Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 30(7-8): 649-658, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37029201

RESUMO

Gene therapy with AAV vectors carrying genes for neuropeptide Y and its receptor Y2 has been shown to inhibit seizures in multiple animal models of epilepsy. It is however unknown how the AAV serotype or the sequence order of these two transgenes in the expression cassette affects the actual parenchymal gene expression levels and the seizure-suppressant efficacy. To address these questions, we compared three viral vector serotypes (AAV1, AAV2 and AAV8) and two transgene sequence orders (NPY-IRES-Y2 and Y2-IRES-NPY) in a rat model of acutely induced seizures. Wistar male rats were injected bilaterally with viral vectors and 3 weeks later acute seizures were induced by a subcutaneous injection of kainate. The latency until 1st motor seizure, time spent in motor seizure and latency to status epilepticus were measured to evaluate the seizure-suppressing efficacy of these vectors compared to an empty cassette control vector. Based on the results, the effect of the AAV1-NPY-IRES-Y2 vector was further investigated by in vitro electrophysiology, and its ability to achieve transgene overexpression in resected human hippocampal tissue was evaluated. The AAV1-NPY-IRES-Y2 proved to be better to any other serotype or gene sequence considering both transgene expression and ability to suppress induced seizures in rats. The vector also demonstrated transgene-induced decrease of glutamate release from excitatory neuron terminals and significantly increased both NPY and Y2 expression in resected human hippocampal tissue from patients with drug-resistant temporal lobe epilepsy. These results validate the feasibility of NPY/Y2 receptor gene therapy as a therapeutic opportunity in focal epilepsies.


Assuntos
Epilepsia , Convulsões , Ratos , Masculino , Humanos , Animais , Sorogrupo , Ratos Wistar , Convulsões/genética , Convulsões/terapia , Epilepsia/terapia , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Terapia Genética/métodos , Hipocampo/metabolismo
2.
J Neurosci Res ; 100(10): 1921-1932, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35822521

RESUMO

Neuroglobin (Ngb) is found in the neurones of several different brain areas and is known to bind oxygen and other gaseous molecules and reactive oxygen species (ROS) in vitro, but it does not seem to act as a respiratory molecule for neurones. Using male and female Ngb-knockout (KO) mice, we addressed the role of Ngb in neuronal brain activity using behavioral tests but found no differences in general behaviors, memory processes, and anxiety-/depression-like behaviors. Oxidative stress and ROS play key roles in epileptogenesis, and oxidative injury produced by an excessive production of free radicals is involved in the initiation and progression of epilepsy. The ROS binding properties led us to hypothesize that lack of Ngb could affect central coping with excitatory stimuli. We consequently explored whether exposure to the excitatory molecule kainate (KA) would increase severity of seizures in mice lacking Ngb. We found that the duration and severity of seizures were increased, while the latency time to develop seizures was shortened in Ngb-KO compared to wildtype adult female mice. Consistently, c-fos expression after KA was significantly increased in Ngb-KO mice in the amygdala and piriform cortex, regions rich in Ngb and known to be centrally involved in seizure generation. Moreover, the measured c-fos expression levels were correlated with seizure susceptibility. With these new findings combined with previous studies we propose that Ngb could constitute an intrinsic defense mechanism against neuronal hyperexcitability and oxidative stress by buffering of ROS in amygdala and other Ngb-containing brain regions.


Assuntos
Neuroglobina , Convulsões , Animais , Feminino , Masculino , Camundongos , Neuroglobina/deficiência , Neuroglobina/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
4.
Sci Rep ; 10(1): 18041, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33093500

RESUMO

Classically, neurexins are thought to mediate synaptic connections through trans interactions with a number of different postsynaptic partners. Neurexins are cleaved by metalloproteases in an activity-dependent manner, releasing the soluble extracellular domain. Here, we report that in both immature (before synaptogenesis) and mature (after synaptogenesis) hippocampal neurons, the soluble neurexin-1ß ectodomain triggers acute Ca2+-influx at the dendritic/postsynaptic side. In both cases, neuroligin-1 expression was required. In immature neurons, calcium influx required N-type calcium channels and stimulated dendritic outgrowth and neuronal survival. In mature glutamatergic neurons the neurexin-1ß ectodomain stimulated calcium influx through NMDA-receptors, which increased presynaptic release probability. In contrast, prolonged exposure to the ectodomain led to inhibition of synaptic transmission. This secondary inhibition was activity- and neuroligin-1 dependent and caused by a reduction in the readily-releasable pool of vesicles. A synthetic peptide modeled after the neurexin-1ß:neuroligin-1 interaction site reproduced the cellular effects of the neurexin-1ß ectodomain. Collectively, our findings demonstrate that the soluble neurexin ectodomain stimulates growth of neurons and exerts acute and chronic effects on trans-synaptic signaling involved in setting synaptic strength.


Assuntos
Proteínas de Ligação ao Cálcio/farmacologia , Cálcio/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Moléculas de Adesão de Célula Nervosa/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Solubilidade , Estimulação Química
5.
Mol Ther Methods Clin Dev ; 15: 179-193, 2019 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-31660420

RESUMO

Gene therapy has been suggested as a plausible novel approach to achieve seizure control in patients with focal epilepsy that do not adequately respond to pharmacological treatment. We investigated the seizure-suppressant potential of combinatorial neuropeptide Y and Y2 receptor single vector gene therapy based on adeno-associated virus serotype 1 (AAV1) in rats. First, a dose-response study in the systemic kainate-induced acute seizure model was performed, whereby the 1012 genomic particles (gp)/mL titer of the vector was selected as an optimal concentration. Second, an efficacy study was performed in the intrahippocampal kainate chronic model of spontaneous recurrent seizures (SRSs), designed to reflect a likely clinical scenario, with magnetic resonance image (MRI)-guided focal unilateral administration of the vector in the hippocampus during the chronic stage of the disease. The efficacy study demonstrated a favorable outcome of the gene therapy, with a 31% responder rate (more than 50% reduction in SRS frequency) and 13% seizure-freedom rate, whereas no such effects were observed in the control animals. The inter-SRS and SRS cluster intervals were also significantly prolonged in the treated group compared to controls. In addition, the SRS duration was significantly reduced in the treated group but not in the controls. This study establishes the SRS-suppressant ability of the single vector combinatorial neuropeptide Y/Y2 receptor gene therapy in a clinically relevant chronic model of epilepsy.

6.
Behav Brain Res ; 367: 28-34, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30914309

RESUMO

Neuropeptide S (NPS) has shown anxiolytic-like effects in rodents after acute administration, but its long-term effects remain unknown. Gene therapy enables the targeted delivery of DNA to cell nuclei, and recombinant adeno-associated viral (rAAV) vectors have been identified as suitable tools for stable overexpression. Thus, to explore the effects of long-term expression of NPS, the present study examined anxiety- and depressive-like effects after rAAV-mediated NPS overexpression in the rat amygdala. Compared to rats injected with an empty control vector (rAAV-Empty), rAAV-NPS treatment was associated with reduced anxiety-like behavior in the elevated plus maze and light-dark box, but did not affect depressive-like behavior in the forced swim test. Importantly, rAAV-NPS did not cause confounding effects on locomotion or bodyweight as opposed to currently used anxiolytic drugs. Immunohistochemical stainings revealed NPS-positive cells in the central and basolateral region of the amygdala in rAAV-NPS but not rAAV-Empty rats, indicating successful transduction. Our study provides novel evidence for sustained anxiolytic-like properties of NPS by transgenic overexpression. These data suggest that rAAV-NPS application deserves further attention as a potential treatment strategy for anxiety in humans.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Depressão/metabolismo , Neuropeptídeos/metabolismo , Animais , Peso Corporal/fisiologia , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Locomoção/fisiologia , Masculino , Ratos , Ratos Wistar
7.
Physiol Behav ; 164(Pt A): 300-5, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27306083

RESUMO

It is well known that physical activity has several health benefits, yet many people do not exercise. Dopamine levels in the striatum of the brain are thought to be important for the motivation to exercise. Conversely, we hypothesized that muscle quality can affect the motivation to exercise through alterations of the brain dopamine levels specifically in the striatal region. To test this hypothesis, transgenic mice overexpressing an inactivatable dominant negative α2 AMPK construct (AMPK α2 KD) in muscles and littermate wildtype (WT) mice were tested. AMPK α2 KD mice have impaired running capacity and display reduced voluntary wheel running activity. Striatal content of dopamine and its metabolites were measured under basal physiological conditions and after cocaine-induced dopamine efflux from the ventral striatum by in vivo microdialysis. Moreover, cocaine-induced locomotor activity was tested in an open field test. Furthermore, we investigated maximal running capacity and voluntary running over a period of 19days. AMPK α2 KD mice ran 30% less in daily distance compared to WT. Furthermore, AMPK α2 KD mice showed significantly decreased locomotor activity in the open field test compared to WT when treated with saline or cocaine, respectively, but the increase induced by cocaine was similar in AMPK α2 KD and WT mice. The efflux of dopamine in ventral striatum after cocaine treatment increased similarly by 2.5-fold in the two genotypes, and basal levels of dopamine and its metabolites DOPAC and HVA were also similar between genotypes. These findings show that decreased AMPK activity in muscle leads to decreased voluntary activity which is not due to secondary abnormalities in dopamine levels in the ventral striatum or sensitivity to cocaine. Thus, decreased voluntary activity in AMPK muscle deficient mice is most likely unrelated to regulation of brain dopamine content and metabolism.


Assuntos
Proteínas Quinases Ativadas por AMP/deficiência , Corpo Estriado/metabolismo , Dopamina/metabolismo , Músculo Esquelético/metabolismo , Corrida/fisiologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Cocaína/farmacologia , Corpo Estriado/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Ácido Homovanílico/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutação
8.
Neurobiol Dis ; 86: 52-61, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26607785

RESUMO

Although novel treatment strategies based on the gene therapy approach for epilepsy has been encouraging, there is still a gap in demonstrating a proof-of-concept in a clinically relevant animal model and study design. In the present study, a conceptually novel framework reflecting a plausible clinical trial for gene therapy of temporal lobe epilepsy was explored: We investigated (i) whether the post intrahippocampal kainate-induced status epilepticus (SE) model of chronic epilepsy in rats could be clinically relevant; and (ii) whether a translationally designed neuropeptide Y (NPY)/Y2 receptor-based gene therapy approach targeting only the seizure-generating focus unilaterally can decrease seizure frequency in this chronic model of epilepsy. Our data suggest that the intrahippocampal kainate model resembles the disease development of human chronic mesial temporal lobe epilepsy (mTLE): (i) spontaneous seizures originate in the sclerotic hippocampus; (ii) only a part of the animals develops chronic epilepsy; (iii) animals show largely variable seizure frequency that (iv) tends to progressively increase over time. Despite significant hippocampal degeneration caused by the kainate injection, the use of MRI allowed targeting the recombinant adeno-associated viral (rAAV) vectors encoding NPY and Y2 receptor genes to the remaining dorsal and ventral hippocampal areas ipsilateral to the kainate injection. Continuous video-EEG monitoring demonstrated not only prevention of the progressive increase in seizure frequency in rAAV-NPY/Y2 treated animals as compared to the controls, but even 45% decrease of seizure frequency in 80% of the epileptic animals. This translationally designed study in a clinically relevant model of epilepsy suggests that simultaneous overexpression of NPY and Y2 receptors unilaterally in the seizure focus is a relevant and promising approach that can be further validated in more extensive preclinical studies to develop a future treatment strategy for severe, often pharmacoresistant focal epilepsy cases that cannot be offered alternative therapeutic options.


Assuntos
Córtex Cerebral/fisiopatologia , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/terapia , Terapia Genética/métodos , Receptores de Neuropeptídeo Y/genética , Animais , Córtex Cerebral/efeitos dos fármacos , Dependovirus/genética , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Vetores Genéticos/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Ácido Caínico/administração & dosagem , Masculino , Ratos , Ratos Wistar , Pesquisa Translacional Biomédica
9.
ASN Neuro ; 7(4)2015.
Artigo em Inglês | MEDLINE | ID: mdl-26311075

RESUMO

Neuropeptide Y (NPY) is expressed in mammalian retina but the location and potential modulatory effects of NPY receptor activation remain largely unknown. Retinal ganglion cell (RGC) death is a hallmark of several retinal degenerative diseases, particularly glaucoma. Using purified RGCs and ex vivo rat retinal preparations, we have measured RGC intracellular free calcium concentration ([Ca2+]i) and RGC spiking activity, respectively. We found that NPY attenuated the increase in the [Ca2+]i triggered by glutamate mainly via Y1 receptor activation. Moreover, (Leu31, Pro34)-NPY, a Y1/Y5 receptor agonist, increased the initial burst response of OFF-type RGCs, although no effect was observed on RGC spontaneous spiking activity. The Y1 receptor activation was also able to directly modulate RGC responses by attenuating the NMDA-induced increase in RGC spiking activity. These results suggest that Y1 receptor activation, at the level of inner or outer plexiform layers, leads to modulation of RGC receptive field properties. Using in vitro cultures of rat retinal explants exposed to NMDA, we found that NPY pretreatment prevented NMDA-induced cell death. However, in an animal model of retinal ischemia-reperfusion injury, pretreatment with NPY or (Leu31, Pro34)-NPY was not able to prevent apoptosis or rescue RGCs. In conclusion, we found modulatory effects of NPY application that for the first time were detected at the level of RGCs. However, further studies are needed to evaluate whether NPY neuroprotective actions detected in retinal explants can be translated into animal models of retinal degenerative diseases.


Assuntos
Receptores de Neuropeptídeo Y/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Eletrorretinografia , Regulação da Expressão Gênica/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Marcação In Situ das Extremidades Cortadas , Masculino , Neuropeptídeo Y/agonistas , Neuropeptídeo Y/análogos & derivados , Neuropeptídeo Y/antagonistas & inibidores , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/farmacologia , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Ratos Wistar , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/genética , Doenças Retinianas/patologia , Doenças Retinianas/fisiopatologia , Isótopos de Enxofre/farmacocinética , Fator de Transcrição Brn-3A/metabolismo
10.
J Neurosci ; 35(26): 9622-31, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26134645

RESUMO

Development of novel disease-modifying treatment strategies for neurological disorders, which at present have no cure, represents a major challenge for today's neurology. Translation of findings from animal models to humans represents an unresolved gap in most of the preclinical studies. Gene therapy is an evolving innovative approach that may prove useful for clinical applications. In animal models of temporal lobe epilepsy (TLE), gene therapy treatments based on viral vectors encoding NPY or galanin have been shown to effectively suppress seizures. However, how this translates to human TLE remains unknown. A unique possibility to validate these animal studies is provided by a surgical therapeutic approach, whereby resected epileptic tissue from temporal lobes of pharmacoresistant patients are available for neurophysiological studies in vitro. To test whether NPY and galanin have antiepileptic actions in human epileptic tissue as well, we applied these neuropeptides directly to human hippocampal slices in vitro. NPY strongly decreased stimulation-induced EPSPs in dentate gyrus and CA1 (up to 30 and 55%, respectively) via Y2 receptors, while galanin had no significant effect. Receptor autoradiographic binding revealed the presence of both NPY and galanin receptors, while functional receptor binding was only detected for NPY, suggesting that galanin receptor signaling may be impaired. These results underline the importance of validating findings from animal studies in human brain tissue, and advocate for NPY as a more appropriate candidate than galanin for future gene therapy trials in pharmacoresistant TLE patients.


Assuntos
Epilepsia/patologia , Galanina/farmacologia , Hipocampo/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Adolescente , Adulto , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Hipocampo/patologia , Humanos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos , Pessoa de Meia-Idade , Técnicas de Patch-Clamp , Ensaio Radioligante , Receptores de Galanina/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Isótopos de Enxofre/farmacocinética , Adulto Jovem
11.
Synapse ; 68(10): 427-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24985894

RESUMO

Kainate-induced seizures constitute a model of temporal lobe epilepsy where prominent changes are observed in the hippocampal neuropeptide Y (NPY) system. However, little is known about the functional state and signal transduction of the NPY receptor population resulting from kainate exposure. Thus, in this study, we explored functional NPY receptor activity in the mouse hippocampus and neocortex after kainate-induced seizures using NPY-stimulated [(35) S]GTPγS binding. Moreover, we also studied levels of [(125) I]-peptide YY (PYY) binding and NPY, Y1, Y2, and Y5 receptor mRNA in these kainate-treated mice. Functional NPY binding was unchanged up to 12 h post-kainate, but decreased significantly in all hippocampal regions after 24 h and 1 week. Similarly, a decrease in [(125) I]-PYY binding was found in the dentate gyrus (DG) 1 week post-kainate. However, at 2 h, 6 h, and 12 h, [(125) I]-PYY binding was increased in all regions, and in the CA1 also at 24 h post-kainate. NPY mRNA levels were prominently increased in hippocampal regions, reaching maximum at 12 and 24 h. Y1 and Y5 mRNA levels were lowered in the DG at 24 and 2 h, respectively, while Y2 mRNA levels were elevated at 24 h in the DG and CA3. This study confirms rat kainate studies by showing pronounced adaptive changes in the mouse hippocampus both with regard to NPY synthesis and NPY receptor synthesis and binding, which may contribute to regulating neuronal seizure susceptibility after kainate. However, the potential seizure-suppressant effects of increased NPY gene expression at late time points post-kainate could be attenuated by the novel finding of reduced NPY-receptor G-protein activation.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipocampo/metabolismo , Neuropeptídeo Y/metabolismo , Convulsões/metabolismo , Animais , Autorradiografia , Modelos Animais de Doenças , Ácido Caínico , Masculino , Camundongos , Neocórtex/metabolismo , Peptídeo YY/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Convulsões/induzido quimicamente , Fatores de Tempo
12.
Neurobiol Dis ; 45(1): 288-96, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21884793

RESUMO

We recently demonstrated that recombinant adeno-associated viral vector-induced hippocampal overexpression of neuropeptide Y receptor, Y2, exerts a seizure-suppressant effect in kindling and kainate-induced models of epilepsy in rats. Interestingly, additional overexpression of neuropeptide Y in the hippocampus strengthened the seizure-suppressant effect of transgene Y2 receptors. Here we show for the first time that another neuropeptide Y receptor, Y5, can also be overexpressed in the hippocampus. However, unlike Y2 receptor overexpression, transgene Y5 receptors in the hippocampus had no effect on kainate-induced motor seizures in rats. However, combined overexpression of Y5 receptors and neuropeptide Y exerted prominent suppression of seizures. This seizure-suppressant effect of combination gene therapy with Y5 receptors and neuropeptide Y was significantly stronger as compared to neuropeptide Y overexpression alone. These results suggest that overexpression of Y5 receptors in combination with neuropeptide Y could be an alternative approach for more effective suppression of hippocampal seizures.


Assuntos
Hipocampo/metabolismo , Excitação Neurológica/genética , Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/genética , Convulsões/genética , Animais , Ácido Caínico , Excitação Neurológica/metabolismo , Masculino , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Ratos , Ratos Transgênicos , Ratos Wistar , Receptores de Neuropeptídeo Y/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo
13.
Brain ; 133(9): 2778-88, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20688813

RESUMO

Gene therapy using recombinant adeno-associated viral vectors overexpressing neuropeptide Y in the hippocampus exerts seizure-suppressant effects in rodent epilepsy models and is currently considered for clinical application in patients with intractable mesial temporal lobe epilepsy. Seizure suppression by neuropeptide Y in the hippocampus is predominantly mediated by Y2 receptors, which, together with neuropeptide Y, are upregulated after seizures as a compensatory mechanism. To explore whether such upregulation could prevent seizures, we overexpressed Y2 receptors in the hippocampus using recombinant adeno-associated viral vectors. In two temporal lobe epilepsy models, electrical kindling and kainate-induced seizures, vector-based transduction of Y2 receptor complementary DNA in the hippocampus of adult rats exerted seizure-suppressant effects. Simultaneous overexpression of Y2 and neuropeptide Y had a more pronounced seizure-suppressant effect. These results demonstrate that overexpression of Y2 receptors (alone or in combination with neuropeptide Y) could be an alternative strategy for epilepsy treatment.


Assuntos
Terapia Genética/métodos , Hipocampo/metabolismo , Receptores de Neuropeptídeo Y/uso terapêutico , Convulsões/patologia , Convulsões/terapia , Análise de Variância , Animais , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Vetores Genéticos/fisiologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Ácido Caínico/efeitos adversos , Excitação Neurológica/genética , Excitação Neurológica/fisiologia , Masculino , Ligação Proteica/genética , Radiografia/métodos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Convulsões/etiologia , Isótopos de Enxofre/farmacocinética , Transcrição Gênica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA