Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Epilepsia ; 61(3): 359-386, 2020 03.
Article in English | MEDLINE | ID: mdl-32196665

ABSTRACT

Prevention of epilepsy is a great unmet need. Acute central nervous system (CNS) insults such as traumatic brain injury (TBI), cerebrovascular accidents (CVA), and CNS infections account for 15%-20% of all epilepsy. Following TBI and CVA, there is a latency of days to years before epilepsy develops. This allows treatment to prevent or modify postinjury epilepsy. No such treatment exists. In animal models of acquired epilepsy, a number of medications in clinical use for diverse indications have been shown to have antiepileptogenic or disease-modifying effects, including medications with excellent side effect profiles. These include atorvastatin, ceftriaxone, losartan, isoflurane, N-acetylcysteine, and the antiseizure medications levetiracetam, brivaracetam, topiramate, gabapentin, pregabalin, vigabatrin, and eslicarbazepine acetate. In addition, there are preclinical antiepileptogenic data for anakinra, rapamycin, fingolimod, and erythropoietin, although these medications have potential for more serious side effects. However, except for vigabatrin, there have been almost no translation studies to prevent or modify epilepsy using these potentially "repurposable" medications. We may be missing an opportunity to develop preventive treatment for epilepsy by not evaluating these medications clinically. One reason for the lack of translation studies is that the preclinical data for most of these medications are disparate in terms of types of injury, models within different injury type, dosing, injury-treatment initiation latencies, treatment duration, and epilepsy outcome evaluation mode and duration. This makes it difficult to compare the relative strength of antiepileptogenic evidence across the molecules, and difficult to determine which drug(s) would be the best to evaluate clinically. Furthermore, most preclinical antiepileptogenic studies lack information needed for translation, such as dose-blood level relationship, brain target engagement, and dose-response, and many use treatment parameters that cannot be applied clinically, for example, treatment initiation before or at the time of injury and dosing higher than tolerated human equivalent dosing. Here, we review animal and human antiepileptogenic evidence for these medications. We highlight the gaps in our knowledge for each molecule that need to be filled in order to consider clinical translation, and we suggest a platform of preclinical antiepileptogenesis evaluation of potentially repurposable molecules or their combinations going forward.


Subject(s)
Anticonvulsants/therapeutic use , Antioxidants/therapeutic use , Epilepsy, Post-Traumatic/prevention & control , Epilepsy/prevention & control , GABA Agents/therapeutic use , Immunologic Factors/therapeutic use , Neuroprotective Agents/therapeutic use , Acetylcysteine/therapeutic use , Animals , Atorvastatin/therapeutic use , Brain Injuries, Traumatic/complications , Ceftriaxone/therapeutic use , Dibenzazepines/therapeutic use , Drug Repositioning , Epilepsy/etiology , Erythropoietin/therapeutic use , Fingolimod Hydrochloride/therapeutic use , Gabapentin/therapeutic use , Humans , Inflammation , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Isoflurane/therapeutic use , Levetiracetam/therapeutic use , Losartan/therapeutic use , Oxidative Stress , Pregabalin/therapeutic use , Pyrrolidinones/therapeutic use , Sirolimus/therapeutic use , Stroke/complications , Topiramate/therapeutic use , Translational Research, Biomedical , Vigabatrin/therapeutic use
2.
Neurotoxicology ; 78: 99-105, 2020 05.
Article in English | MEDLINE | ID: mdl-32084435

ABSTRACT

Organophosphates (OPs) are widely used as pesticides and have been employed as warfare agents. OPs inhibit acetylcholinesterase, leading to over-stimulation of cholinergic synapses and can cause status epilepticus (SE). OPs poisoning can result in irreversible brain damage and death. Despite termination of SE, recurrent seizures and abnormal brain activity remain common sequelae often associated with long-term neural damage and cognitive dysfunction. Therefore, early treatment for prevention of seizures is of high interest. Using a rat model of paraoxon poisoning, we tested the efficacy of different neuroprotective and anti-epileptic drugs (AEDs) in suppressing early seizures and preventing brain damage. Electrocorticographic recordings were performed prior, during and after injection of 4.5 LD50 paraoxon, followed by injections of atropine and toxogonin (obidoxime) to prevent death. Thirty minutes later, rats were injected with midazolam alone or in combination with different AEDs (lorazepam, valproic acid, phenytoin) or neuroprotective drugs (losartan, isoflurane). Outcome measures included SE duration, early seizures frequency and epileptiform activity duration in the first 24 -hs after poisoning. To assess delayed brain damage, we performed T2-weighted magnetic resonance imaging one month after poisoning. SE duration and the number of recurrent seizures were not affected by the addition of any of the drugs tested. Delayed brain injury was most prominent in the septum, striatum, amygdala and piriform network. Only isoflurane anesthesia significantly reduced brain damage. We show that acute treatment with isoflurane, but not AEDs, reduces brain damage following SE. This may offer a new therapeutic approach for exposed individuals.


Subject(s)
Anticonvulsants/administration & dosage , Brain/drug effects , Isoflurane/administration & dosage , Midazolam/administration & dosage , Paraoxon/toxicity , Status Epilepticus/prevention & control , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Male , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/pathology
3.
Sci Transl Med ; 11(521)2019 12 04.
Article in English | MEDLINE | ID: mdl-31801888

ABSTRACT

A growing body of evidence shows that epileptic activity is frequent but often undiagnosed in patients with Alzheimer's disease (AD) and has major therapeutic implications. Here, we analyzed electroencephalogram (EEG) data from patients with AD and found an EEG signature of transient slowing of the cortical network that we termed paroxysmal slow wave events (PSWEs). The occurrence per minute of the PSWEs was correlated with level of cognitive impairment. Interictal (between seizures) PSWEs were also found in patients with epilepsy, localized to cortical regions displaying blood-brain barrier (BBB) dysfunction, and in three rodent models with BBB pathology: aged mice, young 5x familial AD model, and status epilepticus-induced epilepsy in young rats. To investigate the potential causative role of BBB dysfunction in network modifications underlying PSWEs, we infused the serum protein albumin directly into the cerebral ventricles of naïve young rats. Infusion of albumin, but not artificial cerebrospinal fluid control, resulted in high incidence of PSWEs. Our results identify PSWEs as an EEG manifestation of nonconvulsive seizures in patients with AD and suggest BBB pathology as an underlying mechanism and as a promising therapeutic target.


Subject(s)
Alzheimer Disease/physiopathology , Blood-Brain Barrier/physiopathology , Cerebral Cortex/physiopathology , Electroencephalography , Epilepsy/physiopathology , Aged , Aging/pathology , Animals , Dementia/physiopathology , Humans , Male , Mice , Nerve Net/physiopathology , Perfusion , Rats , Serum Albumin/metabolism
4.
Epilepsia ; 60(5): 1005-1016, 2019 05.
Article in English | MEDLINE | ID: mdl-31032909

ABSTRACT

OBJECTIVE: Dogs with spontaneous or acquired epilepsy exhibit resemblance in etiology and disease course to humans, potentially offering a translational model of the human disease. Blood-brain barrier dysfunction (BBBD) has been shown to partake in epileptogenesis in experimental models of epilepsy. To test the hypothesis that BBBD can be detected in dogs with naturally occurring seizures, we developed a linear dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) analysis algorithm that was validated in clinical cases of seizing dogs and experimental epileptic rats. METHODS: Forty-six dogs with naturally occurring seizures of different etiologies and 12 induced epilepsy rats were imaged using DCE-MRI. Six healthy dogs and 12 naive rats served as control. DCE-MRI was analyzed by linear-dynamic method. BBBD scores were calculated in whole brain and in specific brain regions. Immunofluorescence analysis for transforming growth factor beta (TGF-ß) pathway proteins was performed on the piriform cortex of epileptic dogs. RESULTS: We found BBBD in 37% of dogs with seizures. A significantly higher cerebrospinal fluid to serum albumin ratio was found in dogs with BBBD relative to dogs with intact blood-brain barrier (BBB). A significant difference was found between epileptic and control rats when BBBD scores were calculated for the piriform cortex at 48 hours and 1 month after status epilepticus. Mean BBBD score of the piriform lobe in idiopathic epilepsy (IE) dogs was significantly higher compared to control. Immunohistochemistry results suggested active TGF-ß signaling and neuroinflammation in the piriform cortex of dogs with IE, showing increased levels of serum albumin colocalized with glial acidic fibrillary protein and pSMAD2 in an area where BBBD had been detected by linear DCE-MRI. SIGNIFICANCE: Detection of BBBD in dogs with naturally occurring epilepsy provides the ground for future studies for evaluation of novel treatment targeting the disrupted BBB. The involvement of the piriform lobe seen using our linear DCE-MRI protocol and algorithm emphasizes the possibility of using dogs as a translational model for the human disease.


Subject(s)
Blood-Brain Barrier , Dog Diseases/physiopathology , Epilepsy/veterinary , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Albumins/cerebrospinal fluid , Algorithms , Animals , Brain Neoplasms/complications , Brain Neoplasms/physiopathology , Brain Neoplasms/veterinary , Contrast Media , Convulsants/toxicity , Dog Diseases/blood , Dog Diseases/cerebrospinal fluid , Dog Diseases/diagnostic imaging , Dogs , Epilepsy/diagnostic imaging , Epilepsy/metabolism , Epilepsy/physiopathology , Gliosis/etiology , Paraoxon/toxicity , Piriform Cortex/blood supply , Piriform Cortex/diagnostic imaging , Piriform Cortex/metabolism , Piriform Cortex/pathology , Prospective Studies , Rats , Serum Albumin/analysis , Signal Transduction , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Transforming Growth Factor beta/physiology
5.
Epilepsia ; 60(2): 322-336, 2019 02.
Article in English | MEDLINE | ID: mdl-30609012

ABSTRACT

OBJECTIVE: Blood-brain barrier (BBB) impairment, redistribution of pericytes, and disturbances in cerebral blood flow may contribute to the increased seizure propensity and neurological comorbidities associated with epilepsy. However, despite the growing evidence of postictal disturbances in microcirculation, it is not known how recurrent seizures influence pericytic membrane currents and subsequent vasodilation. METHODS: Here, we investigated successive changes in capillary neurovascular coupling and BBB integrity during recurrent seizures induced by 4-aminopyridine or low-Mg2+ conditions. To avoid the influence of arteriolar dilation and cerebral blood flow changes on the capillary response, we measured seizure-associated pericytic membrane currents, capillary motility, and permeability changes in a brain slice preparation. Arteriolar responses to 4-aminopyridine-induced seizures were further studied in anesthetized Sprague Dawley rats by using electrocorticography and tissue oxygen recordings simultaneously with intravital imaging of arteriolar diameter, BBB permeability, and cellular damage. RESULTS: Within the preserved vascular network in hippocampal slice cultures, pericytes regulated capillary diameter in response to vasoactive agents and neuronal activity. Seizures induced distinct patterns of membrane currents that contributed to the regulation of pericytic length. During the course of recurrent seizures, individual vasodilation responses eroded and BBB permeability increased, despite unaltered neurometabolic coupling. Reduced vascular responsiveness was associated with mitochondrial depolarization in pericytes. Subsequent capillary constriction preceded BBB opening, suggesting that pericyte injury mediates the breach in capillary integrity. In vivo findings were consistent with slice experiments, showing seizure-related neurovascular decoupling and BBB dysfunction in small cortical arterioles, accompanied by perivascular cellular injury despite normoxic conditions. SIGNIFICANCE: Our study presents a direct observation of gradually developing neurovascular decoupling during recurrent seizures and suggests pericytic injury as an inducer of vascular dysfunction in epilepsy.


Subject(s)
Blood-Brain Barrier/physiopathology , Capillaries/injuries , Capillary Permeability/physiology , Seizures/physiopathology , Animals , Brain/physiopathology , Capillaries/physiopathology , Cerebrovascular Circulation/physiology , Neurons/physiology , Neurovascular Coupling/physiology , Rats, Sprague-Dawley , Seizures/complications
6.
Brain ; 140(6): 1692-1705, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28444141

ABSTRACT

A biomarker that will enable the identification of patients at high-risk for developing post-injury epilepsy is critically required. Microvascular pathology and related blood-brain barrier dysfunction and neuroinflammation were shown to be associated with epileptogenesis after injury. Here we used prospective, longitudinal magnetic resonance imaging to quantitatively follow blood-brain barrier pathology in rats following status epilepticus, late electrocorticography to identify epileptic animals and post-mortem immunohistochemistry to confirm blood-brain barrier dysfunction and neuroinflammation. Finally, to test the pharmacodynamic relevance of the proposed biomarker, two anti-epileptogenic interventions were used; isoflurane anaesthesia and losartan. Our results show that early blood-brain barrier pathology in the piriform network is a sensitive and specific predictor (area under the curve of 0.96, P < 0.0001) for epilepsy, while diffused pathology is associated with a lower risk. Early treatments with either isoflurane anaesthesia or losartan prevented early microvascular damage and late epilepsy. We suggest quantitative assessment of blood-brain barrier pathology as a clinically relevant predictive, diagnostic and pharmaco!dynamics biomarker for acquired epilepsy.


Subject(s)
Anesthetics, Inhalation/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/physiopathology , Isoflurane/pharmacology , Losartan/pharmacology , Magnetic Resonance Imaging/methods , Status Epilepticus/diagnostic imaging , Status Epilepticus/physiopathology , Anesthesia, Inhalation , Anesthetics, Inhalation/administration & dosage , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Biomarkers , Blood-Brain Barrier/drug effects , Disease Models, Animal , Electrocorticography , Isoflurane/administration & dosage , Losartan/administration & dosage , Male , Prospective Studies , Rats , Rats, Sprague-Dawley , Status Epilepticus/drug therapy
7.
Ann Neurol ; 80(6): 896-908, 2016 12.
Article in English | MEDLINE | ID: mdl-27761920

ABSTRACT

OBJECTIVE: Acquired epilepsy is a devastating long-term risk of various brain insults, including trauma, stroke, infections, and status epilepticus (SE). There is no preventive treatment for patients at risk. Attributable to the complex alterations involved in epileptogenesis, it is likely that multitargeted approaches are required for epilepsy prevention. We report novel preclinical findings with isoflurane, which exerts various nonanesthetic effects that may be relevant for antiepileptogenesis. METHODS: The effects of isoflurane were investigated in two rat models of SE-induced epilepsy: intrahippocampal kainate and systemic administration of paraoxon. Isoflurane was either administered during (kainate) or after (paraoxon) induction of SE. Magnetic resonance imaging was used to assess blood-brain barrier (BBB) dysfunction. Positron emission tomography was used to visualize neuroinflammation. Long-term electrocorticographic recordings were used to monitor spontaneous recurrent seizures. Neuronal damage was assessed histologically. RESULTS: In the absence of isoflurane, spontaneous recurrent seizures were common in the majority of rats in both models. When isoflurane was administered during kainate injection, duration and severity of SE were not affected, but only few rats developed spontaneous recurrent seizures. A similar antiepileptogenic effect was found when paraoxon-treated rats were exposed to isoflurane after SE. Moreover, in the latter model, isoflurane prevented BBB dysfunction and neurodegeneration, whereas isoflurane reduced neuroinflammation in the kainate model. INTERPRETATION: Given that isoflurane is a widely used volatile anesthetic, and is used for inhalational long-term sedation in critically ill patients at risk to develop epilepsy, our findings hold a promising potential to be successfully translated into the clinic. Ann Neurol 2016;80:896-908.


Subject(s)
Epilepsy, Temporal Lobe/prevention & control , Isoflurane/pharmacology , Animals , Blood-Brain Barrier/diagnostic imaging , Disease Models, Animal , Electrocorticography , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Female , Inflammation/diagnostic imaging , Inflammation/prevention & control , Kainic Acid , Magnetic Resonance Imaging , Male , Neuroimaging , Neurons/pathology , Paraoxon , Positron-Emission Tomography , Rats
8.
Semin Cell Dev Biol ; 38: 43-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25455024

ABSTRACT

A dysfunctional BBB is a common feature in a variety of brain disorders, a fact stressing the need for diagnostic tools designed to assess brain vessels' permeability in space and time. Biological research has benefited over the years various means to analyze BBB integrity. The use of biomarkers for improper BBB functionality is abundant. Systemic administration of BBB impermeable tracers can both visualize brain regions characterized by BBB impairment, as well as lead to its quantification. Additionally, locating molecular, physiological content in regions from which it is restricted under normal BBB functionality undoubtedly indicates brain pathology-related BBB disruption. However, in-depth research into the BBB's phenotype demands higher analytical complexity than functional vs. pathological BBB; criteria which biomarker based BBB permeability analyses do not meet. The involvement of accurate and engineering sciences in recent brain research, has led to improvements in the field, in the form of more accurate, sensitive imaging-based methods. Improvements in the spatiotemporal resolution of many imaging modalities and in image processing techniques, make up for the inadequacies of biomarker based analyses. In pre-clinical research, imaging approaches involving invasive procedures, enable microscopic evaluation of BBB integrity, and benefit high levels of sensitivity and accuracy. However, invasive techniques may alter normal physiological function, thus generating a modality-based impact on vessel's permeability, which needs to be corrected for. Non-invasive approaches do not affect proper functionality of the inspected system, but lack in spatiotemporal resolution. Nevertheless, the benefit of medical imaging, even in pre-clinical phases, outweighs its disadvantages. The innovations in pre-clinical imaging and the development of novel processing techniques, have led to their implementation in clinical use as well. Specialized analyses of vessels' permeability add valuable information to standard anatomical inspections which do not take the latter into consideration.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Animals , Blood Vessels/cytology , Blood Vessels/pathology , Blood Vessels/physiology , Blood-Brain Barrier/anatomy & histology , Blood-Brain Barrier/physiology , Brain/cytology , Brain/pathology , Brain/physiology , Capillary Permeability , Humans , Magnetic Resonance Imaging , Radiography
9.
Epilepsia ; 55(12): 1953-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25377630

ABSTRACT

OBJECTIVE: Organophosphates (OPs) are commonly used insecticides for agriculture and domestic purposes, but may also serve as nerve agents. Exposure to OPs result in overstimulation of the cholinergic system and lead to status epilepticus (SE), a life-threatening condition that is often resistant to treatment. SE is associated with significant neuronal damage, neurocognitive dysfunction, and the development of lifelong epilepsy. Therefore, rapid termination of SE and prevention of brain damage is of high interest. Here we tested the efficacy of sec-butyl-propylacetamide (SPD) and two of its individual stereoisomers, (2S,3S)-SPD and (2R,3R)-SPD, in discontinuing OP-induced seizures. SPD is a one carbon homolog of valnoctamide, a central nervous system (CNS)-active constitutional isomer of valproic acid (VPA) corresponding amide valpromide. METHODS: Rats were implanted with epidural telemetric electrodes to allow electrocorticography (ECoG) recording 24 h prior, during and 24 h after poisoning with the OP paraoxon (at a dose equivalent to 1.4 LD50 Median lethal dose). All rats were provided with antidotal treatment of atropine and toxogonin. Epileptic activity was measured using a novel automated system to evaluate the different effects of midazolam, SPD, and its individual stereoisomers in comparison to nontreated controls. RESULTS: Treatment with SPD or its individual stereoisomer (2S,3S)-SPD significantly shorten paraoxon-induced SE and reduced the duration of recorded pathologic activity after SE was terminated. (2S,3S)-SPD was superior to racemic-SPD in diminishing delayed pathologic epileptiform activity within the first 8 h after SE. SIGNIFICANCE: These results suggest SPD as an efficient drug for the rapid termination of SE and pathological epileptiform activity following OP poisoning, a strategy to reduce neuronal dysfunction and the risk for lifelong epilepsy.


Subject(s)
Amides/therapeutic use , Anticonvulsants/therapeutic use , Status Epilepticus/drug therapy , Valproic Acid/analogs & derivatives , Amides/chemistry , Animals , Anticonvulsants/chemistry , Disease Models, Animal , Electroencephalography , Insecticides/toxicity , Male , Paraoxon/toxicity , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Status Epilepticus/chemically induced , Stereoisomerism , Treatment Outcome , Valproic Acid/chemistry , Valproic Acid/therapeutic use
10.
Expert Rev Neurother ; 14(12): 1365-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25346269

ABSTRACT

Brain injury is a major health concern and associated with delayed neurological complications, including post-injury epilepsy, cognitive and emotional disabilities. Currently, there is no strategy to prevent post-injury delayed complications. We recently showed that dysfunction of the blood-brain barrier, often reported in brain injuries, can lead to epilepsy and neurodegeneration via activation of inflammatory TGF-ß signaling in astrocytes. We further showed that the FDA approved angiotensin II type 1 receptor antagonist, losartan, blocks brain TGF-ß signaling and prevents epilepsy in the albumin or blood-brain barrier breakdown models of epileptogenesis. Here we discuss the potential of losartan as an anti-epileptogenic and a neuroprotective drug, the rationale of its use following brain injury and the challenges of designing clinical trials. We highlight the urgent need to develop reliable biomarkers for epileptogenesis (and other complications) after brain injury as a pre-requisite to challenge neuroprotective therapies.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Injuries/drug therapy , Brain/drug effects , Losartan/therapeutic use , Signal Transduction/drug effects , Animals , Brain/physiopathology , Brain Injuries/physiopathology , Humans , Losartan/administration & dosage , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
11.
Toxicology ; 323: 19-25, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-24881594

ABSTRACT

Poisoning with organophosphates (OPs) may induce status epilepticus (SE), leading to severe brain damage. Our objectives were to investigate whether OP-induced SE leads to the emergence of spontaneous recurrent seizures (SRSs), the hallmark of chronic epilepsy, and if so, to assess the efficacy of benzodiazepine therapy following SE onset in preventing the epileptogenesis. We also explored early changes in hippocampal pyramidal cells excitability in this model. Adult rats were poisoned with the paraoxon (450µg/kg) and immediately treated with atropine (3mg/kg) and obidoxime (20mg/kg) to reduce acute mortality due to peripheral acetylcholinesterase inhibition. Electrical brain activity was assessed for two weeks during weeks 4-6 after poisoning using telemetric electrocorticographic intracranial recordings. All OP-poisoned animals developed SE, which could be suppressed by midazolam. Most (88%) rats which were not treated with midazolam developed SRSs, indicating that they have become chronically epileptic. Application of midazolam 1min following SE onset had a significant antiepileptogenic effect (only 11% of the rats became epileptic; p=0.001 compared to non-midazolam-treated rats). Applying midazolam 30min after SE onset did not significantly prevent chronic epilepsy. The electrophysiological properties of CA1 pyramidal cells, assessed electrophysiologically in hippocampal slices, were not altered by OP-induced SE. Thus we show for the first time that a single episode of OP-induced SE in rats leads to the acquisition of chronic epilepsy, and that this epileptogenic outcome can be largely prevented by immediate, but not delayed, administration of midazolam. Extrapolating these results to humans would suggest that midazolam should be provided together with atropine and an oxime in the immediate pharmacological treatment of OP poisoning.


Subject(s)
Antidotes/therapeutic use , Cholinesterase Inhibitors/toxicity , Epilepsy/prevention & control , Midazolam/therapeutic use , Paraoxon/toxicity , Status Epilepticus/chemically induced , Animals , Atropine/therapeutic use , Cholinesterase Reactivators/therapeutic use , Chronic Disease , Epilepsy/chemically induced , Muscarinic Agonists , Obidoxime Chloride/therapeutic use , Pesticides/toxicity , Pilocarpine , Rats , Rats, Sprague-Dawley , Status Epilepticus/physiopathology
12.
Ann Neurol ; 75(6): 864-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659129

ABSTRACT

OBJECTIVE: Acquired epilepsy is frequently associated with structural lesions after trauma, stroke, and infections. Although seizures are often difficult to treat, there is no clinically applicable strategy to prevent the development of epilepsy in patients at risk. We have recently shown that vascular injury is associated with activation of albumin-mediated transforming growth factor ß (TGF-ß) signaling, and followed by local inflammatory response and epileptiform activity ex vivo. Here we investigated albumin-mediated TGF-ß signaling and tested the efficacy of blocking the TGF-ß pathway in preventing epilepsy. METHODS: We addressed the role of TGF-ß signaling in epileptogenesis in 2 different rat models of vascular injury, combining in vitro and in vivo biochemical assays, gene expression, and magnetic resonance and direct optical imaging for blood-brain barrier permeability and vascular reactivity. Long-term electrocorticographic recordings were acquired in freely behaving animals. RESULTS: We demonstrate that serum-derived albumin preferentially induces activation of the activin receptor-like kinase 5 pathway of TGF-ß receptor I in astrocytes. We further show that the angiotensin II type 1 receptor antagonist, losartan, previously identified as a blocker of peripheral TGF-ß signaling, effectively blocks albumin-induced TGF-ß activation in the brain. Most importantly, losartan prevents the development of delayed recurrent spontaneous seizures, an effect that persists weeks after drug withdrawal. INTERPRETATION: TGF-ß signaling, activated in astrocytes by serum-derived albumin, is involved in epileptogenesis. We propose losartan, a drug approved by the US Food and Drug Administration, as an efficient antiepileptogenic therapy for epilepsy associated with vascular injury.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/prevention & control , Losartan/therapeutic use , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Animals , Animals, Newborn , Anticonvulsants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Benzamides/pharmacology , Blood-Brain Barrier/physiology , Cells, Cultured , Cerebral Cortex/cytology , Dioxoles/pharmacology , Disease Models, Animal , Embryo, Mammalian , Endocytosis/drug effects , Epilepsy/chemically induced , Epilepsy/pathology , Epilepsy/physiopathology , Male , Neurons/drug effects , Neurons/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics
13.
J Neurosci ; 29(28): 8927-35, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605630

ABSTRACT

Brain injury may result in the development of epilepsy, one of the most common neurological disorders. We previously demonstrated that albumin is critical in the generation of epilepsy after blood-brain barrier (BBB) compromise. Here, we identify TGF-beta pathway activation as the underlying mechanism. We demonstrate that direct activation of the TGF-beta pathway by TGF-beta1 results in epileptiform activity similar to that after exposure to albumin. Coimmunoprecipitation revealed binding of albumin to TGF-beta receptor II, and Smad2 phosphorylation confirmed downstream activation of this pathway. Transcriptome profiling demonstrated similar expression patterns after BBB breakdown, albumin, and TGF-beta1 exposure, including modulation of genes associated with the TGF-beta pathway, early astrocytic activation, inflammation, and reduced inhibitory transmission. Importantly, TGF-beta pathway blockers suppressed most albumin-induced transcriptional changes and prevented the generation of epileptiform activity. Our present data identifies the TGF-beta pathway as a novel putative epileptogenic signaling cascade and therapeutic target for the prevention of injury-induced epilepsy.


Subject(s)
Brain/metabolism , Epilepsy/etiology , Gene Expression Profiling/methods , Gene Expression/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Action Potentials/physiology , Albumins/metabolism , Animals , Antibodies/pharmacology , Astrocytes/metabolism , Benzamides/pharmacology , Blood-Brain Barrier/drug effects , Brain/physiopathology , Cluster Analysis , Dioxoles/pharmacology , Disease Models, Animal , Electric Stimulation/adverse effects , Epilepsy/complications , Epilepsy/pathology , Gene Expression Regulation , Genome-Wide Association Study/methods , Glutamic Acid/genetics , Glutamic Acid/metabolism , Immunoprecipitation/methods , In Vitro Techniques , Inflammation/etiology , Ion Channels/genetics , Ion Channels/metabolism , Male , Microarray Analysis/methods , Rats , Rats, Wistar , Smad2 Protein/metabolism , Statistics, Nonparametric , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta2/antagonists & inhibitors , Transforming Growth Factor beta2/immunology , Transforming Growth Factor beta2/metabolism , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...