Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Discov Med ; 36(183): 842-852, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38665032

RESUMO

BACKGROUND: Following traumatic brain injury (TBI), an imbalance arises in the central nervous system within the hippocampus region, resulting in the proliferation of mossy cell fibers, causing abnormal membrane discharge. Moreover, disruptions in cellular neurotransmitter secretion induce post-traumatic epilepsy. Extensive experimental and clinical data indicate that the orexin system plays a regulatory role in the hippocampal central nervous system, but the specific regulatory effects are unclear. Therefore, further experimental evaluation of its relevance is needed. OBJECTIVE: This study aims to investigate the effects of orexin receptor agonists (OXA) on the seizure threshold and intensity in controlled cortical impact (CCI) mice, and to understand the role of the orexin system in post-traumatic epilepsy (PTE). METHODS: Male C57BL/6 mice weighing 18-22 g were randomly divided into three groups: Sham, CCI, and CCI+OXA. The three groups of mice were sequentially constructed with models, implanted with electrodes, and established drug-delivery cannulas. After a 30-day recovery, the Sham and CCI groups were injected with physiological saline through the administration cannulas, while the CCI+OXA group was injected with OXA. Subsequently, all mice underwent electrical stimulation every 30 minutes for a total of 15 times. Epileptic susceptibility, duration, intensity, and cognitive changes were observed. Concurrently, the expression levels and changes of GABAergic neurons in the hippocampus of each group were examined by immunofluorescence. RESULTS: Injecting OXA into hippocampal CA1 reduces the threshold of post-traumatic seizures, prolongs the post-discharge duration, prolongs seizure duration, reduces cognitive ability, and exacerbates the loss of GABAergic neurons in the hippocampal region. CONCLUSIONS: Based on the results, we can find that injecting OXA antagonists into the CA1 region of the hippocampus can treat or prevent the occurrence and progression of post-traumatic epilepsy.


Assuntos
Lesões Encefálicas Traumáticas , Camundongos Endogâmicos C57BL , Orexinas , Animais , Masculino , Camundongos , Orexinas/metabolismo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Receptores de Orexina/metabolismo , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Epilepsia/etiologia , Epilepsia/metabolismo , Convulsões/etiologia , Convulsões/metabolismo
2.
Neurotherapeutics ; 18(3): 1582-1601, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34595732

RESUMO

Traumatic brain injury (TBI) is defined as an alteration in brain function or other evidence of brain pathology caused by an external force. When epilepsy develops following TBI, it is known as post-traumatic epilepsy (PTE). PTE occurs in a subset of patients suffering from different types and severities of TBI, occurs more commonly following severe injury, and greatly impacts the quality of life for patients recovering from TBI. Similar to other types of epilepsy, PTE is often refractory to drug treatment with standard anti-seizure drugs. No therapeutic approaches have proven successful in the clinic to prevent the development of PTE. Therefore, novel treatment strategies are needed to stop the development of PTE and improve the quality of life for patients after TBI. Interestingly, TBI represents an excellent clinical opportunity for intervention to prevent epileptogenesis as typically the time of initiation of epileptogenesis (i.e., TBI) is known, the population of at-risk patients is large, and animal models for preclinical studies of mechanisms and treatment targets are available. If properly identified and treated, there is a true opportunity to prevent epileptogenesis after TBI and stop seizures from ever happening. With that goal in mind, here we review previous attempts to prevent PTE both in animal studies and in humans, we examine how biomarkers could enable better-targeted therapeutics, and we discuss how genetic variation may predispose individuals to PTE. Finally, we highlight exciting new advances in the field that suggest that there may be novel approaches to prevent PTE that should be considered for further clinical development.


Assuntos
Anticonvulsivantes/uso terapêutico , Lesões Encefálicas Traumáticas/terapia , Epilepsia Pós-Traumática/terapia , Terapia Genética/métodos , Variação Genética/genética , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Anticonvulsivantes/farmacologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/metabolismo , Terapia Baseada em Transplante de Células e Tecidos/métodos , Epilepsia Pós-Traumática/genética , Epilepsia Pós-Traumática/metabolismo , Variação Genética/efeitos dos fármacos , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo
3.
Med Sci Monit ; 26: e923919, 2020 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-32687486

RESUMO

BACKGROUND Post-traumatic epilepsy (PTE) is a common type of acquired epilepsies secondary to traumatic brain injury (TBI), accounting for approximately 10-25% of patients. The present study evaluated activity of PP-4-one against mTOR signaling activation in a rat model of FeCl2-induced post-traumatic epilepsy. MATERIAL AND METHODS Epilepsy in rats was induced by injecting 10 µl FeCl2 (concentration 100 mM) at a uniform rate of 1 µl/minute. The iNOS expression was detected using a Leica microscope connected to a digital camera system. Reverse transcription polymerase chain reaction (RT­PCR) was used for determination of NR1 mRNA expression. RESULTS The post-traumatic epilepsy induced neuronal degeneration in the hippocampus and frontal cortex of the rats. Treatment with PP-4-one prevented neuronal degeneration in the hippocampus and frontal cortex in rats with post-traumatic epilepsy. The data revealed markedly higher levels of p-mTOR and p-P70S6K in rat hippocampal tissues after induction of traumatic epilepsy. Treatment of post-traumatic epilepsy rats with PP-4-one significantly suppressed p-mTOR and p-P70S6K expression, and PP-4-one treatment reduced epileptic brain injury in the rats with post-traumatic epilepsy. CONCLUSIONS PP-4-one exhibits an anti-epileptogenic effect in the rat model of PTE by inhibiting behavioral seizures through suppression of iNOS and astrocytic proliferation. Moreover, PP-4-one treatment suppressed NR1 expression and targeted the mTOR pathway in PTE-induced rats. Thus, PP-4-one shows promise as a novel and effective therapeutic agent for treatment of epilepsy induced by PTE.


Assuntos
Epilepsia Pós-Traumática/tratamento farmacológico , Pirazóis/química , Pirazóis/farmacologia , Piridinas/química , Piridinas/farmacologia , Animais , Anticonvulsivantes/farmacologia , Astrócitos/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas , Lesões Encefálicas Traumáticas , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia Pós-Traumática/metabolismo , Lobo Frontal/metabolismo , Hipocampo/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 70-kDa/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Convulsões , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
4.
Epilepsia ; 61(8): 1774-1785, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32592416

RESUMO

OBJECTIVE: Posttraumatic epilepsy (PTE) is defined as recurrent and unprovoked seizures occurring >1 week after traumatic brain injury (TBI). Animal studies of PTE are lengthy and expensive. In this study, we developed a cost-effective PTE animal model using zebrafish to bridge the gap between in vitro studies and low-throughput animal studies. METHODS: We used two different sets of parameters (G1 and G2) to induce closed-head TBI in adult zebrafish using pulsed high-intensity focused ultrasound. Injured fish and naive controls were evaluated for behavioral deficits and spontaneous behavioral seizure activity up to 21 days postinjury (DPI). We also assessed behavioral seizure susceptibility to a subconvulsive dose of pentylenetetrazole (PTZ; 2.5 mmol·L-1 ) and recorded electrophysiological signals to confirm seizure activity up to 40 DPI. In addition, we investigated injury-related changes in the blood-brain barrier and expression levels of various proteins altered in rodent and human TBI. RESULTS: The G2 parameters resulted in a more severe TBI, with a mortality rate of 25%, as well as motor dysfunction and heightened anxiety persisting at 21 DPI. One hundred percent of the G2 group showed spontaneous myocloniclike behavior, and 80% demonstrated tonic-clonic-like behavioral seizures by 21 DPI. Such activities were not detected in the naive group. After the application of 2.5 mmol·L-1 PTZ, 100% of injured zebrafish had cloniclike seizures at 21 DPI, versus 30% of the naive group. We also demonstrated electrographic seizure activity at 40 DPI, which was not detected in the naive controls. Lastly, we observed acute blood-brain barrier dysfunction and increased levels of HMGB1 and ratios of phosphorylated/total Akt and tau through 21 DPI. SIGNIFICANCE: Together, the results indicate that severe TBI in the adult zebrafish leads to similar behavioral and physiological changes to those of more traditional models, including the development of PTE, and suggest this may be a useful model that can accelerate research in TBI/PTE.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Epilepsia Pós-Traumática/fisiopatologia , Peixe-Zebra , Animais , Comportamento Animal , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , Convulsivantes/farmacologia , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/metabolismo , Proteína HMGB1/metabolismo , Pentilenotetrazol/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ondas Ultrassônicas , Proteínas tau/metabolismo
5.
Epilepsia ; 61(7): 1503-1514, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32484924

RESUMO

OBJECTIVE: To determine the role of aquaporin-4 (AQP4) in posttraumatic epileptogenesis using long-term video-electroencephalographic (vEEG) recordings. Here, differences in EEG were analyzed between wild-type (WT) and AQP4 knockout (KO) mice and between mice with and without posttraumatic epilepsy (PTE). METHODS: WT and AQP4 KO mice were subjected to a single controlled cortical impact traumatic brain injury (TBI) in the frontal cortex, and vEEG was recorded in the ipsilateral hippocampus at 14, 30, 60, and 90 days postinjury (dpi). Intrahippocampal electrical stimulation was also used to assess electrographic seizure threshold and electrographic seizure duration (ESD). RESULTS: The mean seizure frequency per day for WT mice was 0.07 ± 0.07, 0.11 ± 0.07, 0.26 ± 0.13, and 0.12 ± 0.10 at 14, 30, 60, and 90 dpi, respectively. The mean seizure frequency per day for AQP4 KO mice was 0.45 ± 0.27, 0.29 ± 0.12, and 0.26 ± 0.19 at 14, 30, and 60 dpi, respectively. The mean seizure duration was 15 ± 2 seconds and 24 ± 3 seconds for WT and AQP4 KO mice, respectively. The percentage of mice that developed PTE were 28% and 37% for WT and AQP4 KO mice, respectively. Power spectral density (PSD) analysis revealed alterations in EEG frequency bands between sham and TBI in both genotypes. Additionally, PSD analysis of spontaneous recurrent seizures revealed alterations in delta power between genotypes. Morlet wavelet analysis detected heterogeneity in EEG seizure subtypes and dynamic EEG power patterns after TBI. Compared with AQP4 KO mice, a significant increase in ESD was observed in WT mice at 14 dpi. SIGNIFICANCE: Posttraumatic seizures (PTSs) may be modulated by the astrocyte water channel AQP4. Absence of AQP4 increases the number of spontaneous seizures, increases seizure duration, and alters EEG power patterns of PTSs.


Assuntos
Aquaporina 4/deficiência , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/fisiopatologia , Epilepsia Pós-Traumática/metabolismo , Epilepsia Pós-Traumática/fisiopatologia , Animais , Eletroencefalografia/métodos , Masculino , Camundongos , Camundongos Knockout , Gravação em Vídeo/métodos
6.
J Neurotrauma ; 37(5): 692-705, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32000582

RESUMO

Targeting neuroinflammation is a novel frontier in the prevention and treatment of epilepsy. A substantial body of evidence supports a key role for neuroinflammation in epileptogenesis, the pathological process that leads to the development and progression of spontaneous recurrent epileptic seizures. It is also well recognized that traumatic brain injury (TBI) induces a vigorous neuroinflammatory response and that a significant proportion of patients with TBI suffer from debilitating post-traumatic epilepsy. The complement system is a potent effector of innate immunity and a significant contributor to secondary tissue damage and to epileptogenesis following central nervous system injury. Several therapeutic agents targeting the complement system are already on the market to treat other central nervous system disorders or are well advanced in their development. The purpose of this review is to summarize findings on complement activation in experimental TBI and epilepsy models, highlighting the potential of drug repurposing in the development of therapeutics to ameliorate post-traumatic epileptogenesis.


Assuntos
Anticonvulsivantes/uso terapêutico , Lesões Encefálicas Traumáticas/complicações , Proteínas do Sistema Complemento/metabolismo , Reposicionamento de Medicamentos , Epilepsia Pós-Traumática/tratamento farmacológico , Anticonvulsivantes/farmacologia , Lesões Encefálicas Traumáticas/metabolismo , Ativação do Complemento/efeitos dos fármacos , Epilepsia Pós-Traumática/metabolismo , Humanos
7.
Neuroscience ; 428: 140-153, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31866558

RESUMO

Posttraumatic epilepsy (PTE) is a long-term negative consequence of traumatic brain injury (TBI) in which recurrent spontaneous seizures occur after the initial head injury. PTE develops over an undefined period during which circuitry reorganization in the brain causes permanent hyperexcitability. The pathophysiology by which trauma leads to spontaneous seizures is unknown and clinically relevant models of PTE are key to understanding the molecular and cellular mechanisms underlying the development of PTE. In the present study, we used the controlled-cortical impact (CCI) injury model of TBI to induce PTE in mice and to characterize changes in aquaporin-4 (AQP4) expression. A moderate-severe TBI was induced in the right frontal cortex and video-electroencephalographic (vEEG) recordings were performed in the ipsilateral hippocampus to monitor for spontaneous seizures at 14, 30, 60, and 90 days post injury (dpi). The percentage of mice that developed PTE were 13%, 20%, 27%, and 14% at 14, 30, 60, and 90 dpi, respectively. We found a significant increase in AQP4 in the ipsilateral frontal cortex and hippocampus of mice that developed PTE compared to those that did not develop PTE. Interestingly, AQP4 was found to be mislocalized away from the perivascular endfeet and towards the neuropil in mice that developed PTE. Here, we report for the first time, AQP4 dysregulation in a model of PTE which may carry significant implications for epileptogenesis after TBI.


Assuntos
Aquaporina 4 , Lesões Encefálicas Traumáticas/fisiopatologia , Epilepsia Pós-Traumática/metabolismo , Convulsões/fisiopatologia , Animais , Aquaporina 4/metabolismo , Aquaporina 4/farmacologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/fisiopatologia , Masculino , Camundongos , Gravação em Vídeo/métodos
8.
Epilepsia ; 60(11): 2151-2162, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31595501

RESUMO

Traumatic brain injury (TBI) affects 2.5 million people annually within the United States alone, with over 300 000 severe injuries resulting in emergency room visits and hospital admissions. Severe TBI can result in long-term disability. Posttraumatic epilepsy (PTE) is one of the most debilitating consequences of TBI, with an estimated incidence that ranges from 2% to 50% based on severity of injury. Conducting studies of PTE poses many challenges, because many subjects with TBI never develop epilepsy, and it can be more than 10 years after TBI before seizures begin. One of the unmet needs in the study of PTE is an accurate biomarker of epileptogenesis, or a panel of biomarkers, which could provide early insights into which TBI patients are most susceptible to PTE, providing an opportunity for prophylactic anticonvulsant therapy and enabling more efficient large-scale PTE studies. Several recent reviews have provided a comprehensive overview of this subject (Neurobiol Dis, 123, 2019, 3; Neurotherapeutics, 11, 2014, 231). In this review, we describe acute and chronic imaging methods that detect biomarkers for PTE and potential mechanisms of epileptogenesis. We also describe shortcomings in current acquisition methods, analysis, and interpretation that limit ongoing investigations that may be mitigated with advancements in imaging techniques and analysis.


Assuntos
Epilepsia Pós-Traumática/diagnóstico por imagem , Epilepsia Pós-Traumática/metabolismo , Imageamento por Ressonância Magnética/métodos , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada por Raios X/métodos , Anticonvulsivantes/uso terapêutico , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Epilepsia Pós-Traumática/tratamento farmacológico , Humanos
9.
JCI Insight ; 52019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31038473

RESUMO

Traumatic brain injury (TBI) causes cortical dysfunction and can lead to post-traumatic epilepsy. Multiple studies demonstrate that GABAergic inhibitory network function is compromised following TBI, which may contribute to hyperexcitability and motor, behavioral, and cognitive deficits. Preserving the function of GABAergic interneurons, therefore, is a rational therapeutic strategy to preserve cortical function after TBI and prevent long-term clinical complications. Here, we explored an approach based on the ketogenic diet, a neuroprotective and anticonvulsant dietary therapy which results in reduced glycolysis and increased ketosis. Utilizing a pharmacologic inhibitor of glycolysis (2-deoxyglucose, or 2-DG), we found that acute in vitro application of 2-DG decreased the excitability of excitatory neurons, but not inhibitory interneurons, in cortical slices from naïve mice. Employing the controlled cortical impact (CCI) model of TBI in mice, we found that in vitro 2-DG treatment rapidly attenuated epileptiform activity seen in acute cortical slices 3 to 5 weeks after TBI. One week of in vivo 2-DG treatment immediately after TBI prevented the development of epileptiform activity, restored excitatory and inhibitory synaptic activity, and attenuated the loss of parvalbumin-expressing inhibitory interneurons. In summary, 2-DG may have therapeutic potential to restore network function following TBI.


Assuntos
Antimetabólitos/farmacologia , Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/efeitos dos fármacos , Excitabilidade Cortical/efeitos dos fármacos , Desoxiglucose/farmacologia , Epilepsia Pós-Traumática/metabolismo , Neurônios GABAérgicos/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Animais , Contusão Encefálica/metabolismo , Córtex Cerebral/metabolismo , Dieta Cetogênica , Modelos Animais de Doenças , Neurônios GABAérgicos/metabolismo , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Camundongos , Inibição Neural/efeitos dos fármacos , Parvalbuminas/metabolismo
10.
Mol Biol Rep ; 46(2): 1757-1773, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30706359

RESUMO

Post-traumatic epilepsy (PTE) is a common long-term risk associated with traumatic brain injury (TBI). PTE rat model, proposed by Willmore et al., is a well known model that mimics human PTE. The present study explored the lipid metabolism in this PTE rat model by using in vitro, high-resolution NMR (nuclear magnetic resonance) spectroscopy and lipid staining based investigations. The level of gene expression, cytokines and enzyme activity was estimated. Level of TG (triglycerides), PL (phospholipids) and CHOL (cholesterol) was found to increase in brain tissue of PTE rats. This is an indication of the altered lipid metabolism in PTE rats. Level of lipid peroxidation and cytokines was enhanced in the brain tissue of PTE rats. A positive correlation was also observed in cytokines vs. lipid peroxidation. These results make available the evidence of the oxidative stress induced damage or destruction of the lipid components and also the cause of the inflammatory events in PTE rats. Antioxidant enzyme activity and respective gene expression were found to increase in brain tissue of PTE rats. A positive correlation was also observed in antioxidant enzyme's activity vs. respective enzyme gene expression and lipid peroxidation vs. activity of antioxidant enzymes. Such outcomes reflect the oxidative stress induced lipid damage responsible for production enhancement of antioxidant enzymes, which further responsible for enhancing the activity of antioxidant enzymes. A positive correlation was observed in lipid peroxidation vs. lipid components (TG, PL and CHOL) and provides the confirmatory verification of alteration in the level of lipid components. A negative correlation was observed in the level of cytokines and the quantity of TG. This showed that TG is consumed in the production of cytokines. MUA (Motor unit activity) is highly correlated with the level of LP and indicated that oxidative stress is responsible for the event of epileptogenesis. Positive correlation of MUA with RA (rearing activity) and MWM (Morris-water maze) showed that epileptogenesis also influences the memory of PTE rats. Overall results based analyses clearly indicate that the inflammatory activity and oxidative stress in brain tissue of PTE rats, which are responsible to establish a significant change in the lipid metabolism. This can be visualized through a well constructed possible pathway of altered lipid metabolism. This study will improve our understanding and approach in the field of epilepsy that need to be considered for the development of new drugs or therapy for patients with PTE. Representation of the proposed pathway of altered lipid metabolism in posttraumatic epileptic rats.


Assuntos
Epilepsia Pós-Traumática/metabolismo , Metabolismo dos Lipídeos/genética , Animais , Antioxidantes/metabolismo , Antioxidantes/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Colesterol/metabolismo , Modelos Animais de Doenças , Epilepsia Pós-Traumática/patologia , Metabolismo dos Lipídeos/fisiologia , Peroxidação de Lipídeos/fisiologia , Lipídeos/fisiologia , Masculino , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar , Triglicerídeos/metabolismo
11.
Epilepsy Res ; 151: 7-16, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30711714

RESUMO

Multi-center preclinical studies can facilitate the discovery of biomarkers of antiepileptogenesis and thus facilitate the diagnosis and treatment development of patients at risk of developing post-traumatic epilepsy. However, these studies are often limited by the difficulty in harmonizing experimental protocols between laboratories. Here, we assess whether the production of traumatic brain injury (TBI) using the lateral fluid-percussion injury (FPI) in adult male Sprague-Dawley rats (12 weeks at the time of injury) was harmonized between three laboratories - located in the University of Eastern Finland (UEF), Monash University in Melbourne, Australia (Melbourne) and The University of California, Los Angeles, USA (UCLA). These laboratories are part of the international multicenter-based project, the Epilepsy Bioinformatics Study for Antiepileptogenesis Therapy (EpiBioS4Rx). Lateral FPI was induced in adult male Sprague-Dawley rats. The success of methodological harmonization was assessed by performing inter-site comparison of injury parameters including duration of anesthesia during surgery, impact pressure, post-impact transient apnea, post-impact seizure-like behavior, acute mortality (<72 h post-injury), time to self-right after the impact, and severity of the injury (assessed with the neuroscore). The data was collected using Common Data Elements and Case Report Forms. The acute mortality was 15% (UEF), 50% (Melbourne) and 57% (UCLA) (p < 0.001). The sites differed in the duration of anesthesia, the shortest being at UEF < Melbourne < UCLA (p < 0.001). The impact pressure used also differed between the sites, the highest being in UEF > Melbourne > UCLA (p < 0.001). The impact pressure associated with the severity of the functional deficits (low neuroscore) (P < 0.05) only at UEF, but not at any of the other sites. Additionally, the sites differed in the duration of post-impact transient apnea (p < 0.001) and time to self-right (P < 0.001), the highest values in both parameters was registered in Melbourne. Post-impact seizure-like behavior was observed in 51% (UEF), 25% (Melbourne) and 2% (UCLA) of rats (p < 0.001). Despite the differences in means when all sites were compared there was significant overlap in injury parameters between the sites. The data reflects the technical difficulties in the production of lateral FPI across multiple sites. On the other hand, the data can be used to model the heterogeneity in human cohorts with closed-head injury. Our animal cohort will provide a good starting point to investigate the factors associated with epileptogenesis after lateral FPI.


Assuntos
Lesões Encefálicas/complicações , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/metabolismo , Cooperação Internacional , Animais , Anticonvulsivantes , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia Pós-Traumática/diagnóstico por imagem , Epilepsia Pós-Traumática/tratamento farmacológico , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas
12.
Neurobiol Dis ; 123: 20-26, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30030025

RESUMO

Traumatic brain injury (TBI) accounts for approximately 16% of acute symptomatic seizures which usually occur in the first week after trauma. Children are at higher risk for post-traumatic seizures than adults. Post-traumatic seizures are a risk factor for delayed development of epilepsy. Delayed, chronic post-traumatic epilepsy is preceded by a silent period during which therapeutic interventions may arrest, revert or prevent epileptogenesis. A number of recent review articles summarize the most important features of post-traumatic seizures and epilepsy; this review will instead focus on the link between cerebrovascular permeability, epileptogenesis and ictal events after TBI. The possibility of acting on the blood-brain barrier (BBB) and the neurovascular unit to prevent, disrupt or treat post-traumatic epilepsy is also discussed. Finally, we describe the latest quest for biomarkers of epileptogenesis which may allow for a more targeted intervention.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Lesões Encefálicas Traumáticas/metabolismo , Epilepsia Pós-Traumática/metabolismo , Animais , Biomarcadores , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/diagnóstico , Permeabilidade Capilar , Epilepsia Pós-Traumática/diagnóstico , Epilepsia Pós-Traumática/etiologia , Humanos
13.
Neurobiol Dis ; 123: 86-99, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29936231

RESUMO

Post-traumatic epilepsy (PTE) is diagnosed in 20% of individuals with acquired epilepsy, and can impact significantly the quality of life due to the seizures and other functional or cognitive and behavioral outcomes of the traumatic brain injury (TBI) and PTE. There is no available antiepileptogenic or disease modifying treatment for PTE. Animal models of TBI and PTE have been developed, offering useful insights on the value of inflammatory, neurodegenerative pathways, hemorrhages and iron accumulation, calcium channels and other target pathways that could be used for treatment development. Most of the existing preclinical studies test efficacy towards pathologies of functional recovery after TBI, while a few studies are emerging testing the effects towards induced or spontaneous seizures. Here we review the existing preclinical trials testing new candidate treatments for TBI sequelae and PTE, and discuss future directions for efforts aiming at developing antiepileptogenic and disease-modifying treatments.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Epilepsia Pós-Traumática/terapia , Animais , Anticonvulsivantes/uso terapêutico , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Encefalite/etiologia , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/metabolismo , Humanos , Transdução de Sinais
14.
Epilepsy Res ; 149: 92-101, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30553097

RESUMO

The Epilepsy Bioinformatics Study for Antiepileptogenic Therapy (EpiBioS4Rx) is an international, multicenter, multidisciplinary study aimed at preventing epileptogenesis (EpiBioS4Rx: https://epibios.loni.usc.edu/). One of the study's major objectives is the discovery of diagnostic, prognostic, and predictive plasma protein and microRNA (miRNA) biomarkers that are sensitive, specific, and translatable to the human condition. Epilepsy due to structural brain abnormalities, secondary to neurological insults such as traumatic brain injury (TBI), currently represents ∼50% of all epilepsy cases. In the preclinical EpiBioS4Rx study, TBI was induced in adult male Sprague Dawley rats using a standardized protocol for lateral fluid-percussion injury. Whole blood was collected from the tail vein at baseline and 2, 9 and 30 days post-injury and processed for plasma separation. Biomaterial properties, sample preparation and integrity, and choice of analysis platform can significantly impact measured marker levels and, in turn, interpretation with respect to injury and/or other variables. We present here the results of procedural harmonization for the first 320 rats included in the EpiBioS4Rx study study, from three international research centers, and preliminary proteomic and miRNA analyses. We also discuss experimental considerations for establishing rigorous quality controls with the goal of harmonizing operating procedures across study sites, and delivering high-quality specimens for preclinical biomarker discovery in a rat model of post-traumatic epilepsy (PTE).


Assuntos
Proteínas Sanguíneas/metabolismo , Epilepsia Pós-Traumática/metabolismo , Homeostase/fisiologia , MicroRNAs/metabolismo , Animais , Biomarcadores/metabolismo , Biologia Computacional , Modelos Animais de Doenças , Hemoglobinas/metabolismo , Cooperação Internacional , MicroRNAs/genética , Proteínas do Tecido Nervoso/metabolismo , Análise Serial de Proteínas , Proteômica , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Fatores de Tempo
15.
Epilepsy Behav ; 87: 188-194, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30146352

RESUMO

This study assessed neuropeptide Y (NPY) expression in the hippocampus after long-term survival following traumatic brain injury (TBI) induced by controlled cortical impact (CCI) with or without the development of posttraumatic epilepsy (PTE). We hypothesized that following long-term survival after CCI, the severity of tissue injury and the development of PTE would correlate with the degree of hippocampal neurodegeneration as reflected by NPY+ and neuronal nuclear antigen (NeuN)+ cell loss. Adult Sprague-Dawley rats of 2-3 months of age were lesioned in the right parietal cortex and monitored for seizure activity by video and/or video-EEG. Neuropeptide Y and NeuN immunoreactivities (IRs) were quantified by light microscopy and semiautomatic image analysis approaches for unbiased quantification. Severely injured animals, marked by extensive tissue loss in the ipsilateral neocortex and adjacent hippocampus, resulted in significantly lower NeuN+ hilar cell density and NPY+ cell loss in the contralateral Cornu Ammonis (CA)-3 and dentate hilus (DH). The degree of NPY+ cell loss was more severe in CCI-injured animals with PTE than those animals that did not develop PTE. Mildly injured animals demonstrated no significant change of NPY expression compared with control animals. Our findings of long-term alterations of NPY expression in the hippocampus of severely brain-injured animals can provide important insights into the cellular and molecular consequences of severe TBI and posttraumatic epileptogenesis.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/lesões , Epilepsia Pós-Traumática/metabolismo , Hipocampo/metabolismo , Neuropeptídeo Y/biossíntese , Animais , Lesões Encefálicas Traumáticas/fisiopatologia , Eletroencefalografia/métodos , Epilepsia Pós-Traumática/fisiopatologia , Expressão Gênica , Hipocampo/fisiopatologia , Masculino , Neurônios/metabolismo , Neuropeptídeo Y/genética , Ratos , Ratos Sprague-Dawley
16.
Epilepsy Res ; 128: 35-42, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27810514

RESUMO

Post-traumatic epilepsy (PTE) is one of the most common complications resulting from brain injury, however, antiepileptic drugs usually fail to prevent it. Several lines of evidence have demonstrated that the endogenous cannabinoid system (ECS) plays a pivotal role during epileptogenesis in several animal models. A recent study has shown that a cannabinoid type 1 (CB1) receptor antagonist could suppress long-term neuron hyperexcitability after brain injury, but the underlying mechanisms remain largely unknown. In this study, we first analyzed the dynamic expression of different components of the ECS at various time points after brain injury in rats. Then, we conducted a 12-month-long session of behavioral monitoring after the brain injury, and based on the results, the rats were divided into a PTE group and a non-PTE group. Finally, the changes in the ECS between the two groups were compared. We found that the ECS exhibited a biphasic alteration after brain injury; the expression of the CB1 receptor and 2-arachidonoylglycerol (2-AG) in the PTE group was significantly higher than that of the non-PTE group 12 months after traumatic brain injury. Our preliminary results indicated that the ECS might be involved in post-traumatic epileptogenesis.


Assuntos
Lesões Encefálicas/complicações , Lesões Encefálicas/metabolismo , Endocanabinoides/metabolismo , Epilepsia Pós-Traumática/metabolismo , Animais , Ácidos Araquidônicos/metabolismo , Western Blotting , Modelos Animais de Doenças , Progressão da Doença , Eletrocorticografia , Epilepsia Pós-Traumática/etiologia , Expressão Gênica/fisiologia , Glicerídeos/metabolismo , Hipocampo/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptor CB1 de Canabinoide/metabolismo
17.
Rev Neurosci ; 27(3): 329-46, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581067

RESUMO

Traumatic brain injury (TBI) is a complex neurotrauma in civilian life and the battlefield with a broad spectrum of symptoms, long-term neuropsychological disability, as well as mortality worldwide. Posttraumatic epilepsy (PTE) is a common outcome of TBI with unknown mechanisms, followed by posttraumatic epileptogenesis. There are numerous rodent models of TBI available with varying pathomechanisms of head injury similar to human TBI, but there is no evidence for an adequate TBI model that can properly mimic all aspects of clinical TBI and the first successive spontaneous focal seizures follow a single episode of neurotrauma with respect to epileptogenesis. This review aims to provide current information regarding the various experimental animal models of TBI relevant to clinical TBI. Mossy fiber sprouting, loss of dentate hilar neurons along with recurrent seizures, and epileptic discharge similar to human PTE have been studied in fluid percussion injury, weight-drop injury, and cortical impact models, but further refinement of animal models and functional test is warranted to better understand the underlying pathophysiology of posttraumatic epileptogenesis. A multifaceted research approach in TBI model may lead to exploration of the potential treatment measures, which are a major challenge to the research community and drug developers. With respect to clinical setting, proper patient data collection, improved clinical trials with advancement in drug delivery strategies, blood-brain barrier permeability, and proper monitoring of level and effects of target drug are also important.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Epilepsia Pós-Traumática/tratamento farmacológico , Neurônios/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Epilepsia Pós-Traumática/metabolismo , Humanos
18.
PLoS One ; 8(5): e64078, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691153

RESUMO

Posttraumatic epilepsy is a major source of disability following traumatic brain injury (TBI) and a common cause of medically-intractable epilepsy. Previous attempts to prevent the development of posttraumatic epilepsy with treatments administered immediately following TBI have failed. Recently, the mammalian target of rapamycin complex 1 (mTORC1) pathway has been implicated in mechanisms of epileptogenesis and the mTORC1 inhibitor, rapamycin, has been proposed to have antiepileptogenic effects in preventing some types of epilepsy. In this study, we have tested the hypothesis that rapamycin has antiepileptogenic actions in preventing the development of posttraumatic epilepsy in an animal model of TBI. A detailed characterization of posttraumatic epilepsy in the mouse controlled cortical impact model was first performed using continuous video-EEG monitoring for 16 weeks following TBI. Controlled cortical impact injury caused immediate hyperactivation of the mTORC1 pathway lasting at least one week, which was reversed by rapamycin treatment. Rapamycin decreased neuronal degeneration and mossy fiber sprouting, although the effect on mossy fiber sprouting was reversible after stopping rapamycin and did not directly correlate with inhibition of epileptogenesis. Most posttraumatic seizures occurred greater than 10 weeks after TBI, and rapamycin treatment for one month after TBI decreased the seizure frequency and rate of developing posttraumatic epilepsy during the entire 16 week monitoring session. These results suggest that rapamycin may represent a rational treatment for preventing posttraumatic epilepsy in patients with TBI.


Assuntos
Epilepsia Pós-Traumática/prevenção & controle , Sirolimo/farmacologia , Animais , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia Pós-Traumática/metabolismo , Epilepsia Pós-Traumática/patologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Fibras Musgosas Hipocampais/efeitos dos fármacos , Fibras Musgosas Hipocampais/patologia , Complexos Multiproteicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
19.
Neuroscience ; 210: 451-66, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22433297

RESUMO

Epileptogenesis following traumatic brain injury (TBI) is likely due to a combination of increased excitability, disinhibition, and increased excitatory connectivity via aberrant axon sprouting. Targeting these pathways could be beneficial in the prevention and treatment of posttraumatic epilepsy. Here, we tested this possibility using the novel anticonvulsant (R)-N-benzyl 2-acetamido-3-methoxypropionamide ((R)-lacosamide [LCM]), which acts on both voltage-gated sodium channels and collapsin response mediator protein 2 (CRMP2), an axonal growth/guidance protein. LCM inhibited CRMP2-mediated neurite outgrowth, an effect phenocopied by CRMP2 knockdown. Mutation of LCM-binding sites in CRMP2 reduced the neurite inhibitory effect of LCM by ∼8-fold. LCM also reduced CRMP2-mediated tubulin polymerization. Thus, LCM selectively impairs CRMP2-mediated microtubule polymerization, which underlies its neurite outgrowth and branching. To determine whether LCM inhibits axon sprouting in vivo, LCM was injected into rats subjected to partial cortical isolation, an animal model of posttraumatic epileptogenesis that exhibits axon sprouting in cortical pyramidal neurons. Two weeks following injury, excitatory synaptic connectivity of cortical layer V pyramidal neurons was mapped using patch clamp recordings and laser scanning photostimulation of caged glutamate. In comparison with injured control animals, there was a significant decrease in the map size of excitatory synaptic connectivity in LCM-treated rats, suggesting that LCM treatment prevented enhanced excitatory synaptic connectivity due to posttraumatic axon sprouting. These findings suggest, for the first time, that LCM's mode of action involves interactions with CRMP2 to inhibit posttraumatic axon sprouting.


Assuntos
Anticonvulsivantes/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Acetamidas/farmacologia , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Modelos Animais de Doenças , Epilepsia Pós-Traumática/metabolismo , Epilepsia Pós-Traumática/patologia , Epilepsia Pós-Traumática/fisiopatologia , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intercelular , Lacosamida , Regeneração Nervosa/fisiologia , Neuritos/metabolismo , Técnicas de Patch-Clamp , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
20.
Epilepsy Res ; 95(1-2): 20-34, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21439793

RESUMO

Traumatic brain injury (TBI) can result in the development of posttraumatic epilepsy (PTE). Recently, we reported differential alterations in tonic and phasic GABA(A) receptor (GABA(A)R) currents in hippocampal dentate granule cells 90 days after controlled cortical impact (CCI) (Mtchedlishvili et al., 2010). In the present study, we investigated long-term changes in the protein expression of GABA(A)R α1, α4, γ2, and δ subunits, NMDA (NR2B) and AMPA (GluR1) receptor subunits, and heat shock proteins (HSP70 and HSP90) in the hippocampus of Sprague-Dawley rats evaluated by Western blotting in controls, CCI-injured animals without PTE (CCI group), and CCI-injured animals with PTE (PTE group). No differences were found among all three groups for α1 and α4 subunits. Significant reduction of γ2 protein was observed in the PTE group compared to control. CCI caused a 194% and 127% increase of δ protein in the CCI group compared to control (p<0.0001), and PTE (p<0.0001) groups, respectively. NR2B protein was increased in CCI and PTE groups compared to control (p=0.0001, and p=0.011, respectively). GluR1 protein was significantly decreased in CCI and PTE groups compared to control (p=0.003, and p=0.001, respectively), and in the PTE group compared to the CCI group (p=0.036). HSP70 was increased in CCI and PTE groups compared to control (p=0.014, and p=0.005, respectively); no changes were found in HSP90 expression. These results provide for the first time evidence of long-term alterations of GABA(A) and glutamate receptor subunits and a HSP following CCI.


Assuntos
Lesões Encefálicas/metabolismo , Epilepsia Pós-Traumática/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP90/biossíntese , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Receptores de AMPA/biossíntese , Receptores de GABA-A/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Animais , Lesões Encefálicas/genética , Epilepsia Pós-Traumática/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP90/genética , Hipocampo/lesões , Masculino , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/genética , Receptores de GABA-A/genética , Receptores de N-Metil-D-Aspartato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA