Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 112: 119-135, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29413716

RESUMO

Information about epileptogenesis-associated changes in protein expression patterns is of particular interest for future selection of target and biomarker candidates. Bioinformatic analysis of proteomic data sets can increase our knowledge about molecular alterations characterizing the different phases of epilepsy development following an initial epileptogenic insult. Here, we report findings from a focused analysis of proteomic data obtained for the hippocampus and parahippocampal cortex samples collected during the early post-insult phase, latency phase, and chronic phase of a rat model of epileptogenesis. The study focused on proteins functionally associated with cell stress, cell death, extracellular matrix (ECM) remodeling, cell-ECM interaction, cell-cell interaction, angiogenesis, and blood-brain barrier function. The analysis revealed prominent pathway enrichment providing information about the complex expression alterations of the respective protein groups. In the hippocampus, the number of differentially expressed proteins declined over time during the course of epileptogenesis. In contrast, a peak in the regulation of proteins linked with cell stress and death as well as ECM and cell-cell interaction became evident at later phases during epileptogenesis in the parahippocampal cortex. The data sets provide valuable information about the time course of protein expression patterns during epileptogenesis for a series of proteins. Moreover, the findings provide comprehensive novel information about expression alterations of proteins that have not been discussed yet in the context of epileptogenesis. These for instance include different members of the lamin protein family as well as the fermitin family member 2 (FERMT2). Induction of FERMT2 and other selected proteins, CD18 (ITGB2), CD44 and Nucleolin were confirmed by immunohistochemistry. Taken together, focused bioinformatic analysis of the proteomic data sets completes our knowledge about molecular alterations linked with cell death and cellular plasticity during epileptogenesis. The analysis provided can guide future selection of target and biomarker candidates.


Assuntos
Epilepsia/genética , Matriz Extracelular/genética , Perfilação da Expressão Gênica/métodos , Neovascularização Patológica/genética , Proteômica/métodos , Animais , Morte Celular/fisiologia , Epilepsia/metabolismo , Epilepsia/patologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley
2.
Epilepsia ; 59(4): 765-777, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29479675

RESUMO

OBJECTIVE: Rodent epilepsy models can significantly contribute to our understanding of pathophysiological mechanisms and to validation of biomarker and target candidates. Evidence-based severity assessment is a presupposition for the ethical evaluation of animal experimentation allowances as well as for the development of efficacious refinement concepts. METHODS: Aiming to improve our understanding of the impact of experimental procedures and repeated seizures, we have completed a comprehensive behavioral and biochemical analysis assessing various parameters that can inform about the influence of an electrical kindling paradigm on well-being in rats. Thereby, we have focused on the immediate effects of phases with focal and generalized seizures with behavioral testing during kindling acquisition. RESULTS: Electrode implantation exerted mild effects on anxiety-associated behavior and reduced serum corticosterone at 3 weeks, but not 7 weeks, following surgery. Analysis in kindled rats excluded any relevant impact of focal seizures on behavioral and biochemical parameters. Assessment in rats with generalized seizures revealed an impact on nest complexity scores, nest soiling, and selected parameters in paradigms evaluating anxiety-associated behavior. Moreover, serum corticosterone levels, but neither hair corticosterone nor fecal corticosterone metabolite concentrations were lowered as a consequence of repeated generalized seizures. The assessment of various other behavioral and biochemical parameters did not reveal any other relevant effects of generalized seizures. Cross-correlation analysis suggested that assessment of nest building and maintenance can provide information comparable to that from more elaborate behavioral assays. This finding provides first evidence that nest scoring might serve as a simple and valid approach to evaluate rat well-being during routine assessment schemes. SIGNIFICANCE: The findings argue against a persistent level of pronounced distress and suggest a classification of the kindling paradigm as a model with moderate severity based on a longer-lasting mild impact on animal behavioral patterns. This suggestion provides a basis for a prospective and retrospective case-by-case severity assessment.


Assuntos
Modelos Animais de Doenças , Relações Interpessoais , Excitação Neurológica/fisiologia , Convulsões/fisiopatologia , Índice de Gravidade de Doença , Animais , Eletrodos Implantados , Feminino , Ratos , Ratos Sprague-Dawley , Convulsões/psicologia
3.
Neurobiol Dis ; 105: 164-178, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28576708

RESUMO

Despite intense research efforts, the knowledge about the mechanisms of epileptogenesis and epilepsy is still considered incomplete and limited. However, an in-depth understanding of molecular pathophysiological processes is crucial for the rational selection of innovative biomarkers and target candidates. Here, we subjected proteomic data from different phases of a chronic rat epileptogenesis model to a comprehensive systems level analysis. Weighted Gene Co-expression Network analysis identified several modules of interconnected protein groups reflecting distinct molecular aspects of epileptogenesis in the hippocampus and the parahippocampal cortex. Characterization of these modules did not only further validate the data but also revealed regulation of molecular processes not described previously in the context of epilepsy development. The data sets also provide valuable information about temporal patterns, which should be taken into account for development of preventive strategies in particular when it comes to multi-targeting network pharmacology approaches. In addition, principal component analysis suggests candidate biomarkers, which might inform the design of novel molecular imaging approaches aiming to predict epileptogenesis during different phases or confirm epilepsy manifestation. Further studies are necessary to distinguish between molecular alterations, which correlate with epileptogenesis versus those reflecting a mere consequence of the status epilepticus.


Assuntos
Encéfalo/metabolismo , Proteoma/metabolismo , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia , Animais , Encéfalo/efeitos dos fármacos , Cromatografia Líquida , Modelos Animais de Doenças , Feminino , Redes Reguladoras de Genes , Agonistas Muscarínicos/toxicidade , Pilocarpina/toxicidade , Análise de Componente Principal , Proteoma/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estado Epiléptico/induzido quimicamente , Espectrometria de Massas em Tandem , Fatores de Tempo
4.
Neuroimage Clin ; 15: 35-44, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28462087

RESUMO

Excessive activation of inflammatory signaling pathways seems to be a hallmark of epileptogenesis. Positron emission tomography (PET) allows in vivo detection of brain inflammation with spatial information and opportunities for longitudinal follow-up scanning protocols. Here, we assessed whether molecular imaging of the 18 kDa translocator protein (TSPO) can serve as a biomarker for the development of epilepsy. Therefore, brain uptake of [18F]GE-180, a highly selective radioligand of TSPO, was investigated in a longitudinal PET study in a chronic rat model of temporal lobe epilepsy. Analyses revealed that the influence of the epileptogenic insult on [18F]GE-180 brain uptake was most pronounced in the earlier phase of epileptogenesis. Differences were evident in various brain regions during earlier phases of epileptogenesis with [18F]GE-180 standardized uptake value enhanced by 2.1 to 2.7fold. In contrast, brain regions exhibiting differences seemed to be more restricted with less pronounced increases of tracer uptake by 1.8-2.5fold four weeks following status epilepticus and by 1.5-1.8fold in the chronic phase. Based on correlation analysis, we were able to identify regions with a predictive value showing a correlation with seizure development. These regions include the amygdala as well as a cluster of brain areas. This cluster comprises parts of different brain regions, e.g. the hippocampus, parietal cortex, thalamus, and somatosensory cortex. In conclusion, the data provide evidence that [18F]GE-180 PET brain imaging can serve as a biomarker of epileptogenesis. The identification of brain regions with predictive value might facilitate the development of preventive concepts as well as the early assessment of the interventional success. Future studies are necessary to further confirm the predictivity of the approach.


Assuntos
Encéfalo/diagnóstico por imagem , Carbazóis , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/diagnóstico por imagem , Radioisótopos de Flúor , Tomografia por Emissão de Pósitrons/tendências , Animais , Encéfalo/metabolismo , Carbazóis/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Feminino , Radioisótopos de Flúor/metabolismo , Inflamação/diagnóstico por imagem , Inflamação/metabolismo , Estudos Longitudinais , Valor Preditivo dos Testes , Ratos , Ratos Sprague-Dawley
5.
ACS Chem Neurosci ; 7(11): 1585-1594, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27609046

RESUMO

Regulatory RNAs play a key role in the regulation of protein expression patterns in neurological diseases. Here we studied the regulation of miRNAs in a chronic rat model of temporal lobe epilepsy. The analysis was focused on a putative link with pharmacoresponsiveness as well as the functional implications of the regulation of a selected miRNA. The findings did not reveal a difference in hippocampal miRNA expression between phenobarbital responders and nonresponders. However, when comparing rats following status epilepticus with control rats we identified 13 differentially expressed miRNAs with miRNA-187-3p being most strongly regulated. mRNAs encoding KCNK10/TREK-2 as well as DYRK2 were confirmed as targets of miRNA-187-3p. Expression of the potassium channel protein KCNK10/TREK-2 negatively correlated with hippocampal miRNA-187-3p expression and proved to be upregulated in the chronic phase of the epilepsy model. In conclusion, our data do not suggest a relevant impact of miRNA expression patterns on pharmacoresponsiveness. However, we confirmed regulation of miRNA-187-3p and demonstrated that it impacts the expression of the two-pore domain potassium channel protein KCNK10/TREK-2. Considering evidence from brain ischemia models, KCNK10/TREK-2 upregulation might serve a protective function with a beneficial impact on astrocytic potassium and glutamate homeostasis.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Hipocampo/metabolismo , MicroRNAs/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Anticonvulsivantes/farmacologia , Modelos Animais de Doenças , Epilepsia Resistente a Medicamentos/tratamento farmacológico , Epilepsia Resistente a Medicamentos/metabolismo , Estimulação Elétrica , Epilepsia do Lobo Temporal/tratamento farmacológico , Feminino , Expressão Gênica , Células Hep G2 , Hipocampo/efeitos dos fármacos , Humanos , Neuroestimuladores Implantáveis , MicroRNAs/genética , Mutação , Fenobarbital/farmacologia , Canais de Potássio de Domínios Poros em Tandem/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Ratos Sprague-Dawley , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo , Quinases Dyrk
6.
Epilepsia ; 57(4): 638-47, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26854031

RESUMO

OBJECTIVE: Anticonvulsive monotherapy fails to be effective in one third of patients with epilepsy resulting in the need for polytherapy regimens. However, with the still limited knowledge, drug choices for polytherapy remain empirical. Here we report experimental data from a chronic epilepsy model for the combination of perampanel and zonisamide, which can render guidance for clinical studies and individual drug choices. METHODS: The anticonvulsant effects of the combination of perampanel and zonisamide were evaluated in a rat amygdala kindling model. Furthermore, the potential for motor impairment was evaluated. The type of interaction was quantitatively assessed based on isobolographic analysis. RESULTS: When administered alone, zonisamide dose-dependently increased the afterdischarge threshold in fully kindled rats. Moreover, data confirmed efficacy of perampanel to inhibit seizure initiation and progression with an impact on propagation of activity from the focus. Pronounced threshold increases were observed following administration of a constant zonisamide dosage combined with different doses of perampanel. Isobolographic analysis of drug responses, which is based on individual drug dose-effect data, revealed a synergistic interaction substantiating the high efficacy of the combination. Furthermore, rotarod data indicated that the combination has a favorable tolerability profile when zonisamide is coadministered with low doses of perampanel. Plasma concentration analysis argued against a pharmacokinetic interaction as a basis for the synergism. SIGNIFICANCE: The findings clearly indicate a pronounced synergistic anticonvulsant effect for the combination of the noncompetitive α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist perampanel with zonisamide, which modulates voltage-sensitive sodium channels and T-type calcium currents. Consequently, polytherapy using these two antiepileptic drugs might be efficacious for clinical management of partial-onset seizures. The findings indicate that the impact of dose ratios on tolerability needs be taken into account. With regard to conclusions about the extent of the synergism and its implications further antiepileptic drug combinations need to be evaluated allowing direct comparison.


Assuntos
Anticonvulsivantes/administração & dosagem , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/tratamento farmacológico , Isoxazóis/administração & dosagem , Excitação Neurológica/efeitos dos fármacos , Piridonas/administração & dosagem , Animais , Anticonvulsivantes/sangue , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Epilepsia do Lobo Temporal/sangue , Feminino , Isoxazóis/sangue , Excitação Neurológica/metabolismo , Nitrilas , Piridonas/sangue , Ratos , Ratos Sprague-Dawley , Zonisamida
7.
Eur J Pharmacol ; 771: 29-39, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26681545

RESUMO

The tetracycline antibiotic minocycline can exert strong anti-inflammatory, antioxidant, and antiapoptotic effects. There is cumulating evidence that epileptogenic brain insults trigger neuroinflammation and anti-inflammatory concepts can modulate the process of epileptogenesis. Based on the mechanisms of action discussed for minocycline, the compound is of interest for intervention studies as it can prevent the polarization of microglia into a pro-inflammatory state. Here, we assessed the efficacy of sub-chronic minocycline administration initiated immediately following an electrically-induced status epilepticus in rats. The treatment did not affect the development of spontaneous seizures. However, minocycline attenuated behavioral long-term consequences of status epilepticus with a reduction in hyperactivity and hyperlocomotion. Furthermore, the compound limited the spatial learning deficits observed in the post-status epilepticus model. The typical status epilepticus-induced neuronal cell loss was evident in the hippocampus and the piriform cortex. Minocycline exposure selectively protected neurons in the piriform cortex and the hilus, but not in the hippocampal pyramidal layer. In conclusion, the data argue against an antiepileptogenic effect of minocycline in adult rats. However, the findings suggest a disease-modifying impact of the tetracycline affecting the development of behavioral co-morbidities, as well as long-term consequences on spatial learning. In addition, minocycline administration resulted in a selective neuroprotective effect. Although strong anti-inflammatory effects have been proposed for minocycline, we could not verify these effects in our experimental model. Considering the multitude of mechanisms claimed to contribute to minocycline's effects, it is of interest to further explore the exact mechanisms underlying the beneficial effects in future studies.


Assuntos
Anticonvulsivantes/uso terapêutico , Minociclina/uso terapêutico , Estado Epiléptico/tratamento farmacológico , Animais , Comportamento Animal/efeitos dos fármacos , Córtex Cerebral/patologia , Estimulação Elétrica , Feminino , Hipocampo/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Microglia/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico
8.
Brain Behav Immun ; 53: 138-158, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26685804

RESUMO

Detailed knowledge about the patterns of molecular alterations during epileptogenesis is a presupposition for identifying targets for preventive or disease-modifying approaches, as well as biomarkers of the disease. Large-scale differential proteome analysis can provide unique and novel perspectives based on comprehensive data sets informing about the complex regulation patterns in the disease proteome. Thus, we have completed an elaborate differential proteome analysis based on label-free LC-MS/MS in a rat model of epileptogenesis. Hippocampus and parahippocampal cortex tissues were sampled and analyzed separately at three key time points chosen for monitoring disease development following electrically-induced status epilepticus, namely, the early post-insult phase, the latency phase, and the chronic phase with spontaneous recurrent seizures. We focused the bioinformatics analysis on proteins linked to immune and inflammatory responses, because of the emerging evidence of the specific pathogenic role of inflammatory signalings during epileptogenesis. In the early post-insult and the latency phases, pathway enrichment analysis revealed an extensive over-representation of Toll-like receptor signaling, pro-inflammatory cytokines, heat shock protein regulation, and transforming growth factor beta signaling and leukocyte transendothelial migration. The inflammatory response in the chronic phase proved to be more moderate with differential expression in the parahippocampal cortex exceeding that in the hippocampus. The data sets provide novel information about numerous differentially expressed proteins, which serve as interaction partners or modulators in key disease-associated inflammatory signaling events. Noteworthy, a set of proteins which act as modulators of the ictogenic Toll-like receptor signaling proved to be differentially expressed. In addition, we report novel data demonstrating the regulation of different Toll-like receptor ligands during epileptogenesis. Taken together, the findings deepen our understanding of modulation of inflammatory signaling during epileptogenesis providing an excellent and comprehensive basis for the identification of target and biomarker candidates.


Assuntos
Epilepsia/metabolismo , Inflamação/metabolismo , Animais , Biomarcadores/metabolismo , Córtex Cerebral/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Epilepsia/etiologia , Epilepsia/genética , Feminino , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Inflamação/genética , Giro Para-Hipocampal/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos/metabolismo , Transdução de Sinais , Espectrometria de Massas em Tandem/métodos , Receptores Toll-Like/metabolismo
9.
Neurosci Lett ; 604: 151-6, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26259695

RESUMO

The neuroprotective and anti-inflammatory effects of the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone are of particular interest for disease-modifying and antiepileptogenic approaches. We studied the expression of PPARγ and the impact of rosiglitazone on the consequences of status epilepticus (SE) in a rat post-SE model. Immunohistochemical analysis revealed a selective overexpression of PPARγ in the piriform cortex of rats with spontaneous seizures. Rosiglitazone administration initiated following SE failed to exert relevant effects on the development of spontaneous seizures and neuronal cell loss. Whereas spatial learning in the Morris water maze was delayed in SE animals with vehicle administration, the learning curve of rosiglitazone-treated SE rats showed no significant difference to that of controls. The study provides first evidence arguing against a robust antiepileptogenic effect. However, the findings in the spatial learning paradigm indicate disease-modifying effects.


Assuntos
PPAR gama/metabolismo , Estado Epiléptico/metabolismo , Animais , Comportamento Animal , Estimulação Elétrica , Feminino , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Aprendizagem em Labirinto , Neurônios/efeitos dos fármacos , Neurônios/patologia , PPAR gama/agonistas , Lobo Parietal/efeitos dos fármacos , Lobo Parietal/patologia , Córtex Piriforme/efeitos dos fármacos , Córtex Piriforme/metabolismo , Córtex Piriforme/patologia , Ratos Sprague-Dawley , Rosiglitazona , Convulsões/fisiopatologia , Aprendizagem Espacial , Estado Epiléptico/patologia , Estado Epiléptico/fisiopatologia , Tiazolidinedionas/farmacologia
10.
Eur J Pharmacol ; 740: 72-80, 2014 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-25016931

RESUMO

Blockade of KCa3.1 channels has been suggested as a novel strategy to reduce microglia activation. The concept has been confirmed by neuroprotective effects in a rat brain ischemia-reperfusion model and reduced microglia activation surrounding glioblastomas. Cumulating evidence exists that microglia activation significantly contributes to epileptogenesis as well as intrinsic severity in the chronic epileptic brain. Taken together these data raised the question whether the KCa3.1 channel blocker triarylmethane-34 (TRAM-34) might also exert beneficial effects in chronic epilepsy models. In a rat post-status epilepticus model TRAM-34 treatment following the insult did not result in neuroprotective effects. Whereas status epilepticus-associated neurodegeneration remained unaffected in the piriform cortex, loss of pyramidal cells in the hippocampal CA1 and CA3a region and of neuropeptide Y-positive interneurons in the hilus proved to be exacerbated by pharmacological KCa3.1 blockade. The development of spontaneous seizures and of behavioral and cognitive alterations was comparable in animals receiving TRAM-34 treatment or the respective vehicle. The kindling model of temporal lobe epilepsy with a massive stimulation paradigm with frequent seizure elicitation in fully kindled rats was used to assess a putative disease-modifying effect. However, sub-chronic TRAM-34 treatment failed to exert relevant effects on seizure generation and thresholds. In conclusion, the data obtained in two different chronic epilepsy models argue against using KCa3.1 blockers as disease-modifying or antiepileptogenic agents. Exacerbation of neuronal cell loss in TRAM-34 pre-treated epileptic animals rather indicates that translational development of the compound needs to carefully consider the pathophysiological mechanisms associated with different brain insults.


Assuntos
Epilepsia do Lobo Temporal/tratamento farmacológico , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/uso terapêutico , Pirazóis/uso terapêutico , Estado Epiléptico/tratamento farmacológico , Animais , Epilepsia do Lobo Temporal/patologia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Bloqueadores dos Canais de Potássio/farmacologia , Pirazóis/farmacologia , Ratos Sprague-Dawley , Estado Epiléptico/patologia
11.
Neuropharmacology ; 85: 104-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24880085

RESUMO

Neuroinflammation has been suggested as a key determinant of the intrinsic severity of epilepsy. Glial cell activation and associated inflammatory signaling can influence seizure thresholds as well as the pharmacodynamics and pharmacokinetics of antiepileptic drugs. Based on these data, we hypothesized that molecular imaging of microglia activation might serve as a tool to predict drug refractoriness of epilepsy. Brain uptake of (R)-[11C]PK11195, a ligand of the translocator protein 18 kDa and molecular marker of microglia activation, was studied in a chronic model of temporal lobe epilepsy in rats with selection of phenobarbital responders and non-responders. In rats with drug-sensitive epilepsy, (R)-[11C]PK11195 brain uptake values were comparable to those in non-epileptic controls. Analysis in non-responders revealed enhanced brain uptake of up to 39% in different brain regions. The difference might be related to the fact that non-responders exhibited higher baseline seizure frequencies than responders indicating a more pronounced intrinsic disease severity. In hippocampal sections, ED1 immunostaining argued against a general difference in microglia activation between both groups. Our data suggest that TSPO PET imaging might serve as a biomarker for drug resistance in temporal lobe epilepsy. However, it needs to be considered that our findings indicate that the TSPO PET data might merely reflect seizure frequency. Future experimental and clinical studies should further evaluate the validity of TSPO PET data to predict the response to phenobarbital and other antiepileptic drugs in longitudinal studies with scanning before drug exposure and with a focus on the early phase following an epileptogenic brain insult.


Assuntos
Anticonvulsivantes/farmacologia , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Epilepsia do Lobo Temporal/diagnóstico por imagem , Epilepsia do Lobo Temporal/fisiopatologia , Fenobarbital/farmacologia , Animais , Anticonvulsivantes/sangue , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Radioisótopos de Carbono , Proteínas de Transporte/metabolismo , Doença Crônica , Modelos Animais de Doenças , Resistência a Medicamentos/fisiologia , Eletrodos Implantados , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/patologia , Feminino , Isoquinolinas , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neuroimunomodulação/fisiologia , Fenobarbital/sangue , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Estado Epiléptico/diagnóstico por imagem , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/patologia , Estado Epiléptico/fisiopatologia
12.
ACS Chem Neurosci ; 5(3): 185-93, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24456603

RESUMO

The neural cell adhesion molecule peptide mimetic fibroblast growth loop (FGL) proved to exert neuroprotective, neurotrophic, and anti-inflammatory effects in different in vitro and in vivo experiments. Based on this beneficial efficacy profile, it is currently in clinical development for neurodegenerative diseases and brain insults. Here, we addressed the hypothesis that the peptide might affect development of seizures in a kindling paradigm, as well as associated behavioral and cellular alterations. Both doses tested, 2 and 10 mg/kg FGL, significantly reduced the number of stimulations necessary to induce a generalized seizure. FGL did not exert relevant effects on the behavioral patterns of kindled animals. As expected, kindling increased the hippocampal cell proliferation rate. Whereas the low dose of FGL did not affect this kindling-associated alteration, 10 mg/kg FGL proved to attenuate the expansion of the doublecortin-positive cell population. These data suggest that FGL administration might have an impact on disease-associated alterations in the hippocampal neuronal progenitor cell population. In conclusion, the effects of the peptide mimetic FGL in the kindling model do not confirm a disease-modifying effect with a beneficial impact on the development or course of epilepsy. The results obtained with FGL rather raise some concern regarding a putative effect, which might promote the formation of a hyperexcitable network. Future studies are required to further assess the risks in models with development of spontaneous seizures.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Hipocampo/efeitos dos fármacos , Moléculas de Adesão de Célula Nervosa/farmacologia , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Tonsila do Cerebelo/fisiopatologia , Animais , Materiais Biomiméticos/farmacologia , Bromodesoxiuridina , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas do Domínio Duplacortina , Hipocampo/fisiopatologia , Imuno-Histoquímica , Neuroestimuladores Implantáveis , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neuropeptídeos/metabolismo
13.
Neurosci Lett ; 556: 170-5, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24120433

RESUMO

Ciliary neurotrophic growth factor is considered a potential therapeutic agent for central nervous system diseases. We report first in vivo data of the ciliary neurotrophic growth factor peptide mimetic Cintrofin in a rat post-status epilepticus model. Cintrofin prevented long-term alterations in the number of doublecortin-positive neuronal progenitor cells and attenuated the persistence of basal dendrites. In contrast, Cintrofin did neither affect acute status epilepticus-associated alterations in hippocampal cell proliferation and neurogenesis nor reveal any relevant effect on seizure activity. Whereas status epilepticus caused a significant disturbance in spatial learning in reversed peptide-treated rats, the performance of Cintrofin-treated rats did not differ from controls. The study confirms that Cintrofin comprises an active sequence mimicking effects of its parent molecule. While the data argue against an antiepileptogenic effect, they indicate a putative disease-modifying impact of Cintrofin.


Assuntos
Fator Neurotrófico Ciliar/farmacologia , Aprendizagem/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Estado Epiléptico/tratamento farmacológico , Animais , Fator Neurotrófico Ciliar/uso terapêutico , Proteína Duplacortina , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Microglia/patologia , Mimetismo Molecular , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Neurogênese , Neurônios/patologia , Fragmentos de Peptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/fisiopatologia , Estado Epiléptico/psicologia
14.
Nucl Med Biol ; 40(6): 764-75, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23827307

RESUMO

INTRODUCTION: To analyse the impact of both epilepsy and pharmacological modulation of P-glycoprotein on brain uptake and kinetics of positron emission tomography (PET) radiotracers [(11)C]quinidine and [(11)C]laniquidar. METHODS: Metabolism and brain kinetics of both [(11)C]quinidine and [(11)C]laniquidar were assessed in naive rats, electrode-implanted control rats, and rats with spontaneous recurrent seizures. The latter group was further classified according to their response to the antiepileptic drug phenobarbital into "responders" and "non-responders". Additional experiments were performed following pre-treatment with the P-glycoprotein modulator tariquidar. RESULTS: [(11)C]quinidine was metabolized rapidly, whereas [(11)C]laniquidar was more stable. Brain concentrations of both radiotracers remained at relatively low levels at baseline conditions. Tariquidar pre-treatment resulted in significant increases of [(11)C]quinidine and [(11)C]laniquidar brain concentrations. In the epileptic subgroup "non-responders", brain uptake of [(11)C]quinidine in selected brain regions reached higher levels than in electrode-implanted control rats. However, the relative response to tariquidar did not differ between groups with full blockade of P-glycoprotein by 15 mg/kg of tariquidar. For [(11)C]laniquidar differences between epileptic and control animals were only evident at baseline conditions but not after tariquidar pretreatment. CONCLUSIONS: We confirmed that both [(11)C]quinidine and [(11)C]laniquidar are P-glycoprotein substrates. At full P-gp blockade, tariquidar pre-treatment only demonstrated slight differences for [(11)C]quinidine between drug-resistant and drug-sensitive animals.


Assuntos
Benzazepinas , Epilepsia/diagnóstico por imagem , Epilepsia/tratamento farmacológico , Tomografia por Emissão de Pósitrons/métodos , Quinidina , Quinolinas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Benzazepinas/sangue , Benzazepinas/química , Benzazepinas/metabolismo , Radioisótopos de Carbono , Doença Crônica , Modelos Animais de Doenças , Epilepsia/sangue , Epilepsia/metabolismo , Feminino , Regulação da Expressão Gênica , Cinética , Masculino , Fenobarbital/farmacologia , Fenobarbital/uso terapêutico , Quinidina/sangue , Quinidina/química , Quinidina/metabolismo , Quinolinas/sangue , Quinolinas/química , Quinolinas/metabolismo , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Radioquímica , Ratos , Ratos Sprague-Dawley , Recidiva , Resultado do Tratamento
15.
Epilepsia ; 54(7): 1176-85, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23614482

RESUMO

PURPOSE: The antiepileptic drug, lacosamide, exerts its therapeutic activity by enhancing slow inactivation of voltage-gated sodium channels. Because putative preventive or disease-modifying effects of drugs may affect epileptogenesis, intrinsic severity, and comorbidities, it is of particular interest to assess the effect of lacosamide on the development of epilepsy and associated cellular alterations. METHODS: The effect of lacosamide was evaluated in an electrical rat status epilepticus (SE) model with a 24-day treatment phase following induction of SE. The impact of lacosamide on the development of spontaneous seizures based on continuous video-electroencephalography (EEG) monitoring, as well as the impact on neuronal cell loss and alterations in hippocampal neurogenesis, was assessed. KEY FINDINGS: Neither low-dose nor high-dose lacosamide affected the development of spontaneous seizures. A dose-dependent neuroprotective effect of lacosamide with significant reduction of neuronal cell loss was observed in the hippocampal CA1 region, as well as in the piriform cortex. In addition, lacosamide attenuated the impact of SE on the rate of hippocampal cell neurogenesis. Moreover, lacosamide prevented a significant rise in the number of persistent basal dendrites. SIGNIFICANCE: Our data do not support an antiepileptogenic effect of lacosamide. However, because lacosamide reduced SE-associated cellular alterations, it would be of interest to determine whether these effects indicate a putative disease-modifying effect of lacosamide in future studies.


Assuntos
Hipocampo/patologia , Degeneração Neural/prevenção & controle , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/patologia , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas do Domínio Duplacortina , Estimulação Elétrica/efeitos adversos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Proteínas Associadas aos Microtúbulos/metabolismo , Degeneração Neural/etiologia , Degeneração Neural/patologia , Células-Tronco Neurais/efeitos dos fármacos , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/complicações , Estado Epiléptico/etiologia
16.
Epilepsia ; 52(12): 2333-43, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22050420

RESUMO

PURPOSE: The selection of a minimal active sequence of erythropoietin allowed the design of peptide mimetics that exert beneficial effects in the central nervous system but lack an erythropoietic effect. Erythropoietin has been suggested as a promising therapeutic and prophylactic for epilepsies based on its neuroprotective, neuroregenerative, and antiinflammatory potency. Therefore, it is of particular interest to evaluate whether the nonerythropoietic erythropoietin-derived peptide pHBSP can affect epileptogenesis. METHODS: In a post-status epilepticus model in rats, we determined the effects of pHBSP and of recombinant human erythropoietin with short-term administration following status epilepticus. KEY FINDINGS: Both pHBSP and erythropoietin further enhanced the status epilepticus-associated increase in hippocampal cell proliferation. Thereby, pHBSP seemed to promote neuronal differentiation and survival resulting in a significant increase in neurogenesis. Neither pHBSP nor erythropoietin affected the number of animals exhibiting spontaneous recurrent seizures as well as the seizure frequency in the chronic phase. In the Morris water maze, pHBSP attenuated cognitive deficits in epileptic animals. SIGNIFICANCE: In conclusion, the helix B-derived erythropoietin peptide pHBSP can modulate the cellular and cognitive consequences of a status epilepticus. The impact of pHBSP on spatial learning might indicate that the peptide allows beneficial effects on epileptogenesis-associated cognitive deficits. However, it needs to be considered that learning deficits were not abolished by pHBSP and that the effects were not observed consistently until the end of the study. Therefore, adjustment of timing, duration, and dose of peptide administration might be necessary to further evaluate the efficacy of pHBSP.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Eritropoetina/química , Serina Endopeptidases/uso terapêutico , Estado Epiléptico/complicações , Adaptação Fisiológica/efeitos dos fármacos , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Comportamento Exploratório/efeitos dos fármacos , Feminino , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Microglia/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/etiologia , Estado Epiléptico/patologia
17.
Epilepsy Res ; 96(3): 241-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21741213

RESUMO

The design of peptide mimetics offers interesting opportunities to selectively include beneficial and exclude undesirable effects of a parent molecule. Epotris represents a novel erythropoietin mimetic, which lacks an erythropoietic activity. The present study evaluates the potential of this peptide to interfere with the histopathological consequences of electrical-induced status epilepticus in rats. The peptide attenuated status epilepticus-associated expansion of the neuronal progenitor cell population in a significant manner. Moreover, Epotris affected the number of persistent basal dendrites exhibited by neuronal progenitor cells. In contrast, hippocampal cell loss remained unaffected by administration of this peptide mimetic. Status epilepticus resulted in obvious microglial activation in different brain regions involved in seizure generation and spread. Epotris diminished the microglial response caused by prolonged seizure activity in the thalamus but not in other brain regions. The study renders support that the Epotris' sequences from binding site 2 in helix C of Epo play a role in receptor interaction and cytokine function. In addition, the data demonstrate that Epotris can exert limited in vivo effects on the cellular consequences of prolonged seizure activity. When considering further testing it should be taken in mind that Epotris administration only attenuated selected cellular consequences of status epilepticus and did not completely prevent cellular alterations.


Assuntos
Eritropoetina/farmacologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Fragmentos de Peptídeos/farmacologia , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/patologia , Animais , Proteínas do Domínio Duplacortina , Estimulação Elétrica , Eletrodos Implantados , Feminino , Microglia/efeitos dos fármacos , Microglia/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mimetismo Molecular , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...