencephalography , Epilepsies, Partial/physiopathology , Gyrus Cinguli/physiopathology , 4-Aminopyridine/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Anticonvulsants/pharmacology , Electroencephalography/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Epilepsies, Partial/chemically induced , Evoked Potentials/drug effects , Evoked Potentials/physiology , GABA Antagonists/pharmacology , Gyrus Cinguli/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neurons/physiology , Piperazines/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Receptors, GABA-B/drug effects , Receptors, GABA-B/physiology , Receptors, Glutamate/drug effects , Receptors, Glutamate/physiology , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Opioid/drug effects , Receptors, Opioid/physiology , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
15.
J Neurosci Methods ; 172(2): 143-57, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18550176

ABSTRACT

Understanding the pathophysiogenesis of temporal lobe epilepsy (TLE) largely rests on the use of models of status epilepticus (SE), as in the case of the pilocarpine model. The main features of TLE are: (i) epileptic foci in the limbic system; (ii) an "initial precipitating injury"; (iii) the so-called "latent period"; and (iv) the presence of hippocampal sclerosis leading to reorganization of neuronal networks. Many of these characteristics can be reproduced in rodents by systemic injection of pilocarpine; in this animal model, SE is followed by a latent period and later by the appearance of spontaneous recurrent seizures (SRSs). These processes are, however, influenced by experimental conditions such as rodent species, strain, gender, age, doses and routes of pilocarpine administration, as well as combinations with other drugs administered before and/or after SE. In the attempt to limit these sources of variability, we evaluated the methodological procedures used by several investigators in the pilocarpine model; in particular, we have focused on the behavioural, electrophysiological and histopathological findings obtained with different protocols. We addressed the various experimental approaches published to date, by comparing mortality rates, onset of SRSs, neuronal damage, and network reorganization. Based on the evidence reviewed here, we propose that the pilocarpine model can be a valuable tool to investigate the mechanisms involved in TLE, and even more so when standardized to reduce mortality at the time of pilocarpine injection, differences in latent period duration, variability in the lesion extent, and SRS frequency.


Subject(s)
Epilepsy, Temporal Lobe/chemically induced , Hippocampus/drug effects , Pilocarpine/pharmacology , Status Epilepticus/chemically induced , Animals , Convulsants/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Species Specificity , Status Epilepticus/pathology , Status Epilepticus/physiopathology , Time Factors
18.
Front Pharmacol ; 8: 676, 2017.
Article in English | MEDLINE | ID: mdl-29018345

ABSTRACT

Ghrelin, des-acyl ghrelin and other related peptides possess anticonvulsant activities. Although ghrelin and cognate peptides were shown to physiologically regulate only the ghrelin receptor, some of them were pharmacologically proved to activate the peroxisome proliferator-activated receptor gamma (PPARγ) through stimulation of the scavenger receptor CD36 in macrophages. In our study, we challenged the hypothesis that PPARγ could be involved in the anticonvulsant effects of EP-80317, a ghrelin receptor antagonist. For this purpose, we used the PPARγ antagonist GW9662 to evaluate the modulation of EP-80317 anticonvulsant properties in two different models. Firstly, the anticonvulsant effects of EP-80317 were studied in rats treated with pilocarpine to induce status epilepticus (SE). Secondly, the anticonvulsant activity of EP-80317 was ascertained in the repeated 6-Hz corneal stimulation model in mice. Behavioral and video electrocorticographic (ECoG) analyses were performed in both models. We also characterized levels of immunoreactivity for PPARγ in the hippocampus of 6-Hz corneally stimulated mice. EP-80317 predictably antagonized seizures in both models. Pretreatment with GW9662 counteracted almost all EP-80317 effects both in mice and rats. Only the effects of EP-80317 on power spectra of ECoGs recorded during repeated 6-Hz corneal stimulation were practically unaffected by GW9662 administration. Moreover, GW9662 alone produced a decrease in the latency of tonic-clonic seizures and accelerated the onset of SE in rats. Finally, in the hippocampus of mice treated with EP-80317 we found increased levels of PPARγ immunoreactivity. Overall, these results support the hypothesis that PPARγ is able to modulate seizures and mediates the anticonvulsant effects of EP-80317.

19.
Neuron ; 92(6): 1337-1351, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-27939580

ABSTRACT

A critical feature of neural networks is that they balance excitation and inhibition to prevent pathological dysfunction. How this is achieved is largely unknown, although deficits in the balance contribute to many neurological disorders. We show here that a microRNA (miR-101) is a key orchestrator of this essential feature, shaping the developing network to constrain excitation in the adult. Transient early blockade of miR-101 induces long-lasting hyper-excitability and persistent memory deficits. Using target site blockers in vivo, we identify multiple developmental programs regulated in parallel by miR-101 to achieve balanced networks. Repression of one target, NKCC1, initiates the switch in γ-aminobutyric acid (GABA) signaling, limits early spontaneous activity, and constrains dendritic growth. Kif1a and Ank2 are targeted to prevent excessive synapse formation. Simultaneous de-repression of these three targets completely phenocopies major dysfunctions produced by miR-101 blockade. Our results provide new mechanistic insight into brain development and suggest novel candidates for therapeutic intervention.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental/genetics , MicroRNAs/genetics , Animals , Ankyrins/genetics , Ankyrins/metabolism , Behavior, Animal , Brain/growth & development , Dendrites , Kinesins/genetics , Kinesins/metabolism , Mice , Nerve Net/growth & development , Nerve Net/metabolism , Neural Pathways/growth & development , Neural Pathways/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction , Sequence Analysis, RNA , Solute Carrier Family 12, Member 2/genetics , Solute Carrier Family 12, Member 2/metabolism , gamma-Aminobutyric Acid/metabolism
20.
J Neurosci ; 22(9): 3414-25, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11978818

ABSTRACT

Native rat lactotrophs express thyrotrophin-releasing hormone-dependent K+ currents consisting of fast and slow deactivating components that are both sensitive to the class III anti-arrhythmic drugs that block the eag-related gene (ERG) K+ current (I(ERG)). Here we describe in MMQ prolactin-releasing pituitary cells the isolation of the slowly deactivating long-lasting component (I(ERGS)), which, unlike the fast component (I(ERGF)), is insensitive to verapamil 2 microm but sensitive to a novel scorpion toxin (ErgTx-2) that hardly affects I(ERGF). The time constants of I(ERGS) activation, deactivation, and recovery from inactivation are more than one order of magnitude greater than in I(ERGF), and the voltage-dependent inactivation is left-shifted by approximately 25 mV. The very slow MMQ firing frequency (approximately 0.2 Hz) investigated in perforated patch is increased approximately four times by anti-arrhythmic agents, by ErgTx-2, and by the abrupt I(ERGS) deactivation. Prolactin secretion in the presence of anti-arrhythmics is three- to fourfold higher in comparison with controls. We provide evidence from I(ERGS) and I(ERGF) simulations in a firing model cell to indicate that only I(ERGS) has an accommodating role during the experimentally observed very slow firing. Thus, we suggest that I(ERGS) potently modulates both firing and prolactin release in lactotroph cells.


Subject(s)
Cation Transport Proteins , Membrane Transport Proteins , Pituitary Gland/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium/metabolism , Prolactin/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Brain/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , Computer Simulation , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Models, Neurological , Patch-Clamp Techniques , Pituitary Gland/cytology , Pituitary Gland/drug effects , Potassium Channels/drug effects , Potassium Channels/genetics , RNA/genetics , RNA/metabolism , Rats , Scorpion Venoms/isolation & purification , Scorpion Venoms/pharmacology , Tretinoin/metabolism , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL