Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Epilepsia ; 61(5): 914-923, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32297665

RESUMO

OBJECTIVE: The antiepileptic drug candidate, padsevonil, is the first in a novel class of drugs designed to interact with both presynaptic and postsynaptic therapeutic targets: synaptic vesicle 2 proteins and γ-aminobutyric acid type A receptors (GABAA Rs), respectively. Functional aspects of padsevonil at the postsynaptic target, GABAA Rs, were characterized in experiments reported here. METHODS: The effect of padsevonil on GABA-mediated Cl- currents was determined by patch clamp on recombinant human GABAA Rs (α1ß2γ2) stably expressed in a CHO-K1 cell line and on native GABAA Rs in cultured rat primary cortical neurons. Padsevonil selectivity for GABAA R subtypes was evaluated using a two-electrode voltage clamp on recombinant human GABAA Rs (α1-5/ß2/γ2) in Xenopus oocytes. RESULTS: In recombinant GABAA Rs, padsevonil did not evoke Cl- currents in the absence of the agonist GABA. However, when co-administered with GABA at effective concentration (EC)20 , padsevonil potentiated GABA responses by 167% (EC50 138 nmol/L) and demonstrated a relative efficacy of 41% compared with zolpidem, a reference benzodiazepine site agonist. Similarly, padsevonil demonstrated GABA-potentiating activity at native GABAA Rs (EC50 208 nmol/L) in cultured rat cortical neurons. Padsevonil also potentiated GABA (EC20 ) responses in GABAA Rs expressed in oocytes, with higher potency at α1- and α5-containing receptors (EC50 295 and 281 nmol/L) than at α2- and α3-containing receptors (EC50 1737 and 2089 nmol/L). Compared with chlordiazepoxide-a nonselective, full GABAA R agonist-the relative efficacy of padsevonil was 60% for α1ß2γ2, 26% for α2ß2γ2, 56% for α3ß2γ2, and 41% for α5ß2γ2; no activity was observed at benzodiazepine-insensitive α4ß2γ2 receptors. SIGNIFICANCE: Results of functional investigations on recombinant and native neuronal GABAA Rs show that padsevonil acts as a positive allosteric modulator of these receptors, with a partial agonist profile at the benzodiazepine site. These properties may confer better tolerability and lower potential for tolerance development compared with classic benzodiazepines currently used in the clinic.


Assuntos
Anticonvulsivantes/farmacologia , Imidazóis/farmacologia , Pirrolidinonas/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Tiadiazóis/farmacologia , Animais , Células CHO , Cricetulus , Feminino , Humanos , Neurônios/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Ratos Wistar , Receptores Pré-Sinápticos/efeitos dos fármacos , Proteínas Recombinantes , Potenciais Sinápticos/efeitos dos fármacos , Xenopus laevis
2.
Epilepsia ; 58(1): 27-41, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27864845

RESUMO

OBJECTIVE: In human epilepsy, pharmacoresistance to antiepileptic drug therapy is a major problem affecting ~30% of patients with epilepsy. Many classical antiepileptic drugs target voltage-gated sodium channels, and their potent activity in inhibiting high-frequency firing has been attributed to their strong use-dependent blocking action. In chronic epilepsy, a loss of use-dependent block has emerged as a potential cellular mechanism of pharmacoresistance for anticonvulsants acting on voltage-gated sodium channels. The anticonvulsant drug lacosamide (LCM) also targets sodium channels, but has been shown to preferentially affect sodium channel slow inactivation processes, in contrast to most other anticonvulsants. METHODS: We used whole-cell voltage clamp recordings in acutely isolated cells to investigate the effects of LCM on transient Na+ currents. Furthermore, we used whole-cell current clamp recordings to assess effects on repetitive action potential firing in hippocampal slices. RESULTS: We show here that LCM exerts its effects primarily via shifting the slow inactivation voltage dependence to more hyperpolarized potentials in hippocampal dentate granule cells from control and epileptic rats, and from patients with epilepsy. It is important to note that this activity of LCM was maintained in chronic experimental and human epilepsy. Furthermore, we demonstrate that the efficacy of LCM in inhibiting high-frequency firing is undiminished in chronic experimental and human epilepsy. SIGNIFICANCE: Taken together, these results show that LCM exhibits maintained efficacy in chronic epilepsy, in contrast to conventional use-dependent sodium channel blockers such as carbamazepine. They also establish that targeting slow inactivation may be a promising strategy for overcoming target mechanisms of pharmacoresistance.


Assuntos
Acetamidas/uso terapêutico , Anticonvulsivantes/uso terapêutico , Epilepsia/tratamento farmacológico , Potenciais da Membrana/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Acetamidas/farmacologia , Adulto , Análise de Variância , Animais , Anticonvulsivantes/farmacologia , Biofísica , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Epilepsia/induzido quimicamente , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Técnicas In Vitro , Lacosamida , Agonistas Muscarínicos/toxicidade , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Pilocarpina/toxicidade , Ratos Wistar
3.
Epilepsia ; 58(11): e157-e161, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28850675

RESUMO

Brivaracetam (BRV) is a selective, high-affinity ligand for synaptic vesicle protein 2A (SV2A), recently approved as adjunctive treatment for drug-refractory partial-onset seizures in adults. BRV binds SV2A with higher affinity than levetiracetam (LEV), and was shown to have a differential interaction with SV2A. Because LEV was reported to interact with multiple excitatory and inhibitory ligand-gated ion channels and that may impact its pharmacological profile, we were interested in determining whether BRV directly modulates inhibitory and excitatory ionotropic receptors in central neurons. Voltage-clamp experiments were performed in primary cultures of mouse hippocampal neurons. At a supratherapeutic concentration of 100 µm, BRV was devoid of any direct effect on currents gated by γ-aminobutyric acidergic type A, glycine, kainate, N-methyl-d-aspartate, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid. Similarly to LEV, BRV reveals a potent ability to oppose the action of negative modulators on the inhibitory receptors. In conclusion, these results show that BRV contrasts with LEV by not displaying any direct action on inhibitory or excitatory postsynaptic ligand-gated receptors at therapeutic concentrations and thereby support BRV's role as a selective SV2A ligand. These findings add further evidence to the validity of SV2A as a relevant antiepileptic drug target and emphasize the potential for exploring further presynaptic mechanisms as a novel approach to antiepileptic drug discovery.


Assuntos
Ácido Glutâmico/farmacologia , Glicina/farmacologia , Hipocampo/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Pirrolidinonas/farmacologia , Ácido gama-Aminobutírico/farmacologia , Animais , Anticonvulsivantes/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Glicoproteínas de Membrana/agonistas , Camundongos , Proteínas do Tecido Nervoso/agonistas , Neurônios/efeitos dos fármacos , Neurônios/fisiologia
4.
Epilepsia ; 57(4): 538-48, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26920914

RESUMO

Despite availability of effective antiepileptic drugs (AEDs), many patients with epilepsy continue to experience refractory seizures and adverse events. Achievement of better seizure control and fewer side effects is key to improving quality of life. This review describes the rationale for the discovery and preclinical profile of brivaracetam (BRV), currently under regulatory review as adjunctive therapy for adults with partial-onset seizures. The discovery of BRV was triggered by the novel mechanism of action and atypical properties of levetiracetam (LEV) in preclinical seizure and epilepsy models. LEV is associated with several mechanisms that may contribute to its antiepileptic properties and adverse effect profile. Early findings observed a moderate affinity for a unique brain-specific LEV binding site (LBS) that correlated with anticonvulsant effects in animal models of epilepsy. This provided a promising molecular target and rationale for identifying selective, high-affinity ligands for LBS with potential for improved antiepileptic properties. The later discovery that synaptic vesicle protein 2A (SV2A) was the molecular correlate of LBS confirmed the novelty of the target. A drug discovery program resulted in the identification of anticonvulsants, comprising two distinct families of high-affinity SV2A ligands possessing different pharmacologic properties. Among these, BRV differed significantly from LEV by its selective, high affinity and differential interaction with SV2A as well as a higher lipophilicity, correlating with more potent and complete seizure suppression, as well as a more rapid brain penetration in preclinical models. Initial studies in animal models also revealed BRV had a greater antiepileptogenic potential than LEV. These properties of BRV highlight its promising potential as an AED that might provide broad-spectrum efficacy, associated with a promising tolerability profile and a fast onset of action. BRV represents the first selective SV2A ligand for epilepsy treatment and may add a significant contribution to the existing armamentarium of AEDs.


Assuntos
Anticonvulsivantes/metabolismo , Descoberta de Drogas/tendências , Epilepsia/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pirrolidinonas/metabolismo , Animais , Anticonvulsivantes/uso terapêutico , Relação Dose-Resposta a Droga , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/tendências , Epilepsia/tratamento farmacológico , Humanos , Ligantes , Pirrolidinonas/uso terapêutico , Resultado do Tratamento
5.
Epilepsia ; 56(12): 1899-909, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26515103

RESUMO

OBJECTIVE: Brivaracetam (BRV) decreases seizure activity in a number of epilepsy models and binds to the synaptic vesicle glycoprotein 2A (SV2A) with a higher affinity than the antiepileptic drug levetiracetam (LEV). Experiments were performed to determine if BRV acted similarly to LEV to induce or augment short-term depression (STD) under high-frequency neuronal stimulation and slow synaptic vesicle recycling. METHODS: Electrophysiologic field excitatory postsynaptic potential (fEPSP) recordings were made from CA1 synapses in rat hippocampal slices loaded with BRV or LEV during intrinsic activity or with BRV actively loaded during hypertonic stimulation. STD was examined in response to 5 or 40 Hz stimulus trains. Presynaptic release of FM1-43 was visualized using two-photon microscopy to assess drug effects upon synaptic vesicle mobilization. RESULTS: When hippocampal slices were incubated in 0.1-30 µm BRV or 30 µm-1 mm LEV for 3 h, the relative CA1 field EPSPs decreased over the course of a high-frequency train of stimuli more than for control slices. This STD was frequency- and concentration-dependent, with BRV being 100-fold more potent than LEV. The extent of STD depended on the length of the incubation time for both drugs. Pretreatment with LEV occluded the effects of BRV. Repeated hypertonic sucrose treatments and train stimulation successfully unloaded BRV from recycling vesicles and reversed BRVs effects on STD, as previously reported for LEV. At their maximal concentrations, BRV slowed FM1-43 release to a greater extent than in slices loaded with LEV during prolonged stimulation. SIGNIFICANCE: BRV, similar to LEV, entered into recycling synaptic vesicles and produced a frequency-dependent decrement of synaptic transmission at 100-fold lower concentrations than LEV. In addition, BRV slowed synaptic vesicle mobilization more effectively than LEV, suggesting that these drugs may modify multiple functions of the synaptic vesicle protein SV2A to curb synaptic transmission and limit epileptic activity.


Assuntos
Anticonvulsivantes/farmacologia , Pirrolidinonas/farmacologia , Vesículas Sinápticas/efeitos dos fármacos , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiologia , Relação Dose-Resposta a Droga , Eletrofisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Levetiracetam , Masculino , Microscopia de Fluorescência , Piracetam/análogos & derivados , Piracetam/farmacologia , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo
6.
J Neurosci Res ; 91(3): 436-43, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23239147

RESUMO

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.


Assuntos
Acetamidas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Anticonvulsivantes/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/fisiologia , Animais , Linhagem Celular Tumoral , Lacosamida , Camundongos , Fatores de Tempo
7.
J Neurosci Res ; 90(12): 2317-27, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22847229

RESUMO

Synaptic vesicle protein 2 (SV2) is a glycoprotein that exists in three isoforms, SV2A, SV2B, and SV2C. SV2A knockout (KO) mice and SV2A/SV2B double KO (DKO) mice, but not SV2B KO animals, start to experience severe seizures and weight loss 7 days after birth and die at about postnatal day (P)14-P23. Because excitatory and inhibitory inputs play a major role in controlling neuronal excitability in the hippocampus, we examined the effects of SV2A and/or SV2B deletions on glutamatergic and GABA(A) neurotransmission in hippocampal CA1 pyramidal neurons. Spontaneous and miniature excitatory and inhibitory postsynaptic currents (sEPSCs, mEPSCs, sIPSCs, and mIPSCs, respectively) were recorded using the whole-cell patch-clamp technique in slices from P6-P14 mice. The frequency of sEPSCs was increased in SV2A KO and SV2A/SV2B DKO mice, but their amplitude was unchanged. Such changes were not observed in SV2B KOs. On the contrary, the frequency and amplitude of sIPSCs were decreased in SV2A KO and SV2A/SV2B DKO mice but not in SV2B KO animals, as reported previously for the CA3 region. Kinetic parameters of sIPSCs and sEPSCs were unchanged. Importantly, no changes were observed in any genotype when examining mEPSCs and mIPSCs. We conclude that action potential- and Ca(2+) -dependent glutamatergic and GABAergic synaptic transmission are differentially altered in the hippocampus of SV2A-deficient mice, whereas the mechanism of exocytosis itself is not changed. The altered balance between these major excitatory and inhibitory inputs is probably a contributing factor to seizures in SV2A KO and SV2A/SV2B DKO mice.


Assuntos
Região CA1 Hipocampal/citologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Glicoproteínas de Membrana/deficiência , Proteínas do Tecido Nervoso/deficiência , Células Piramidais/fisiologia , Potenciais de Ação , Animais , Sinalização do Cálcio , Genes Letais , Ácido Glutâmico/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Técnicas de Patch-Clamp , Isoformas de Proteínas/fisiologia , Vesículas Sinápticas/metabolismo
8.
Epilepsia ; 53(11): 1959-67, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23016711

RESUMO

PURPOSE: In chronic epilepsy, a substantial proportion of up to 30% of patients remain refractory to antiepileptic drugs (AEDs). An understanding of the mechanisms of pharmacoresistance requires precise knowledge of how AEDs interact with their targets. Many commonly used AEDs act on the transient and/or the persistent components of the voltage-gated Na(+) current (I(NaT) and I(NaP) , respectively). Lacosamide (LCM) is a novel AED with a unique mode of action in that it selectively enhances slow inactivation of fast transient Na(+) channels. Given that functional loss of accessory Na(+) channel subunits is a feature of a number of neurologic disorders, including epilepsy, we examined the effects of LCM versus carbamazepine (CBZ) on the persistent Na(+) current (I(NaP) ), in the presence and absence of accessory subunits within the channel complex. METHODS: Using patch-clamp recordings in intact hippocampal CA1 neurons of Scn1b null mice, I(NaP) was recorded using slow voltage ramps. Application of 100 µm CBZ or 300 µm LCM reduced the maximal I(NaP) conductance in both wild-type and control mice. KEY FINDINGS: As shown previously by our group in Scn1b null mice, CBZ induced a paradoxical increase of I(NaP) conductance in the subthreshold voltage range, resulting in an ineffective block of repetitive firing in Scn1b null neurons. In contrast, LCM did not exhibit such a paradoxical increase, and accordingly maintained efficacy in blocking repetitive firing in Scn1b null mice. SIGNIFICANCE: These results suggest that the novel anticonvulsant LCM maintains activity in the presence of impaired Na(+) channel ß(1) subunit expression and thus may offer an improved efficacy profile compared with CBZ in diseases associated with an impaired expression of ß sub-units as observed in epilepsy.


Assuntos
Acetamidas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Carbamazepina/farmacologia , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/deficiência , Potenciais de Ação/fisiologia , Animais , Células Cultivadas , Lacosamida , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Subunidades Proteicas/deficiência , Subunidades Proteicas/fisiologia , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/fisiologia
9.
CNS Neurosci Ther ; 25(4): 442-451, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30242974

RESUMO

AIMS: Kv1.1 (KCNA1) channels contribute to the control of neuronal excitability and have been associated with epilepsy. Kv1.1 channels can associate with the cytoplasmic Kvß1 subunit resulting in rapid inactivating A-type currents. We hypothesized that removal of channel inactivation, by modulating Kv1.1/Kvß1 interaction with a small molecule, would lead to decreased neuronal excitability and anticonvulsant activity. METHODS: We applied high-throughput screening to identify ligands able to modulate the Kv1.1-T1 domain/Kvß1 protein complex. We then selected a compound that was characterized on recombinant Kv1.1/Kvß1 channels by electrophysiology and further evaluated on sustained neuronal firing and on in vitro epileptiform activity using a high K+ -low Ca2+ model in hippocampal slices. RESULTS: We identified a novel compound able to modulate the interaction of the Kv1.1/Kvß1 complex and that produced a functional inhibition of Kv1.1/Kvß1 channel inactivation. We demonstrated that this compound reduced the sustained repetitive firing in hippocampal neurons and was able to abolish the development of in vitro epileptiform activity. CONCLUSIONS: This study describes a rational drug discovery approach for the identification of novel ligands that inhibit Kv1.1 channel inactivation and provides pharmacological evidence that such a mechanism translates into physiological effects by reducing in vitro epileptiform activity.


Assuntos
Potenciais de Ação/fisiologia , Descoberta de Drogas/métodos , Hipocampo/fisiologia , Canal de Potássio Kv1.1/fisiologia , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Ensaios de Triagem em Larga Escala/métodos , Hipocampo/efeitos dos fármacos , Humanos , Canal de Potássio Kv1.1/agonistas , Canal de Potássio Kv1.1/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Bloqueadores dos Canais de Potássio/farmacologia , Estrutura Secundária de Proteína , Ratos , Xenopus laevis
10.
Neurotherapeutics ; 15(2): 470-488, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29464573

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Encefalite/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Animais , Astrócitos/metabolismo , Lesões Encefálicas Traumáticas/complicações , Morte Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/complicações , Epilepsia/etiologia , Feminino , Masculino , Microglia/metabolismo , Neurônios/metabolismo , Ratos Sprague-Dawley , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia
11.
Nat Commun ; 9(1): 3561, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30177815

RESUMO

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.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia do Lobo Temporal/genética , Epilepsia/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Animais , Simulação por Computador , Modelos Animais de Doenças , Descoberta de Drogas , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Terapia de Alvo Molecular , Agonistas Muscarínicos/toxicidade , Pilocarpina/toxicidade , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Análise de Sequência de RNA , Biologia de Sistemas
12.
CNS Neurosci Ther ; 21(3): 241-51, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25444522

RESUMO

AIMS: Brivaracetam (BRV) is an antiepileptic drug in Phase III clinical development. BRV binds to synaptic vesicle 2A (SV2A) protein and is also suggested to inhibit voltage-gated sodium channels (VGSCs). To evaluate whether the effect of BRV on VGSCs represents a relevant mechanism participating in its antiepileptic properties, we explored the pharmacology of BRV on VGSCs in different cell systems and tested its efficacy at reducing the sustained repetitive firing (SRF). METHODS: Brivaracetam investigations on the voltage-gated sodium current (I(Na)) were performed in N1E-155 neuroblastoma cells, cultured rat cortical neurons, and adult mouse CA1 neurons. SRF was measured in cultured cortical neurons and in CA1 neurons. All BRV (100-300 µM) experiments were performed in comparison with 100 µM carbamazepine (CBZ). RESULTS: Brivaracetam and CBZ reduced IN a in N1E-115 cells (30% and 40%, respectively) and primary cortical neurons (21% and 47%, respectively) by modulating the fast-inactivated state of VGSCs. BRV, in contrast to CBZ, did not affect I(Na) in CA1 neurons and SRF in cortical and CA1 neurons. CBZ consistently inhibited neuronal SRF by 75-93%. CONCLUSIONS: The lack of effect of BRV on SRF in neurons suggests that the reported inhibition of BRV on VGSC currents does not contribute to its antiepileptic properties.


Assuntos
Anticonvulsivantes/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiologia , Carbamazepina/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Ensaios Clínicos Fase III como Assunto , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos Wistar , Técnicas de Cultura de Tecidos , Canais de Sódio Disparados por Voltagem/metabolismo
13.
Neuroreport ; 14(9): 1273-6, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12824774

RESUMO

The effect of levetiracetam on neuronal hypersynchrony and hyperexcitability was examined using simultaneous extra- and intracellular recordings in rat brain slices perfused with a high K+/low Ca2+ (HKLC) fluid. These findings were compared to results obtained with carbamazepine, valproate and clonazepam. The HKLC milieu induces in hippocampal CA3 area, spontaneous interictal bursts and epileptiform responses. Levetiracetam decreased the number of population spikes per extracellular response but did not affect the number of action potentials per intracellular burst. This contrasts the effects of the reference antiepileptic drugs, which depressed both the extracellular and the intracellular bursts. These results indicate that levetiracetam is distinct from classical antiepileptic drugs by a relatively selective effect on collective neuronal responses, rather than on single neuron activity and suggests a potentially novel desynchronizing effect that probably contributes to its antiepileptic action.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Piracetam/análogos & derivados , Piracetam/farmacologia , Potenciais de Ação/fisiologia , Animais , Hipocampo/fisiologia , Técnicas In Vitro , Levetiracetam , Masculino , Ratos , Ratos Sprague-Dawley
14.
Neuroreport ; 15(6): 1049-52, 2004 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15076732

RESUMO

The persistent Na+ current (INaP) has been proposed as the putative target of the anti-absence antiepileptic drugs. Accordingly, the effect of reference anti-absence drugs ethosuximide (ESM) and valproate (VPA), and of the new antiepileptic drug levetiracetam (LEV), on INaP have been tested in CA1 hippocampal neurons and compared to the classic anticonvulsant phenytoin (PHT) and the neuroprotective agent riluzole (RIL). Whole-cell patch-clamp recordings of the slowly inactivating current, fully characterized as INaP, were performed with a standard voltage-step protocol on thin hippocampal slices prepared from rat brain. Both PHT (100 microM) and RIL (10 microM) strongly depressed INaP, whereas ESM (1 mM) induced a slight decrease of INaP and VPA (1 mM) had no effect. Likewise, 60-min perfusion with relevant concentrations of LEV (10, 32 or 100 microM) did not modify INaP. In conclusion, these data question the impact of INaP depression as an anti-absence mechanism, and also disclaim the involvement of INaP in the antiepileptic mechanism of LEV.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Anticonvulsivantes/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Canais de Sódio/fisiologia , Potenciais de Ação/fisiologia , Animais , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA