Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Neural Regen Res ; 19(9): 1865-1866, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38227501
2.
J Transl Med ; 21(1): 796, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37940957

ABSTRACT

BACKGROUND: Epilepsy affects over 65 million people worldwide and significantly burdens patients, caregivers, and society. Drug-resistant epilepsy occurs in approximately 30% of patients and growing evidence indicates that oxidative stress contributes to the development of such epilepsies. Activation of the Nrf2 pathway, which is involved in cellular defense, offers a potential strategy for reducing oxidative stress and epilepsy treatment. Dimethyl fumarate (DMF), an Nrf2 activator, exhibits antioxidant and anti-inflammatory effects and is used to treat multiple sclerosis. METHODS: The expression of Nrf2 and its related genes in vehicle or DMF treated rats were determined via RT-PCR and Western blot analysis. Neuronal cell death was evaluated by immunohistochemical staining. The effects of DMF in preventing the onset of epilepsy and modifying the disease were investigated in the kainic acid-induced status epilepticus model of temporal lobe epilepsy in rats. The open field, elevated plus maze and T-Maze spontaneous alteration tests were used for behavioral assessments. RESULTS: We demonstrate that administration of DMF following status epilepticus increased Nrf2 activity, attenuated status epilepticus-induced neuronal cell death, and decreased seizure frequency and the total number of seizures compared to vehicle-treated animals. Moreover, DMF treatment reversed epilepsy-induced behavioral deficits in the treated rats. Moreover, DMF treatment even when initiated well after the diagnosis of epilepsy, reduced symptomatic seizures long after the drug was eliminated from the body. CONCLUSIONS: Taken together, these findings suggest that DMF, through the activation of Nrf2, has the potential to serve as a therapeutic target for preventing epileptogenesis and modifying epilepsy.


Subject(s)
Epilepsy , Status Epilepticus , Humans , Rats , Animals , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , NF-E2-Related Factor 2/metabolism , Drug Repositioning , Epilepsy/drug therapy , Epilepsy/prevention & control , Seizures/drug therapy , Status Epilepticus/complications , Status Epilepticus/drug therapy , Disease Models, Animal
3.
Transl Psychiatry ; 13(1): 273, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37524707

ABSTRACT

The endocannabinoid system (ECS) plays a key modulatory role during synaptic plasticity and homeostatic processes in the brain and has an important role in the neurobiological processes underlying drug addiction. We have previously shown that an elevated ECS response to psychostimulant (cocaine) is involved in regulating the development and expression of cocaine-conditioned reward and sensitization. We therefore hypothesized that drug-induced elevation in endocannabinoids (eCBs) and/or eCB-like molecules (eCB-Ls) may represent a protective mechanism against drug insult, and boosting their levels exogenously may strengthen their neuroprotective effects. Here, we determine the involvement of ECS in alcohol addiction. We first measured the eCBs and eCB-Ls levels in different brain reward system regions following chronic alcohol self-administration using LC-MS. We have found that following chronic intermittent alcohol consumption, N-oleoyl glycine (OlGly) levels were significantly elevated in the prefrontal cortex (PFC), and N-oleoyl alanine (OlAla) was significantly elevated in the PFC, nucleus accumbens (NAc) and ventral tegmental area (VTA) in a region-specific manner. We next tested whether exogenous administration of OlGly or OlAla would attenuate alcohol consumption and preference. We found that systemic administration of OlGly or OlAla (60 mg/kg, intraperitoneal) during intermittent alcohol consumption significantly reduced alcohol intake and preference without affecting the hedonic state. These findings suggest that the ECS negatively regulates alcohol consumption and boosting selective eCBs exogenously has beneficial effects against alcohol consumption and potentially in preventing relapse.


Subject(s)
Cocaine , Glycine , Mice , Animals , Glycine/pharmacology , Glycine/metabolism , Ethanol/metabolism , Brain , Nucleus Accumbens , Reward , Ventral Tegmental Area
4.
Neuropharmacology ; 238: 109670, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37482179

ABSTRACT

Epilepsy affects approximately 1% of the global population, with 30% of patients experiencing uncontrolled seizures despite treatment. Reactive oxygen species (ROS) and oxidative stress have been implicated in the pathogenesis of epilepsy. Sestrins are stress-inducible proteins that regulate the ROS response. In particular, Sestrin 3 (SESN3) has been implicated in ROS accumulation and the regulation of proconvulsant genes. To investigate the role of SESN3 in epilepsy, we studied its involvement in rat models of acute seizures and temporal lobe epilepsy. Our results showed that downregulation of SESN3 reduced the oxidative stress induced by seizure activity in neuronal cultures. After acute seizure activity, SESN3 protein levels temporarily increased as early as 3 h after the seizure, whereas kainic acid-induced status epilepticus led to a significant and persistent increase in SESN3 protein levels in the cortex and hippocampus for up to 2 weeks post-status epilepticus. In the chronic epilepsy phase, when spontaneous seizures emerge, SESN3 protein expression is significantly increased in both regions 6 and 12 weeks after status epilepticus. Interestingly, immunohistochemical staining showed a predominant increase in the oxidative stress marker 8-OHdG in neurons in both regions after an acute seizure, whereas following status epilepticus, the marker was detected in both neurons and astrocytes. Our findings suggest that SESN3 may contribute to the development and establishment of epilepsy, and could be a potential therapeutic target for more effective treatments.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Status Epilepticus , Animals , Rats , Disease Models, Animal , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Hippocampus , Kainic Acid/toxicity , Neurons , Oxidative Stress , Reactive Oxygen Species/metabolism , Seizures/drug therapy , Sestrins/metabolism , Status Epilepticus/drug therapy , Transcription Factors/metabolism
5.
Epilepsia Open ; 8(3): 1169-1174, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37328275

ABSTRACT

Transient receptor potential cation subfamily M7 (TRPM7) channels are ion channels permeable to divalent cations. They are abundantly expressed with particularly high expression in the brain. Previous studies have highlighted the importance of TRPM7 channels in brain diseases such as stroke and traumatic brain injury, yet evidence for a role in seizures and epilepsy is lacking. Here, we show that carvacrol, a food additive that inhibits TRPM7 channels, and waixenicin A, a novel selective and potent TRPM7 inhibitor, completely suppressed seizure-like activity in rodent hippocampal-entorhinal brain slices exposed to pentylenetetrazole or low magnesium. These findings support inhibition of TRPM7 channels as a novel target for antiseizure medications.

6.
Cell Mol Life Sci ; 80(5): 127, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37081190

ABSTRACT

Hyperexcitability is associated with neuronal dysfunction, cellular death, and consequently neurodegeneration. Redox disbalance can contribute to hyperexcitation and increased reactive oxygen species (ROS) levels are observed in various neurological diseases. NOX4 is an NADPH oxidase known to produce ROS and might have a regulating function during oxidative stress. We, therefore, aimed to determine the role of NOX4 on neuronal firing, hyperexcitability, and hyperexcitability-induced changes in neural network function. Using a multidimensional approach of an in vivo model of hyperexcitability, proteomic analysis, and cellular function analysis of ROS, mitochondrial integrity, and calcium levels, we demonstrate that NOX4 is neuroprotective by regulating ROS and calcium homeostasis and thereby preventing hyperexcitability and consequently neuronal death. These results implicate NOX4 as a potential redox regulator that is beneficial in hyperexcitability and thereby might have an important role in neurodegeneration.


Subject(s)
Calcium , Proteomics , Humans , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Oxidative Stress , Reactive Oxygen Species
7.
J Neurochem ; 165(4): 550-562, 2023 05.
Article in English | MEDLINE | ID: mdl-36807051

ABSTRACT

The modulation of the nuclear factor erythroid 2-like 2 (Nrf2) activity has been reported to be implicated in the pathology of various neurological disorders, including epilepsy. Previous studies have demonstrated that Nrf2 is activated in the post-status epilepticus rat model; however, the spatiotemporal as well as cell type-specific expression of Nrf2 following brief epileptic seizures remains unclear. Here, we evaluated how an acute epileptic seizure affected the expression of Nrf2 and its downstream genes in the rats' cortex and the hippocampus up to 1 week following the induced seizure. We found that after a pentylenetetrazol-induced seizure, Nrf2 significantly increased at 24 h at the mRNA level and 3 h at the protein level in the cortex. In the hippocampus, the Nrf2 mRNA level peaked at 3 h after the seizure, and no significant changes were observed in the protein level. Interestingly, the mRNA level of Nrf2 downstream genes peaked at 3-6 h after seizure in both the cortex and the hippocampus. A significant increase in the expression of Nrf2 was observed in the neuronal population of CA1 and CA3 regions of the hippocampus, as well as in the cortex. Moreover, we observed no change in the co-localization of Nrf2 with astrocytes neither in the cortex nor in CA1 and CA3. Our results revealed that following a brief acute epileptic seizure, the expression of Nrf2 and its downstream genes is transiently increased and peaked at early timepoints after the seizure predominantly in the hippocampus, and this expression is restricted to the neuronal population.


Subject(s)
Epilepsy , NF-E2-Related Factor 2 , Rats , Animals , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Astrocytes/metabolism , Epilepsy/metabolism , Seizures/chemically induced , Seizures/metabolism , Neurons/metabolism , Hippocampus/metabolism , Disease Models, Animal
8.
Cell Biosci ; 13(1): 3, 2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36600279

ABSTRACT

BACKGROUND: Drug resistance is a particular problem in patients with temporal lobe epilepsy, where seizures originate mainly from the hippocampus. Many of these epilepsies are acquired conditions following an insult to the brain such as a prolonged seizure. Such conditions are characterized by pathophysiological mechanisms including massive oxidative stress that synergistically mediate the secondary brain damage, contributing to the development of epilepsy. The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) has emerged in recent years as an attractive therapeutic approach targeting to upregulate the antioxidative defenses in the cell, to ameliorate the oxidative stress-induced damage. Thus, it is important to understand the characteristics of Nrf2 activation during epileptogenesis and epilepsy. Here, we studied the temporal, regional, and cell-type specific expression of Nrf2 in the brain, in a rat model of temporal lobe epilepsy. RESULTS: Early after status-epilepticus, Nrf2 is mainly activated in the hippocampus and maintained during the whole period of epileptogenesis. Only transient expression of Nrf2 was observed in the cortex. Nevertheless, the expression of several Nrf2 antioxidant target genes was increased within 24 h after status-epilepticus in both the cortex and the hippocampus. We demonstrated that after status-epilepticus in rats, Nrf2 is predominantly expressed in neurons in the CA1 and CA3 regions of the hippocampus, and only astrocytes in the CA1 increase their Nrf2 expression. CONCLUSIONS: In conclusion, our data identify previously unrecognized spatial and cell-type dependent activation of Nrf2 during epilepsy development, highlighting the need for a time-controlled, and cell-type specific activation of the Nrf2 pathway for mediating anti-oxidant response after brain insult, to modify the development of epilepsy.

9.
Redox Biol ; 58: 102549, 2022 12.
Article in English | MEDLINE | ID: mdl-36459714

ABSTRACT

Recent work by us and others has implicated NADPH oxidase (NOX) enzymes as main producers of reactive oxygen species (ROS) following a brain insult such as status epilepticus, contributing to neuronal damage and development of epilepsy. Although several NOX isoforms have been examined in the context of epilepsy, most attention has focused on NOX2. In this present study, we demonstrate the effect of gp91ds-tat, a specific competitive inhibitor of NOX2, in in vitro epileptiform activity model as well as in temporal lobe epilepsy (TLE) model in rats. We showed that in in vitro seizure model, gp91ds-tat modulated Ca2+ oscillation, prevented epileptiform activity-induced ROS generation, mitochondrial depolarization, and neuronal death. Administration of gp91ds-tat 1 h after kainic acid-induced status epilepticus significantly decreased the expression of NOX2, as well as the overall NOX activity in the cortex and the hippocampus. Finally, we showed that upon continuous intracerebroventricular administration to epileptic rats, gp91ds-tat significantly reduced the seizure frequency and the total number of seizures post-treatment compared to the scrambled peptide-treated animals. The results of the study suggest that NOX2 may have an important effect on modulation of epileptiform activity and has a critical role in mediating seizure-induced NOX activation, ROS generation and oxidative stress in the brain, and thus significantly contributes to development of epilepsy following a brain insult.


Subject(s)
Epilepsy , NADPH Oxidase 2 , Status Epilepticus , Animals , Rats , Epilepsy/drug therapy , NADPH Oxidase 2/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Seizures
10.
Science ; 378(6619): 523-532, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378958

ABSTRACT

Several neurodevelopmental and neuropsychiatric disorders are characterized by intermittent episodes of pathological activity. Although genetic therapies offer the ability to modulate neuronal excitability, a limiting factor is that they do not discriminate between neurons involved in circuit pathologies and "healthy" surrounding or intermingled neurons. We describe a gene therapy strategy that down-regulates the excitability of overactive neurons in closed loop, which we tested in models of epilepsy. We used an immediate early gene promoter to drive the expression of Kv1.1 potassium channels specifically in hyperactive neurons, and only for as long as they exhibit abnormal activity. Neuronal excitability was reduced by seizure-related activity, leading to a persistent antiepileptic effect without interfering with normal behaviors. Activity-dependent gene therapy is a promising on-demand cell-autonomous treatment for brain circuit disorders.


Subject(s)
Epilepsy , Genetic Therapy , Kv1.1 Potassium Channel , Humans , Brain/metabolism , Epilepsy/genetics , Epilepsy/therapy , Kv1.1 Potassium Channel/genetics , Seizures/genetics , Seizures/therapy , Seizures/metabolism , Animals , Mice , Neurons/physiology
11.
Free Radic Biol Med ; 190: 158-168, 2022 09.
Article in English | MEDLINE | ID: mdl-35964838

ABSTRACT

The NADPH Oxidase (NOX) enzymes are key producers of reactive oxygen species (ROS) and consist of seven different isoforms, distributed across the tissues and cell types. The increasing level of ROS induces oxidative stress playing a crucial role in neuronal death and the development of epilepsy. Recently, NOX2 was reported as a primary source of ROS production, activated by NMDA receptor, a crucial marker of epilepsy development. Here, we demonstrate spatial, temporal, and cellular expression of NOX2 and NOX4 complexes in in-vitro and in-vivo seizure models. We showed that the expression of NOX2 and NOX4 was increased in the initial 24 h following a brief seizure induced by pentylenetetrazol. Interestingly, while this elevated level returns to baseline 48 h following seizure in the cortex, in the hippocampus these levels remain elevated up to one week following the seizure. Moreover, we showed that 1- and 2- weeks following status epilepticus (SE), expression of NOX2 and NOX4 remains significantly elevated both in the cortex and the hippocampus. Furthermore, in in-vitro seizure model, NOX2 and NOX4 isoforms were overexpressed in neurons and astrocytes following seizures. These results suggest that NOX2 and NOX4 in the brain have a transient response to seizures, and these responses temporally vary depending on, seizure duration, brain region (cortex or hippocampus), and cell types.


Subject(s)
NADPH Oxidases , Seizures , Animals , NADPH Oxidase 1 , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Protein Isoforms/genetics , Rats , Reactive Oxygen Species/metabolism , Seizures/chemically induced , Seizures/genetics
12.
Antioxidants (Basel) ; 10(11)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34829573

ABSTRACT

Epilepsy is a chronic disease of the brain that affects over 65 million people worldwide. Acquired epilepsy is initiated by neurological insults, such as status epilepticus, which can result in the generation of ROS and induction of oxidative stress. Suppressing oxidative stress by upregulation of the transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2) has been shown to be an effective strategy to increase endogenous antioxidant defences, including in brain diseases, and can ameliorate neuronal damage and seizure occurrence in epilepsy. Here, we aim to test the neuroprotective potential of a naturally occurring Nrf2 activator sulforaphane, in in vitro epileptiform activity model and a temporal lobe epilepsy rat model. Sulforaphane significantly decreased ROS generation during epileptiform activity, restored glutathione levels, and prevented seizure-like activity-induced neuronal cell death. When given to rats after 2 h of kainic acid-induced status epilepticus, sulforaphane significantly increased the expression of Nrf2 and related antioxidant genes, improved oxidative stress markers, and increased the total antioxidant capacity in both the plasma and hippocampus. In addition, sulforaphane significantly decreased status epilepticus-induced neuronal cell death. Our results demonstrate that Nrf2 activation following an insult to the brain exerts a neuroprotective effect by reducing neuronal death, increasing the antioxidant capacity, and thus may also modify epilepsy development.

13.
Antioxidants (Basel) ; 9(10)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066477

ABSTRACT

Oxidative stress (OS) and excessive reactive oxygen species (ROS) production have been implicated in many neurological pathologies, including acute seizures and epilepsy. Seizure-induced damage has been demonstrated both in vitro and in several in vivo seizure and epilepsy models by direct determination of ROS, and by measuring indirect markers of OS. In this manuscript, we review the current reliable methods for quantifying ROS-related and OS-related markers in pre-clinical and clinical epilepsy studies. We first provide pieces of evidence for the involvement of different sources of ROS in epilepsy. We then discuss general methods and assays used for the ROS measurements, mainly superoxide anion, hydrogen peroxide, peroxynitrite, and hydroxyl radical in in vitro and in vivo studies. In addition, we discuss the role of these ROS and markers of oxidative injury in acute seizures and epilepsy pre-clinical studies. The indirect detection of secondary products of ROS such as measurements of DNA damage, lipid peroxidation, and protein oxidation will also be discussed. This review also discusses reliable methods for the assessment of ROS, OS markers, and their by-products in epilepsy clinical studies.

14.
Redox Biol ; 26: 101278, 2019 09.
Article in English | MEDLINE | ID: mdl-31382215

ABSTRACT

Many epilepsies are acquired conditions following an insult to the brain such as a prolonged seizure, traumatic brain injury or stroke. The generation of reactive oxygen species (ROS) and induction of oxidative stress are common sequelae of such brain insults and have been shown to contribute to neuronal death and the development of epilepsy. Here, we show that combination therapy targeting the generation of ROS through NADPH oxidase inhibition and the endogenous antioxidant system through nuclear factor erythroid 2-related factor 2 (Nrf2) activation prevents excessive ROS accumulation, mitochondrial depolarisation and neuronal death during in vitro seizure-like activity. Moreover, this combination therapy prevented the development of spontaneous seizures in 40% of animals following status epilepticus (70% of animals were seizure free after 8 weeks) and modified the severity of epilepsy when given to chronic epileptic animals.


Subject(s)
Antioxidants/pharmacology , Epilepsy/etiology , Leprostatic Agents/pharmacology , Animals , Antioxidants/administration & dosage , Antioxidants/chemistry , Biomarkers , Chronic Disease , Epilepsy/drug therapy , Epilepsy/metabolism , Epilepsy/prevention & control , Kainic Acid/metabolism , Leprostatic Agents/administration & dosage , Leprostatic Agents/chemistry , Male , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism
15.
Brain ; 142(7): e39, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31145451

ABSTRACT

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Subject(s)
Acetylcysteine/pharmacology , Epilepsy/prevention & control , Glutathione/metabolism , Isothiocyanates/pharmacology , Oxidative Stress/drug effects , Animals , Astrocytes/metabolism , Biomarkers/metabolism , Case-Control Studies , Cell Count , Cognitive Dysfunction/complications , Cognitive Dysfunction/prevention & control , Disease Models, Animal , Electric Stimulation , Epilepsy/complications , HMGB1 Protein/blood , Hippocampus/metabolism , Humans , Male , Neurons/metabolism , Neurons/pathology , Rats , Status Epilepticus/complications , Status Epilepticus/metabolism , Status Epilepticus/prevention & control , Sulfoxides
16.
J Neurosci ; 39(16): 3159-3169, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30755487

ABSTRACT

Refractory focal epilepsy is a devastating disease for which there is frequently no effective treatment. Gene therapy represents a promising alternative, but treating epilepsy in this way involves irreversible changes to brain tissue, so vector design must be carefully optimized to guarantee safety without compromising efficacy. We set out to develop an epilepsy gene therapy vector optimized for clinical translation. The gene encoding the voltage-gated potassium channel Kv1.1, KCNA1, was codon optimized for human expression and mutated to accelerate the recovery of the channels from inactivation. For improved safety, this engineered potassium channel (EKC) gene was packaged into a nonintegrating lentiviral vector under the control of a cell type-specific CAMK2A promoter. In a blinded, randomized, placebo-controlled preclinical trial, the EKC lentivector robustly reduced seizure frequency in a male rat model of focal neocortical epilepsy characterized by discrete spontaneous seizures. When packaged into an adeno-associated viral vector (AAV2/9), the EKC gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. This demonstration of efficacy in a clinically relevant setting, combined with the improved safety conferred by cell type-specific expression and integration-deficient delivery, identify EKC gene therapy as being ready for clinical translation in the treatment of refractory focal epilepsy.SIGNIFICANCE STATEMENT Pharmacoresistant epilepsy affects up to 0.3% of the population. Although epilepsy surgery can be effective, it is limited by risks to normal brain function. We have developed a gene therapy that builds on a mechanistic understanding of altered neuronal and circuit excitability in cortical epilepsy. The potassium channel gene KCNA1 was mutated to bypass post-transcriptional editing and was packaged in a nonintegrating lentivector to reduce the risk of insertional mutagenesis. A randomized, blinded preclinical study demonstrated therapeutic effectiveness in a rodent model of focal neocortical epilepsy. Adeno-associated viral delivery of the channel to both hippocampi was also effective in a model of temporal lobe epilepsy. These results support clinical translation to address a major unmet need.


Subject(s)
Brain/metabolism , Epilepsy/therapy , Genetic Therapy , Kv1.1 Potassium Channel/genetics , Seizures/therapy , Animals , Disease Models, Animal , Epilepsy/genetics , Genetic Vectors , Kv1.1 Potassium Channel/metabolism , Male , Rats , Seizures/genetics
17.
Br J Pharmacol ; 175(11): 2097-2115, 2018 06.
Article in English | MEDLINE | ID: mdl-29574880

ABSTRACT

BACKGROUND AND PURPOSE: A non-psychoactive phytocannabinoid, cannabidiol (CBD), shows promising results as an effective potential antiepileptic drug in some forms of refractory epilepsy. To elucidate the mechanisms by which CBD exerts its anti-seizure effects, we investigated its effects at synaptic connections and on the intrinsic membrane properties of hippocampal CA1 pyramidal cells and two major inhibitory interneurons: fast spiking, parvalbumin (PV)-expressing and adapting, cholecystokinin (CCK)-expressing interneurons. We also investigated whether in vivo treatment with CBD altered the fate of CCK and PV interneurons using immunohistochemistry. EXPERIMENTAL APPROACH: Electrophysiological intracellular whole-cell recordings combined with neuroanatomy were performed in acute brain slices of rat temporal lobe epilepsy in in vivo (induced by kainic acid) and in vitro (induced by Mg2+ -free solution) epileptic seizure models. For immunohistochemistry experiments, CBD was administered in vivo (100 mg·kg-1 ) at zero time and 90 min post status epilepticus, induced with kainic acid. KEY RESULTS: Bath application of CBD (10 µM) dampened excitability at unitary synapses between pyramidal cells but enhanced inhibitory synaptic potentials elicited by fast spiking and adapting interneurons at postsynaptic pyramidal cells. Furthermore, CBD restored impaired membrane excitability of PV, CCK and pyramidal cells in a cell type-specific manner. These neuroprotective effects of CBD were corroborated by immunohistochemistry experiments that revealed a significant reduction in atrophy and death of PV- and CCK-expressing interneurons after CBD treatment. CONCLUSIONS AND IMPLICATIONS: Our data suggest that CBD restores excitability and morphological impairments in epileptic models to pre-epilepsy control levels through multiple mechanisms to reinstate normal network function.


Subject(s)
Anticonvulsants/pharmacology , Cannabidiol/pharmacology , Disease Models, Animal , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/cytology , Hippocampus/drug effects , Interneurons/drug effects , Administration, Oral , Animals , Anticonvulsants/administration & dosage , Cannabidiol/administration & dosage , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Interneurons/physiology , Kainic Acid/metabolism , Male , Rats , Rats, Sprague-Dawley
18.
Brain ; 141(5): 1390-1403, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29538645

ABSTRACT

Hippocampal sclerosis is a common acquired disease that is a major cause of drug-resistant epilepsy. A mechanism that has been proposed to lead from brain insult to hippocampal sclerosis is the excessive generation of reactive oxygen species, and consequent mitochondrial failure. Here we use a novel strategy to increase endogenous antioxidant defences using RTA 408, which we show activates nuclear factor erythroid 2-related factor 2 (Nrf2, encoded by NFE2L2) through inhibition of kelch like ECH associated protein 1 (KEAP1) through its primary sensor C151. Activation of Nrf2 with RTA 408 inhibited reactive oxygen species production, mitochondrial depolarization and cell death in an in vitro model of seizure-like activity. RTA 408 given after status epilepticus in vivo increased ATP, prevented neuronal death, and dramatically reduced (by 94%) the frequency of late spontaneous seizures for at least 4 months following status epilepticus. Thus, acute KEAP1 inhibition following status epilepticus exerts a neuroprotective and disease-modifying effect, supporting the hypothesis that reactive oxygen species generation is a key event in the development of epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/metabolism , Epilepsy/therapy , Kelch-Like ECH-Associated Protein 1/metabolism , Animals , Animals, Newborn , Anticonvulsants/chemistry , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Epilepsy/chemically induced , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glutathione/metabolism , Kainic Acid/toxicity , Kelch-Like ECH-Associated Protein 1/genetics , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice, Transgenic , Mutation/genetics , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rats , Rats, Sprague-Dawley , Triterpenes/chemistry , Triterpenes/therapeutic use
19.
Brain ; 140(7): 1885-1899, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28575153

ABSTRACT

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented disulfide HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Subject(s)
Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Epilepsy/drug therapy , HMG-Box Domains/drug effects , HMGB1 Protein/blood , HMGB1 Protein/metabolism , Isothiocyanates/therapeutic use , Oxidative Stress/drug effects , Animals , Astrocytes/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Disease Models, Animal , Drug Therapy, Combination , Epilepsy/metabolism , HMGB1 Protein/biosynthesis , Hippocampus/metabolism , Isothiocyanates/pharmacology , Male , Nerve Degeneration/diet therapy , Neurons/metabolism , Rats , Sulfoxides
20.
Bipolar Disord ; 19(4): 285-294, 2017 06.
Article in English | MEDLINE | ID: mdl-28605109

ABSTRACT

OBJECTIVES: Mood stabilizers administered for bipolar disorder during pregnancy, such as valproic acid, can increase the risk of congenital anomalies in offspring. Valnoctamide is a valproic acid derivative associated with a decreased risk for congenital abnormalities in animals. The present study evaluated the efficacy and safety of valnoctamide monotherapy, compared to placebo, in the treatment of patients in an acute manic episode. METHODS: A 3-week, double-blind, randomized, placebo- and risperidone-controlled, parallel group trial was conducted on 173 patients in an acute manic episode. Patients were randomized to receive valnoctamide 1500 mg/d (n=71), risperidone 6 mg/d (n=32), or matching placebo (n=70). The primary outcome measure was the change in Young Mania Rating Scale (YMRS) scores. RESULTS: Valnoctamide did not differ significantly from placebo on any of the study endpoints (YMRS, Positive and Negative Syndrome Scale, and the Clinical Global Impression Scale for Bipolar Disorder [CGI-BP] scales; all P>.60). Mixed models for repeated measures showed that risperidone produced significantly more improvement than placebo in the overall bipolar disorder CGI-BP severity scale (P=.036), and the CGI-BP severity scale for mania (P=.021). The Kaplan-Meier survival curve revealed higher all-cause discontinuation rates (mainly due to lack of efficacy) in the valnoctamide group compared to the other study groups (P=.026). Patients with higher valnoctamide plasma levels had a numerically higher YMRS response, but this was not statistically significant. CONCLUSIONS: Valnoctamide was well tolerated at 1500 mg/d but lacked efficacy in the treatment of symptoms in patients with acute mania. Possible differences between the biological mechanisms of action of valproic acid and valnoctamide are discussed.


Subject(s)
Amides , Bipolar Disorder , Risperidone , Adult , Amides/administration & dosage , Amides/adverse effects , Antimanic Agents/administration & dosage , Antimanic Agents/adverse effects , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Bipolar Disorder/diagnosis , Bipolar Disorder/drug therapy , Bipolar Disorder/psychology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Monitoring/methods , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Psychiatric Status Rating Scales , Risperidone/administration & dosage , Risperidone/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...