1
|
Rawat K, Gautam V, Sandhu A, Kumar A, Sharma A, Bhatia A, Saha L. Wnt Signaling Modulators Exhibit Neuroprotective Effects via Combating Astrogliosis and Balancing Synaptic Density at Early and Late Stage Temporal Lobe Epilepsy. Neurochem Res 2024; 49:3156-3175. [PMID: 39235578 DOI: 10.1007/s11064-024-04236-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/17/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Temporal Lobe Epilepsy (TLE) is a severe neurological condition characterized by recurrent seizures that often do not respond well to available anti-seizure medications. TLE has been associated with epileptogenesis, a process that starts during the latent period following a neurologic insult and is followed by chronic phase. Recent research has linked canonical Wnt signaling to the pathophysiology of epileptogenesis and TLE. Our previous study demonstrated differential regulation of canonical Wnt signaling during early and late stage post status epilepticus (SE) induction. Building on these findings, our current study utilized Wnt modulators: GSK-3β inhibitor 6-bromoindirubin-3'-oxime (6-Bio) and disheveled inhibitor niclosamide and investigated their impact on canonical Wnt signaling during the early (30 days) and later stages (60 days) following SE induction. We assessed several parameters, including seizure frequency, astrogliosis, synaptic density, and neuronal counts in hippocampal tissue. We used immunohistochemistry and Nissl staining to evaluate gliosis, synaptic density, and neuronal counts in micro-dissected hippocampi. Western blotting was used to examine the expression of proteins involved in canonical Wnt/β-catenin signaling, and real-time PCR was conducted to analyze their relative mRNA expression. Wnt modulators, 6-Bio and Niclosamide were found to reduce seizure frequency and various other parameters including behavioral parameters, hippocampal morphology, astrogliosis and synaptic density at different stages of TLE.
Collapse
Affiliation(s)
- Kajal Rawat
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Arushi Sandhu
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Anil Kumar
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Antika Sharma
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Lekha Saha
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
2
|
Gautam V, Rawat K, Sandhu A, Kumar A, Kharbanda PS, Medhi B, Bhatia A, Saha L. Exploring the effect of 6-BIO and sulindac in modulation of Wnt/β-catenin signaling pathway in chronic phase of temporal lobe epilepsy. Neuropharmacology 2024; 251:109931. [PMID: 38570067 DOI: 10.1016/j.neuropharm.2024.109931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
The prospective involvement of the Wnt/β-catenin signaling pathway in epilepsy, with the proposed therapeutic uses of its modulators, has been suggested; however, comprehensive knowledge in this regard is currently limited. Despite postulations about the pathway's significance and treatment potential, a systematic investigation is required to better understand its implications in chronic epilepsy. We investigated the role of key proteins like β-catenin, GSK-3β, and their modulators sulindac and 6-BIO, in Wnt/β-catenin pathway during chronic phase of temporal lobe epilepsy. We also evaluated the role of modulators in seizure score, seizure frequency and neurobehavioral parameters in temporal lobe epilepsy. We developed status epilepticus model using lithium-pilocarpine. The assessment of neurobehavioral parameters was done followed by histopathological examination and immunohistochemistry staining of hippocampus as well as RT-qPCR and western blotting to analyse gene and protein expression. In SE rats, seizure score and frequency were significantly high compared to control rats, with notable changes in neurobehavioral parameters and neuronal damage observed in hippocampus. Our study also revealed a substantial upregulation of the Wnt/β-catenin pathway in chronic epilepsy, as evidenced by gene and protein expression studies. Sulindac emerged as a potent modulator, reducing seizure score, frequency, neuronal damage, apoptosis, and downregulating the Wnt/β-catenin pathway when compared to 6-BIO. Our findings emphasize the potential of GSK-3β and β-catenin as promising drug targets for chronic temporal lobe epilepsy, offering valuable treatment options for chronic epilepsy. The promising outcomes with sulindac encourages further exploration in clinical trials to assess its therapeutic potential.
Collapse
Affiliation(s)
- Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Parampreet Singh Kharbanda
- Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
3
|
Gautam V, Rawat K, Sandhu A, Medhi B, Bhatia A, Kharbanda PS, Saha L. Evaluation of Wnt/β-catenin signaling and its modulators in repeated dose lithium-pilocarpine rat model of status epilepticus: An acute phase study. Eur J Pharmacol 2024; 966:176375. [PMID: 38307381 DOI: 10.1016/j.ejphar.2024.176375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
The role of the Wnt/β-catenin signaling pathway in epilepsy and the effects of its modulators as efficacious treatment options, though postulated, has not been sufficiently investigated. We evaluated the involvement of β-catenin and GSK-3β, the significant proteins in this pathway, in the lithium chloride-pilocarpine-induced status epilepticus model in rodents to study acute phase of temporal lobe epilepsy (TLE). The modulators studied were 6-BIO, a GSK-3β inhibitor and Sulindac, a Dvl protein inhibitor. The disease group exhibited increased seizure score and seizure frequency, and the assessment of neurobehavioral parameters indicated notable alterations. Furthermore, histopathological examination of hippocampal brain tissues revealed significant neurodegeneration. Immunohistochemical study of hippocampus revealed neurogenesis in 6-BIO and sulindac groups. The gene and protein expression by RT-qPCR and western blotting studies indicated Wnt/β-catenin pathway downregulation and increased apoptosis in the acute phase of TLE. 6-BIO was very efficient in upregulating the Wnt pathway, decreasing neuronal damage, increasing neurogenesis in hippocampus and decreasing seizure score and frequency in comparison to sulindac. This suggests that both GSK-3β and β-catenin are potential and novel drug targets for acute phase of TLE, and treatment options targeting these proteins could be beneficial in successfully managing acute epilepsy. Further evaluation of 6-BIO to explore its therapeutic potential in other models of epilepsy should be conducted.
Collapse
Affiliation(s)
- Vipasha Gautam
- Department of Pharmacology, PGIMER Chandigarh, 160012, India
| | - Kajal Rawat
- Department of Pharmacology, PGIMER Chandigarh, 160012, India
| | - Arushi Sandhu
- Department of Pharmacology, PGIMER Chandigarh, 160012, India
| | - Bikash Medhi
- Department of Pharmacology, PGIMER Chandigarh, 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, 160012, India
| | | | - Lekha Saha
- Department of Pharmacology, PGIMER Chandigarh, 160012, India.
| |
Collapse
|
4
|
Rawat K, Gautam V, Sandhu A, Bhatia A, Saha L. Differential Regulation of Wnt/β-catenin Signaling in Acute and Chronic Epilepsy in Repeated Low Dose Lithium-Pilocarpine Rat Model of Status Epilepticus. Neuroscience 2023; 535:36-49. [PMID: 37913863 DOI: 10.1016/j.neuroscience.2023.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Epilepsy is a chronic neurological complication characterized by unprovoked seizure episodes due to the imbalance between excitatory and inhibitory neurons. The epileptogenesis process has been reported to be involved in chronic epilepsy however, the mechanism underlying epileptogenesis remains unclear. Recent studies have shown the possible involvement of Wnt/β-catenin signaling in the neurogenesis and neuronal reorganization in epileptogenesis. In this study, we used repeated low dose lithium-pilocarpine model of status epilepsy (SE) to study the involvement of Wnt/β-catenin signaling at acute and chronic stages post SE induction. The acute study ranged from day 0 to day 28 post SE induction and the chronic study ranged from day 0 to day 56 post SE induction. Several neurobehavioral parameters and seizure score and seizure frequency was analysed until the end of the study. The proteins involved in the regulation of Wnt/β-catenin signaling and downstream cascading were analysed using western blot and quantitative real-time PCR analysis. The Wnt/β-catenin pathway was found inactive in acute SE, while the same was found activated at the chronic stage. Our findings suggest that the activated Wnt/β-catenin signaling in chronic epilepsy might be the possible mechanism underlying epileptogenesis as indicated by increased neuronal count, increased synaptic density, astrogliosis and apoptosis in chronic epilepsy. These findings can help target the Wnt/β-catenin pathway differentially depending upon the type of epilepsy. The acute stage characterized by SE can be improved by targeting GSK-3β levels and the chronic stage characterized by temporal lobe epilepsy can be improved by targeting β-catenin and disheveled proteins.
Collapse
Affiliation(s)
- Kajal Rawat
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Arushi Sandhu
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Lekha Saha
- Department of Pharmacology, Research Block B, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India.
| |
Collapse
|
5
|
Kumar H, Katyal J, Kumar Gupta Y. Effect of U50488, a selective kappa opioid receptor agonist and levetiracetam against lithium-pilocarpine-induced status epilepticus, spontaneous convulsive seizures and related cognitive impairment. Neurosci Lett 2023; 815:137477. [PMID: 37714287 DOI: 10.1016/j.neulet.2023.137477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/16/2023] [Accepted: 08/29/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE Kappa opioid receptor (KOR) agonists have anticonvulsant effect but their antiepileptogenic effect is unknown. U50488, a selective KOR agonist is used to determine its effect on status epilepticus (SE), spontaneous convulsive seizures (SS) and cognitive impairment in rat lithium-pilocarpine SE model. Effect of an antiepileptic drug levetiracetam is also studied. METHOD Male Wistar rats with SE were divided into three groups namely, LiP, LiP + U50488 (10 mg/kg, i.p.) and LiP + levetiracetam (400 mg/kg, i.p.) group. SE was terminated after 90 min of its onset with diazepam (15 mg/kg, i.p.) and phenobarbitone (25 mg/kg, i.p.). Drug treatment was started after 15 min of onset of SE and repeated once after 4 h. Rats were video monitored 12 h daily (9 AM to 9 PM) to determine severity of SE using modified Racine scale and onset and frequency of SS from day 0 to day 21. Morris water maze (MWM) test was done at baseline i.e. day -1 (before lithium administration) and day 22, to assess cognitive impairment. RESULTS As compared to LiP, U50488 decreased the severity of SE (1.98 ± 0.13 vs 2.95 ± 0.12; p-value < 0.0001) but not levetiracetam (2.62 ± 0.09; p-value = 0.3112). Survival increased with both U50488 (90%, n = 10) and levetiracetam (81.8%, n = 11) as compared to NS (56.2%, n = 16). No effect on onset and frequency of SS was found in U50488/levetiracetam group. U50488 improved seizures-induced cognitive impairment. Levetiracetam group showed thigmotactic (wall hugging) behaviour in MWM in 8 out of 9 rats. CONCLUSION Acute treatment with U50488, a kappa opioid receptor agonist has a beneficial effect on SE, SE-related mortality and memory impairment. The dual protective effect of U50488 on seizures and related cognitive impairment is advantageous over currently used antiseizure drugs which are known to cause cognitive impairment.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Pharmacology, Faculty of Medicine and Health Sciences, SGT University, Gurgaon, Haryana, India.
| | - Jatinder Katyal
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | | |
Collapse
|
6
|
Casillas-Espinosa PM, Anderson A, Harutyunyan A, Li C, Lee J, Braine EL, Brady RD, Sun M, Huang C, Barlow CK, Shah AD, Schittenhelm RB, Mychasiuk R, Jones NC, Shultz SR, O'Brien TJ. Disease-modifying effects of sodium selenate in a model of drug-resistant, temporal lobe epilepsy. eLife 2023; 12:e78877. [PMID: 36892461 PMCID: PMC10208637 DOI: 10.7554/elife.78877] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 03/08/2023] [Indexed: 03/10/2023] Open
Abstract
There are no pharmacological disease-modifying treatments with an enduring effect to mitigate the seizures and comorbidities of established chronic temporal lobe epilepsy (TLE). This study aimed to evaluate for disease modifying effects of sodium selenate treatment in the chronically epileptic rat post-status epilepticus (SE) model of drug-resistant TLE. Wistar rats underwent kainic acid-induced SE or sham. Ten-weeks post-SE, animals received sodium selenate, levetiracetam, or vehicle subcutaneousinfusion continuously for 4 weeks. To evaluate the effects of the treatments, one week of continuous video-EEG was acquired before, during, and 4, 8 weeks post-treatment, followed by behavioral tests. Targeted and untargeted proteomics and metabolomics were performed on post-mortem brain tissue to identify potential pathways associated with modified disease outcomes. Telomere length was investigated as a novel surrogate marker of epilepsy disease severity in our current study. The results showed that sodium selenate treatment was associated with mitigation of measures of disease severity at 8 weeks post-treatment cessation; reducing the number of spontaneous seizures (p< 0.05), cognitive dysfunction (p< 0.05), and sensorimotor deficits (p< 0.01). Moreover, selenate treatment was associated with increased protein phosphatase 2A (PP2A) expression, reduced hyperphosphorylated tau, and reversed telomere length shortening (p< 0.05). Network medicine integration of multi-omics/pre-clinical outcomes identified protein-metabolite modules positively correlated with TLE. Our results provide evidence that treatment with sodium selenate results in a sustained disease-modifying effect in chronically epileptic rats in the post-KA SE model of TLE, including improved comorbid learning and memory deficits.
Collapse
Affiliation(s)
- Pablo M Casillas-Espinosa
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
- Monash Proteomics & Metabolomics Facility and Monash Biomedicine Discovery Institute, Monash UniversityClayton, VictoriaAustralia
| | - Alison Anderson
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Anna Harutyunyan
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Crystal Li
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Jiyoon Lee
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
| | - Emma L Braine
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Cheng Huang
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Christopher K Barlow
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Anup D Shah
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Ralf B Schittenhelm
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Nigel C Jones
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
- Monash Proteomics & Metabolomics Facility and Monash Biomedicine Discovery Institute, Monash UniversityClayton, VictoriaAustralia
| |
Collapse
|
7
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Contreras-García IJ, Cárdenas-Rodríguez N, Romo-Mancillas A, Bandala C, Zamudio SR, Gómez-Manzo S, Hernández-Ochoa B, Mendoza-Torreblanca JG, Pichardo-Macías LA. Levetiracetam Mechanisms of Action: From Molecules to Systems. Pharmaceuticals (Basel) 2022; 15:ph15040475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic disease that affects millions of people worldwide. Antiepileptic drugs (AEDs) are used to control seizures. Even though parts of their mechanisms of action are known, there are still components that need to be studied. Therefore, the search for novel drugs, new molecular targets, and a better understanding of the mechanisms of action of existing drugs is still crucial. Levetiracetam (LEV) is an AED that has been shown to be effective in seizure control and is well-tolerable, with a novel mechanism of action through an interaction with the synaptic vesicle protein 2A (SV2A). Moreover, LEV has other molecular targets that involve calcium homeostasis, the GABAergic system, and AMPA receptors among others, that might be integrated into a single mechanism of action that could explain the antiepileptogenic, anti-inflammatory, neuroprotective, and antioxidant properties of LEV. This puts it as a possible multitarget drug with clinical applications other than for epilepsy. According to the above, the objective of this work was to carry out a comprehensive and integrative review of LEV in relation to its clinical uses, structural properties, therapeutical targets, and different molecular, genetic, and systemic action mechanisms in order to consider LEV as a candidate for drug repurposing.
Collapse
Affiliation(s)
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico;
| | - Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| | - Julieta Griselda Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| |
Collapse
|
9
|
de Curtis M, Rossetti AO, Verde DV, van Vliet EA, Ekdahl CT. Brain pathology in focal status epilepticus: evidence from experimental models. Neurosci Biobehav Rev 2021; 131:834-846. [PMID: 34517036 DOI: 10.1016/j.neubiorev.2021.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/02/2021] [Accepted: 09/05/2021] [Indexed: 12/01/2022]
Abstract
Status Epilepticus (SE) is often a neurological emergency characterized by abnormally sustained, longer than habitual seizures. The new ILAE classification reports that SE "…can have long-term consequences including neuronal death, neuronal injury…depending on the type and duration of seizures". While it is accepted that generalized convulsive SE exerts detrimental effects on the brain, it is not clear if other forms of SE, such as focal non-convulsive SE, leads to brain pathology and contributes to long-term deficits in patients. With the available clinical and experimental data, it is hard to discriminate the specific action of the underlying SE etiologies from that exerted by epileptiform activity. This information is highly relevant in the clinic for better treatment stratification, which may include both medical and surgical intervention for seizure control. Here we review experimental studies of focal SE, with an emphasis on focal non-convulsive SE. We present a repertoire of brain pathologies observed in the most commonly used animal models and attempt to establish a link between experimental findings and human condition(s). The extensive literature on focal SE animal models suggest that the current approaches have significant limitations in terms of translatability of the findings to the clinic. We highlight the need for a more stringent description of SE features and brain pathology in experimental studies in animal models, to improve the accuracy in predicting clinical translation.
Collapse
Affiliation(s)
- Marco de Curtis
- Epilepsy Unit, Fondazione IRCCS Istituto NeurologicoCarlo Besta, Milano, Italy.
| | - Andrea O Rossetti
- Department of Clinical Neuroscience, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Diogo Vila Verde
- Epilepsy Unit, Fondazione IRCCS Istituto NeurologicoCarlo Besta, Milano, Italy
| | - Erwin A van Vliet
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, P.O. Box 94246, 1090 GE, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Christine T Ekdahl
- Division of Clinical Neurophysiology, Lund University, Sweden; Lund Epilepsy Center, Dept Clinical Sciences, Lund University, Sweden
| |
Collapse
|
10
|
Scopolamine prevents aberrant mossy fiber sprouting and facilitates remission of epilepsy after brain injury. Neurobiol Dis 2021; 158:105446. [PMID: 34280524 DOI: 10.1016/j.nbd.2021.105446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/01/2021] [Accepted: 07/13/2021] [Indexed: 11/21/2022] Open
Abstract
Prevention or modification of acquired epilepsy in patients at risk is an urgent, yet unmet, clinical need. Following acute brain insults, there is an increased risk of mesial temporal lobe epilepsy (mTLE), which is often associated with debilitating comorbidities and reduced life expectancy. The latent period between brain injury and the onset of epilepsy may offer a therapeutic window for interfering with epileptogenesis. The pilocarpine model of mTLE is widely used in the search for novel antiepileptogenic treatments. Recent biochemical studies indicated that cholinergic mechanisms play a role in the epileptogenic alterations induced by status epilepticus (SE) in this and other models of mTLE, which prompted us to evaluate whether treatment with the muscarinic antagonist scopolamine during the latent period after SE is capable of preventing or modifying epilepsy and associated behavioral and cognitive alterations in female Sprague-Dawley rats. First, in silico pharmacokinetic modeling was used to select a dosing protocol by which M-receptor inhibitory brain levels of scopolamine are maintained during prolonged treatment. This protocol was verified by drug analysis in vivo. Rats were then treated twice daily with scopolamine over 17 days after SE, followed by drug wash-out and behavioral and video/EEG monitoring up to ~6 months after SE. Compared to vehicle controls, rats that were treated with scopolamine during the latent period exhibited a significantly lower incidence of spontaneous recurrent seizures during periods of intermittent recording in the chronic phase of epilepsy, less behavioral excitability, less cognitive impairment, and significantly reduced aberrant mossy fiber sprouting in the hippocampus. The present data may indicate that scopolamine exerts antiepileptogenic/disease-modifying activity in the lithium-pilocarpine rat model, possibly involving increased remission of epilepsy as a new mechanism of disease-modification. For evaluating the rigor of the present data, we envision a study that more thoroughly addresses the gender bias and video-EEG recording limitations of the present study.
Collapse
|
11
|
Welzel L, Bergin DH, Schidlitzki A, Twele F, Johne M, Klein P, Löscher W. Systematic evaluation of rationally chosen multitargeted drug combinations: a combination of low doses of levetiracetam, atorvastatin and ceftriaxone exerts antiepileptogenic effects in a mouse model of acquired epilepsy. Neurobiol Dis 2020; 149:105227. [PMID: 33347976 DOI: 10.1016/j.nbd.2020.105227] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/24/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023] Open
Abstract
Epileptogenesis, the gradual process that leads to epilepsy after brain injury or genetic mutations, is a complex network phenomenon, involving a variety of morphological, biochemical and functional brain alterations. Although risk factors for developing epilepsy are known, there is currently no treatment available to prevent epilepsy. We recently proposed a multitargeted, network-based approach to prevent epileptogenesis by rationally combining clinically available drugs and provided first proof-of-concept that this strategy is effective. Here we evaluated eight novel rationally chosen combinations of 14 drugs with mechanisms that target different epileptogenic processes. The combinations consisted of 2-4 different drugs per combination and were administered systemically over 5 days during the latent epileptogenic period in the intrahippocampal kainate mouse model of acquired temporal lobe epilepsy, starting 6 h after kainate. Doses and dosing intervals were based on previous pharmacokinetic and tolerability studies in mice. The incidence and frequency of spontaneous electrographic and electroclinical seizures were recorded by continuous (24/7) video linked EEG monitoring done for seven days at 4 and 12 weeks post-kainate, i.e., long after termination of drug treatment. Compared to vehicle controls, the most effective drug combination consisted of low doses of levetiracetam, atorvastatin and ceftriaxone, which markedly reduced the incidence of electrographic seizures (by 60%; p<0.05) and electroclinical seizures (by 100%; p<0.05) recorded at 12 weeks after kainate. This effect was lost when higher doses of the three drugs were administered, indicating a synergistic drug-drug interaction at the low doses. The potential mechanisms underlying this interaction are discussed. We have discovered a promising novel multitargeted combination treatment for modifying the development of acquired epilepsy.
Collapse
Affiliation(s)
- Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - David H Bergin
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
12
|
Yang L, Afroz S, Valsamis HA, Michelson HB, Goodman JH, Ling DSF. Early intervention with levetiracetam prevents the development of cortical hyperexcitability and spontaneous epileptiform activity in two models of neurotrauma in rats. Exp Neurol 2020; 337:113571. [PMID: 33340499 DOI: 10.1016/j.expneurol.2020.113571] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 12/13/2020] [Indexed: 10/22/2022]
Abstract
This study examined the antiepileptogenic potential of the antiseizure drug (ASD) levetiracetam (LEV) using the in vitro traumatized-slice and in vivo controlled cortical impact (CCI) models of traumatic brain injury (TBI) in rats when administered early after the injury. For the in vitro model, acute coronal slices (400-450 μm) of rat neocortex (P21-32) were injured via a surgical cut that separated the superficial layers from the deeper regions. Persistent stimulus-evoked epileptiform activity developed within 1-2 h after trauma. In randomly selected slices, LEV (500 μM) was bath-applied for 1 h starting immediately or delayed by 30-80 min after injury. Treated and untreated slices were examined for epileptiform activity via intracellular and extracellular recordings. For the in vivo model, rats (P24-32) were subjected to a non-penetrating, focal, CCI injury targeting the neocortex (5.0 mm diameter; 2.0 mm depth). Immediately after injury, rats were given either a single dose of LEV (60-150 mg/kg, i.p.) or the saline vehicle. At 2-3 weeks after the injury, ex vivo cortical slices were examined for epileptiform activity. The results from the traumatized-slice experiments showed that in vitro treatment with LEV within 60 min of injury significantly reduced (> 50%) the proportion of slices that exhibited stimulus-evoked epileptiform activity. LEV treatment also increased the stimulus intensity required to trigger epileptiform bursts in injured slices by 2-4 fold. Consistent with these findings, LEV treatment of CCI-injured rats (n = 15) significantly reduced the proportion of animals that exhibited spontaneous and stimulus-evoked epileptiform bursts in ex vivo cortical slices compared to saline-treated controls (n = 15 rats), and also significantly increased the stimulus intensity required to evoke epileptiform bursts. These results suggest that early administration of LEV has the potential to prevent or reduce posttraumatic epileptogenesis and that there may be a narrow therapeutic window for successful prophylactic intervention.
Collapse
Affiliation(s)
- Lie Yang
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Sonia Afroz
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Helen A Valsamis
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Neurology Service, Kings County Hospital Center, Brooklyn, NY 11203, USA.
| | - Hillary B Michelson
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| | - Jeffrey H Goodman
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| | - Douglas S F Ling
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| |
Collapse
|
13
|
Miziak B, Konarzewska A, Ułamek-Kozioł M, Dudra-Jastrzębska M, Pluta R, Czuczwar SJ. Anti-Epileptogenic Effects of Antiepileptic Drugs. Int J Mol Sci 2020; 21:ijms21072340. [PMID: 32231010 PMCID: PMC7178140 DOI: 10.3390/ijms21072340] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
Generally, the prevalence of epilepsy does not exceed 0.9% of the population and approximately 70% of epilepsy patients may be adequately controlled with antiepileptic drugs (AEDs). Moreover, status epilepticus (SE) or even a single seizure may produce neurodegeneration within the brain and SE has been recognized as one of acute brain insults leading to acquired epilepsy via the process of epileptogenesis. Two questions thus arise: (1) Are AEDs able to inhibit SE-induced neurodegeneration? and (2) if so, can a probable neuroprotective potential of particular AEDs stop epileptogenesis? An affirmative answer to the second question would practically point to the preventive potential of a given neuroprotective AED following acute brain insults. The available experimental data indicate that diazepam (at low and high doses), gabapentin, pregabalin, topiramate and valproate exhibited potent or moderate neuroprotective effects in diverse models of SE in rats. However, only diazepam (at high doses), gabapentin and pregabalin exerted some protective activity against acquired epilepsy (spontaneous seizures). As regards valproate, its effects on spontaneous seizures were equivocal. With isobolography, some supra-additive combinations of AEDs have been delineated against experimental seizures. One of such combinations, levetiracetam + topiramate proved highly synergistic in two models of seizures and this particular combination significantly inhibited epileptogenesis in rats following status SE. Importantly, no neuroprotection was evident. It may be strikingly concluded that there is no correlation between neuroprotection and antiepileptogenesis. Probably, preclinically verified combinations of AEDs may be considered for an anti-epileptogenic therapy.
Collapse
Affiliation(s)
- Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Agnieszka Konarzewska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Marzena Ułamek-Kozioł
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Monika Dudra-Jastrzębska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| |
Collapse
|
14
|
Klein P, Friedman A, Hameed MQ, Kaminski RM, Bar-Klein G, Klitgaard H, Koepp M, Jozwiak S, Prince DA, Rotenberg A, Twyman R, Vezzani A, Wong M, Löscher W. Repurposed molecules for antiepileptogenesis: Missing an opportunity to prevent epilepsy? Epilepsia 2020; 61:359-386. [PMID: 32196665 PMCID: PMC8317585 DOI: 10.1111/epi.16450] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland
| | - Alon Friedman
- Departments of Physiology and Cell Biology, and Brain and Cognitive Science, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Departments of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Canada
| | - Mustafa Q. Hameed
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rafal M. Kaminski
- Neurosymptomatic Domains Section, Roche Pharma Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Guy Bar-Klein
- McKusick-Nathans Institute of Genetic Medicine, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henrik Klitgaard
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l’Alleud, Belgium
| | - Mathias Koepp
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Sergiusz Jozwiak
- Department of Pediatric Neurology, Warsaw Medical University, Warsaw, Poland
| | - David A. Prince
- Neurology and the Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Scientific Institute for Research and Health Care, Milan, Italy
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
15
|
Romoli M, Mazzocchetti P, D'Alonzo R, Siliquini S, Rinaldi VE, Verrotti A, Calabresi P, Costa C. Valproic Acid and Epilepsy: From Molecular Mechanisms to Clinical Evidences. Curr Neuropharmacol 2020; 17:926-946. [PMID: 30592252 PMCID: PMC7052829 DOI: 10.2174/1570159x17666181227165722] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/03/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
After more than a century from its discovery, valproic acid (VPA) still represents one of the most efficient antiepi-leptic drugs (AEDs). Pre and post-synaptic effects of VPA depend on a very broad spectrum of actions, including the regu-lation of ionic currents and the facilitation of GABAergic over glutamatergic transmission. As a result, VPA indirectly mod-ulates neurotransmitter release and strengthens the threshold for seizure activity. However, even though participating to the anticonvulsant action, such mechanisms seem to have minor impact on epileptogenesis. Nonetheless, VPA has been reported to exert anti-epileptogenic effects. Epigenetic mechanisms, including histone deacetylases (HDACs), BDNF and GDNF modulation are pivotal to orientate neurons toward a neuroprotective status and promote dendritic spines organization. From such broad spectrum of actions comes constantly enlarging indications for VPA. It represents a drug of choice in child and adult with epilepsy, with either general or focal seizures, and is a consistent and safe IV option in generalized convulsive sta-tus epilepticus. Moreover, since VPA modulates DNA transcription through HDACs, recent evidences point to its use as an anti-nociceptive in migraine prophylaxis, and, even more interestingly, as a positive modulator of chemotherapy in cancer treatment. Furthermore, VPA-induced neuroprotection is under investigation for benefit in stroke and traumatic brain injury. Hence, VPA has still got its place in epilepsy, and yet deserves attention for its use far beyond neurological diseases. In this review, we aim to highlight, with a translational intent, the molecular basis and the clinical indications of VPA.
Collapse
Affiliation(s)
- Michele Romoli
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Petra Mazzocchetti
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Renato D'Alonzo
- Pediatric Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | | | - Victoria Elisa Rinaldi
- Pediatric Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Alberto Verrotti
- Department of Pediatrics, University of L'Aquila - San Salvatore Hospital, L'Aquila, Italy
| | - Paolo Calabresi
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy.,IRCCS "Santa Lucia", Rome, Italy
| | - Cinzia Costa
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
16
|
Casillas-Espinosa PM, Shultz SR, Braine EL, Jones NC, Snutch TP, Powell KL, O’Brien TJ. Disease-modifying effects of a novel T-type calcium channel antagonist, Z944, in a model of temporal lobe epilepsy. Prog Neurobiol 2019; 182:101677. [DOI: 10.1016/j.pneurobio.2019.101677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 02/08/2023]
|
17
|
Liu H, Stover KR, Sivanenthiran N, Chow J, Cheng C, Liu Y, Lim S, Wu C, Weaver DF, Eubanks JH, Song H, Zhang L. Impaired Spatial Learning and Memory in Middle-Aged Mice with Kindling-Induced Spontaneous Recurrent Seizures. Front Pharmacol 2019; 10:1077. [PMID: 31611787 PMCID: PMC6768971 DOI: 10.3389/fphar.2019.01077] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
Temporal lobe epilepsy is the most common and often drug-resistant type of epilepsy in the adult and aging populations and has great diversity in etiology, electro-clinical manifestations, and comorbidities. Kindling through repeated brief stimulation of limbic structures is a commonly used model of temporal lobe epilepsy. Particularly, extended kindling can induce spontaneous recurrent seizures in several animal species. However, kindling studies in middle-aged, aging, or aged animals remain scarce, and currently, little is known about kindling-induced behavioral changes in middle-aged/aging animals. We therefore attempted to provide more information in this area using a mouse model of extended hippocampal kindling. We conducted experiments in middle-aged mice (C57BL/6, male, 12-14 months of age) to model new-onset epilepsy in adult/aging populations. Mice experienced twice daily hippocampal stimulations or handling manipulations for 60-70 days and then underwent continuous electroencephalogram (EEG)-video monitoring to detect spontaneous recurrent seizures. Extended kindled mice consistently exhibited spontaneous recurrent seizures with mean incidences of 6-7 events per day, and these seizures featured EEG discharges and corresponding convulsions. The handling control mice showed neither seizure nor aberrant EEG activity. The two groups of mice underwent the Morris water maze test of spatial learning and memory 1-2 weeks after termination of the kindling stimulation or handling manipulation. During visible platform trials, the kindled mice took a longer distance and required more time than the control mice to find the platform. During hidden platform trials, the kindled mice showed no improvement over 5-day trials in finding the platform whereas the control mice improved significantly. During probe tests in which the hidden platform was removed, the kindled mice spent less time than the controls searching in the correct platform location. There were no significant differences between the kindled and control mice with respect to swim speed or total locomotor activity in an open-field test. Together, these observations indicate that the extended kindled mice with spontaneous recurrent seizures are impaired in spatial learning and memory as assessed by the Morris water maze test. We postulate that the extended hippocampal kindling in middle-aged mice may help explore epileptogenic mechanisms and comorbidities potentially relevant to new-onset temporal lobe epilepsy in adult and aging patients. Limitations and confounds of our present experiments are discussed to improve future examinations of epileptic comorbidities in extended kindled mice.
Collapse
Affiliation(s)
- Haiyu Liu
- Department of Neurosurgery, The First Hospital of Jilin University, Jilin, China
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Kurt R. Stover
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Nila Sivanenthiran
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Jonathan Chow
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Chloe Cheng
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Yapeng Liu
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Stellar Lim
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Chiping Wu
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Donald F. Weaver
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - James H. Eubanks
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Hongmei Song
- Department of Neurosurgery, The First Hospital of Jilin University, Jilin, China
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Liang Zhang
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Löscher W. The holy grail of epilepsy prevention: Preclinical approaches to antiepileptogenic treatments. Neuropharmacology 2019; 167:107605. [PMID: 30980836 DOI: 10.1016/j.neuropharm.2019.04.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
A variety of acute brain insults can induce epileptogenesis, a complex process that results in acquired epilepsy. Despite advances in understanding mechanisms of epileptogenesis, there is currently no approved treatment that prevents the development or progression of epilepsy in patients at risk. The current concept of epileptogenesis assumes a window of opportunity following acute brain insults that allows intervention with preventive treatment. Recent results suggest that injury-induced epileptogenesis can be a much more rapid process than previously thought, suggesting that the 'therapeutic window' may only be open for a brief period, as in stroke therapy. However, experimental data also suggest a second, possibly delayed process ("secondary epileptogenesis") that influences the progression and refractoriness of the epileptic state over time, allowing interfering with this process even after onset of epilepsy. In this review, both methodological issues in preclinical drug development and novel targets for antiepileptogenesis will be discussed. Several promising drugs that either prevent epilepsy (antiepileptogenesis) or slow epilepsy progression and alleviate cognitive or behavioral comorbidities of epilepsy (disease modification) have been described in recent years, using diverse animal models of acquired epilepsy. Promising agents include TrkB inhibitors, losartan, statins, isoflurane, anti-inflammatory and anti-oxidative drugs, the SV2A modulator levetiracetam, and epigenetic interventions. Research on translational target validity and on prognostic biomarkers that can be used to stratify patients (or experimental animals) at high risk of developing epilepsy will hopefully soon lead to proof-of-concept clinical trials with the most promising drugs, which will be essential to make prevention of epilepsy a reality. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
19
|
Santana-Gómez CE, Valle-Dorado MG, Domínguez-Valentín AE, Hernández-Moreno A, Orozco-Suárez S, Rocha L. Neuroprotective effects of levetiracetam, both alone and combined with propylparaben, in the long-term consequences induced by lithium-pilocarpine status epilepticus. Neurochem Int 2018; 120:224-232. [PMID: 30213635 DOI: 10.1016/j.neuint.2018.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 12/27/2022]
Abstract
Status epilepticus (SE) is a neurological condition that frequently induces severe neuronal injury in the hippocampus, subsequent epileptogenesis and pharmacoresistant spontaneous recurrent seizures (SRS). The repeated administration of LEV (a broad-spectrum antiepileptic drug) during the post-SE period does not prevent the subsequent development of SRS. However, this treatment reduces SE-induced neurodegeneration in the hippocampus. Conversely, propylparaben (PPB) is a widely used antimicrobial that blocks voltage-dependent Na+ channels, induces neuroprotection and reduces epileptiform activity in vitro. The present study attempted to determine if the neuroprotective effects induced by LEV are augmented when combined with a sub-effective dose of PPB. Long-term SE-induced consequences (hyperexcitability, high glutamate release, neuronal injury and volume loss) were evaluated in the hippocampus of rats. LEV alone, as well as combined with PPB, did not prevent the occurrence of SRS. However, animals treated with LEV plus PPB showed high prevalence of low frequency oscillations (0.1-4 Hz and 8-90 bands, p < 0.001) and low prevalence of high frequency activity (90-250 bands, p < 0.001) during the interictal period. In addition, these animals presented lower extracellular levels of glutamate, decreased rate of neurodegeneration and a similar hippocampal volume compared to the control conditions. This study's results suggest that LEV associated with PPB could represent a new therapeutic strategy to reduce long-term consequences induced by SE that facilitate pharmacoresistant SRS.
Collapse
Affiliation(s)
| | | | | | | | - Sandra Orozco-Suárez
- Unit for Medical Research in Neurological Diseases, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
20
|
Reddy DS, Yoshimura RF, Ramanathan G, Carver C, Johnstone TB, Hogenkamp DJ, Gee KW. Role of β 2/3-specific GABA-A receptor isoforms in the development of hippocampus kindling epileptogenesis. Epilepsy Behav 2018; 82:57-63. [PMID: 29587186 DOI: 10.1016/j.yebeh.2018.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/16/2018] [Accepted: 02/20/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Subunit-specific positive allosteric modulators (PAMs) of gamma-aminobutyric acid-A (GABA-A) receptors are commonly used to uncover the role of GABA-A receptor isoforms in brain function. Recently, we have designed novel PAMs selective for β2/3-subunit containing GABA-A receptors (β2/3-selective PAMs) that are nonbenzodiazepine site-mediated and do not show an α-subunit isoform selectivity, yet exhibit anxiolytic efficacy with reduced potential for sedation, cognitive impairment, and tolerance. In this study, we used three novel β2/3-selective PAMs (2-261, 2-262, and 10029) with differential β2/3-subunit potency to identify the role of β2/3-selective receptor isoforms in limbic epileptogenesis. METHODS Experimental epileptogenesis was induced in mice by daily hippocampus stimulations until each mouse showed generalized (stage 5) seizures. Patch-clamp electrophysiology was used to record GABA-gated currents. Brain levels of β2/3-selective PAMs were determined for mechanistic correlations. RESULTS Treatment with the β2/3-selective PAMs 2-261 (30mg/kg), 2-262 (10mg/kg), and 10029 (30mg/kg), 30min prior to stimulations, significantly suppressed the rate of development of kindled seizure activity without affecting the afterdischarge (AD) signal, indicating their disease-modifying activity. The β2/3-selective agents suppressed chemical epileptogenesis in the pentylenetetrazol model. Test doses of these agents were devoid of acute antiseizure activity in the kindling model. CONCLUSION These findings demonstrate that β2/3-selective PAMs can moderately retard experimental epileptogenesis, indicating the protective role of β2/3-subunit GABA-A receptor isoforms in the development of epilepsy.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, United States.
| | - Ryan F Yoshimura
- Department of Pharmacology, School of Medicine, University of California, Irvine, California, United States
| | - Gunasekaran Ramanathan
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Chase Carver
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Timothy B Johnstone
- Department of Pharmacology, School of Medicine, University of California, Irvine, California, United States
| | - Derk J Hogenkamp
- Department of Pharmacology, School of Medicine, University of California, Irvine, California, United States
| | - Kelvin W Gee
- Department of Pharmacology, School of Medicine, University of California, Irvine, California, United States
| |
Collapse
|
21
|
Abbasova K, Kubová H, Mareš P. Does status epilepticus modify the effect of ifenprodil on cortical epileptic afterdischarges in immature rats? Pharmacol Rep 2018; 70:126-132. [PMID: 29355816 DOI: 10.1016/j.pharep.2017.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/26/2017] [Accepted: 08/23/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Ifenprodil as a specific antagonist of NMDA receptors containing a dominant NR2B subunit exhibits age-dependent anticonvulsant action. Possible changes of this action due to status epilepticus (SE) elicited at early stage of development were studied using cortical epileptic afterdischarges (ADs) as a model. METHODS Lithium-pilocarpine SE was induced at postnatal day 12 and effects of ifenprodil were studied 3, 6, 9, and 13 days after SE in rat pups with implanted epidural electrodes. Controls (LiPAR) received saline instead of pilocarpine. ADs were elicited by low frequency stimulation of sensorimotor cortex. Intensity of stimulation current increased in 18 steps from 0.2 to 15 mA. Ifenprodil (20 mg/kg) was administered intraperitoneally (ip) after the stimulation with 3.5-mA current. Threshold for four different phenomena as well as duration of ADs were evaluated. RESULTS The threshold for the transition into the limbic type of ADs was higher in 15-day-old SE rats than in LiPAR controls. Opposite difference was found in 18-day-old animals, older rats did not exhibit any difference. Isolated significant changes in total duration of ADs were found after high stimulation intensities. These changes appeared in 18-day-old rats where ADs were shorter in SE than in control LiPAR rats. CONCLUSIONS Changes in ifenprodil action were found only in the first week after SE but not in the second week. Interpretation of the results is complicated by failure of significant differences between SE and LiPAR rats probably due to a high dose of paraldehyde.
Collapse
Affiliation(s)
- Kenul Abbasova
- Department of Developmental Epileptology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic; Faculty of Biology, Moscow State University, Moscow, Russia
| | - Hana Kubová
- Department of Developmental Epileptology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Mareš
- Department of Developmental Epileptology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
22
|
Reddy SD, Clossen BL, Reddy DS. Epigenetic Histone Deacetylation Inhibition Prevents the Development and Persistence of Temporal Lobe Epilepsy. J Pharmacol Exp Ther 2018; 364:97-109. [PMID: 29101217 DOI: 10.1124/jpet.117.244939] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/31/2017] [Indexed: 03/08/2025] Open
Abstract
Epilepsy is a chronic brain disease characterized by repeated unprovoked seizures. Currently, no drug therapy exists for curing epilepsy or disease modification in people at risk. Despite several emerging mechanisms, there have been few studies of epigenetic signaling in epileptogenesis, the process whereby a normal brain becomes progressively epileptic because of precipitating factors. Here, we report a novel role of histone deacetylation as a critical epigenetic mechanism in epileptogenesis. Experiments were conducted using the histone deacetylase (HDAC) inhibitor sodium butyrate in the hippocampus kindling model of temporal lobe epilepsy (TLE), a classic model heavily used to approve drugs for treatment of epilepsy. Daily treatment with butyrate significantly inhibited HDAC activity and retarded the development of limbic epileptogenesis without affecting after-discharge signal. HDAC inhibition markedly impaired the persistence of seizure expression many weeks after epilepsy development. Moreover, subchronic HDAC inhibition for 2 weeks resulted in a striking retardation of epileptogenesis. HDAC inhibition, unexpectedly, also showed erasure of the epileptogenic state in epileptic animals. Finally, butyrate-treated animals exhibited a powerful reduction in mossy fiber sprouting, a morphologic index of epileptogenesis. Together these results underscore that HDAC inhibition prevents the development of TLE, indicating HDAC's critical signaling role in epileptogenesis. These findings, therefore, envisage a unique novel therapy for preventing or curing epilepsy by targeting the epigenetic HDAC pathway.
Collapse
Affiliation(s)
- Sandesh D Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, Texas
| | - Bryan L Clossen
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, Texas
| |
Collapse
|
23
|
Abstract
This article highlights the emerging therapeutic potential of specific epigenetic modulators as promising antiepileptogenic or disease-modifying agents for curing epilepsy. Currently, there is an unmet need for antiepileptogenic agents that truly prevent the development of epilepsy in people at risk. There is strong evidence that epigenetic signaling, which exerts high fidelity regulation of gene expression, plays a crucial role in the pathophysiology of epileptogenesis and chronic epilepsy. These modifications are not hard-wired into the genome and are constantly reprogrammed by environmental influences. The potential epigenetic mechanisms, including histone modifications, DNA methylation, microRNA-based transcriptional control, and bromodomain reading activity, can drastically alter the neuronal gene expression profile by exerting their summative effects in a coordinated fashion. Such an epigenetic intervention appears more rational strategy for preventing epilepsy because it targets the primary pathway that initially triggers the numerous downstream cellular and molecular events mediating epileptogenesis. Among currently approved epigenetic drugs, the majority are anticancer drugs with well-established profiles in clinical trials and practice. Evidence from preclinical studies supports the premise that these drugs may be applied to a wide range of brain disorders. Targeting histone deacetylation by inhibiting histone deacetylase enzymes appears to be one promising epigenetic therapy since certain inhibitors have been shown to prevent epileptogenesis in animal models. However, developing neuronal specific epigenetic modulators requires rational, pathophysiology-based optimization to efficiently intercept the upstream pathways in epileptogenesis. Overall, epigenetic agents have been well positioned as new frontier tools towards the national goal of curing epilepsy.
Collapse
Affiliation(s)
- Iyan Younus
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
24
|
Sarangi SC, Kakkar AK, Kumar R, Gupta YK. Effect of lamotrigine, levetiracetam & topiramate on neurobehavioural parameters & oxidative stress in comparison with valproate in rats. Indian J Med Res 2017; 144:104-111. [PMID: 27834333 PMCID: PMC5116881 DOI: 10.4103/0971-5916.193296] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background & objectives: Though newer antiepileptic drugs are considered safer than conventional antiepileptics, the effects of lamotrigine, levetiracetam and topiramate on neurobehavioural functions are yet to be established. This study evaluated neurobehavioural parameters and oxidative stress markers in brain tissue of rats treated with lamotrigine, levetiracetam and topiramate compared to sodium valproate. Methods: Five groups of male Wistar rats were treated respectively with normal saline (control), sodium valproate (370 mg/kg), lamotrigine (50 mg/kg), levetiracetam (310 mg/kg) and topiramate (100 mg/kg) for 45 days. Neurobehavioural parameters were assessed using elevated plus maze (EPM), actophotometer, rotarod, passive avoidance and Morris water maze (MWM) at baseline and at the end of treatment. Oxidative stress parameters [malondialdehyde (MDA), reduced glutathione (GSH) and superoxide dismutase (SOD)] were estimated in rat brain at the end of treatment. Results: Valproate and lamotrigine showed no significant effect on learning and memory in passive avoidance and MWM tests. However, levetiracetam and topiramate reduced retention memory significantly as compared to control (P<0.01) and lamotrigine (P<0.05) groups. Performances on EPM, rotarod and actophotometer were not significantly different between the groups. In comparison to control group, MDA was higher in the levetiracetam and topiramate (360.9 and 345.9 nmol/g of homogenized brain tissue, respectively) groups. GSH and SOD activity were significantly reduced by valproate and levetiracetam treatment. Lamotrigine did not induce significant oxidative stress. Interpretation & conclusions: Long-term and therapeutic dose treatment with levetiracetam and topiramate significantly impaired learning and memory, which was not seen with valproate and lamotrigine in rats. Levetiracetam, topiramate and valproate augmented oxidative stress, whereas lamotrigine has little effect on it. These antiepileptic drugs are used in clinical practice, hence pharmacovigilance studies are required to evaluate their safety profile.
Collapse
Affiliation(s)
| | - Ashish Kumar Kakkar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi; Department of Pharmacology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Ritesh Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
25
|
Clossen BL, Reddy DS. Novel therapeutic approaches for disease-modification of epileptogenesis for curing epilepsy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1519-1538. [PMID: 28179120 PMCID: PMC5474195 DOI: 10.1016/j.bbadis.2017.02.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 11/16/2022]
Abstract
This article describes the recent advances in epileptogenesis and novel therapeutic approaches for the prevention of epilepsy, with a special emphasis on the pharmacological basis of disease-modification of epileptogenesis for curing epilepsy. Here we assess animal studies and human clinical trials of epilepsy spanning 1982-2016. Epilepsy arises from a number of neuronal factors that trigger epileptogenesis, which is the process by which a brain shifts from a normal physiologic state to an epileptic condition. The events precipitating these changes can be of diverse origin, including traumatic brain injury, cerebrovascular damage, infections, chemical neurotoxicity, and emergency seizure conditions such as status epilepticus. Expectedly, the molecular and system mechanisms responsible for epileptogenesis are not well defined or understood. To date, there is no approved therapy for the prevention of epilepsy. Epigenetic dysregulation, neuroinflammation, and neurodegeneration appear to trigger epileptogenesis. Targeted drugs are being identified that can truly prevent the development of epilepsy in at-risk people. The promising agents include rapamycin, COX-2 inhibitors, TRK inhibitors, epigenetic modulators, JAK-STAT inhibitors, and neurosteroids. Recent evidence suggests that neurosteroids may play a role in modulating epileptogenesis. A number of promising drugs are under investigation for the prevention or modification of epileptogenesis to halt the development of epilepsy. Some drugs in development appear rational for preventing epilepsy because they target the initial trigger or related signaling pathways as the brain becomes progressively more prone to seizures. Additional research into the target validity and clinical investigation is essential to make new frontiers in curing epilepsy.
Collapse
Affiliation(s)
- Bryan L Clossen
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
26
|
Russmann V, Brendel M, Mille E, Helm-Vicidomini A, Beck R, Günther L, Lindner S, Rominger A, Keck M, Salvamoser JD, Albert NL, Bartenstein P, Potschka H. Identification of brain regions predicting epileptogenesis by serial [ 18F]GE-180 positron emission tomography imaging of neuroinflammation in a rat model of temporal lobe epilepsy. NEUROIMAGE-CLINICAL 2017; 15:35-44. [PMID: 28462087 PMCID: PMC5403805 DOI: 10.1016/j.nicl.2017.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
Excessive activation of inflammatory signaling pathways seems to be a hallmark of epileptogenesis. Positron emission tomography (PET) allows in vivo detection of brain inflammation with spatial information and opportunities for longitudinal follow-up scanning protocols. Here, we assessed whether molecular imaging of the 18 kDa translocator protein (TSPO) can serve as a biomarker for the development of epilepsy. Therefore, brain uptake of [18F]GE-180, a highly selective radioligand of TSPO, was investigated in a longitudinal PET study in a chronic rat model of temporal lobe epilepsy. Analyses revealed that the influence of the epileptogenic insult on [18F]GE-180 brain uptake was most pronounced in the earlier phase of epileptogenesis. Differences were evident in various brain regions during earlier phases of epileptogenesis with [18F]GE-180 standardized uptake value enhanced by 2.1 to 2.7fold. In contrast, brain regions exhibiting differences seemed to be more restricted with less pronounced increases of tracer uptake by 1.8-2.5fold four weeks following status epilepticus and by 1.5-1.8fold in the chronic phase. Based on correlation analysis, we were able to identify regions with a predictive value showing a correlation with seizure development. These regions include the amygdala as well as a cluster of brain areas. This cluster comprises parts of different brain regions, e.g. the hippocampus, parietal cortex, thalamus, and somatosensory cortex. In conclusion, the data provide evidence that [18F]GE-180 PET brain imaging can serve as a biomarker of epileptogenesis. The identification of brain regions with predictive value might facilitate the development of preventive concepts as well as the early assessment of the interventional success. Future studies are necessary to further confirm the predictivity of the approach.
Collapse
Affiliation(s)
- Vera Russmann
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Erik Mille
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Angela Helm-Vicidomini
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Roswitha Beck
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany; German Center for Vertigo and Balance Disorders, DSGZ, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Lisa Günther
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany; German Center for Vertigo and Balance Disorders, DSGZ, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Michael Keck
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Josephine D Salvamoser
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
27
|
Propylparaben applied after pilocarpine-induced status epilepticus modifies hippocampal excitability and glutamate release in rats. Neurotoxicology 2017; 59:110-120. [DOI: 10.1016/j.neuro.2017.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 01/06/2017] [Accepted: 01/31/2017] [Indexed: 11/19/2022]
|
28
|
Itoh K, Ishihara Y, Komori R, Nochi H, Taniguchi R, Chiba Y, Ueno M, Takata-Tsuji F, Dohgu S, Kataoka Y. Levetiracetam treatment influences blood-brain barrier failure associated with angiogenesis and inflammatory responses in the acute phase of epileptogenesis in post-status epilepticus mice. Brain Res 2016; 1652:1-13. [PMID: 27693413 DOI: 10.1016/j.brainres.2016.09.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022]
Abstract
Our previous study showed that treatment with levetiracetam (LEV) after status epilepticus (SE) termination by diazepam might prevent the development of spontaneous recurrent seizures via the inhibition of neurotoxicity induced by brain edema events. In the present study, we determined the possible molecular and cellular mechanisms of LEV treatment after termination of SE. To assess the effect of LEV against the brain alterations after SE, we focused on blood-brain barrier (BBB) dysfunction associated with angiogenesis and brain inflammation. The consecutive treatment of LEV inhibited the temporarily increased BBB leakage in the hippocampus two days after SE. At the same time point, the LEV treatment significantly inhibited the increase in the number of CD31-positive endothelial immature cells and in the expression of angiogenic factors. These findings suggested that the increase in neovascularization led to an increase in BBB permeability by SE-induced BBB failure, and these brain alterations were prevented by LEV treatment. Furthermore, in the acute phase of the latent period, pro-inflammatory responses for epileptogenic targets in microglia and astrocytes of the hippocampus activated, and these upregulations of pro-inflammatory-related molecules were inhibited by LEV treatment. These findings suggest that LEV is likely involved in neuroprotection via anti-angiogenesis and anti-inflammatory activities against BBB dysfunction in the acute phase of epileptogenesis after SE.
Collapse
Affiliation(s)
- Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan.
| | - Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima 739-8521, Japan
| | - Rie Komori
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Hiromi Nochi
- Laboratory for Pharmaceutical Health Sciences, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Ruri Taniguchi
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima 739-8521, Japan
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Fuyuko Takata-Tsuji
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Yasufumi Kataoka
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| |
Collapse
|
29
|
Ge YX, Tian XZ, Lin YY, Liu XY. Chronic treatment with levetiracetam reverses deficits in hippocampal LTP in vivo in experimental temporal lobe epilepsy rats. Neurosci Lett 2016; 628:194-200. [PMID: 27345386 DOI: 10.1016/j.neulet.2016.06.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/10/2016] [Accepted: 06/21/2016] [Indexed: 12/21/2022]
Abstract
Temporal lobe epilepsy (TLE), the common form of epilepsy in adults, often displays complex partial seizures and cognitive deficits. The underlying mechanisms of such deficits are not yet well understood. Many contributing factors, such as initial epileptogenic lesion, seizure type, age of onset, and treatment side effects have been proposed. Levetiracetam (LEV) is a novel anti-epileptic drug (AED) used to treat partial seizures and idiopathic generalized epilepsy. It has been suggested that LEV exerts antiepileptic properties by modulation of synaptic release of neurotransmitters. However, its neuroprotective effects on learning and memory are not yet well demonstrated. Here we showed the impairment of spatial memory in the pilocarpine-induced experimental TLE rats, which can be improved by LEV. Furthermore, we found chronic LEV treatment partially reversed the SE-induced synaptic dysfunction in hippocampal LTP induction in vivo. In addition, LEV treatment can alleviate the SE-induced abnormal GluR1 phosphorylation at Ser(831) site, which may contribute to the rescue of synaptic transmission. These results indicate the neuroprotective role for LEV while it exhibits an antiseizure effect on experimental epileptic models.
Collapse
Affiliation(s)
- Yu-Xing Ge
- Department of Neurology, Tongji University Affiliated Tenth People's Hospital, 200072 Shanghai, PR China
| | - Xiang-Zhu Tian
- Department of Neurology, Tongji University Affiliated Tenth People's Hospital, 200072 Shanghai, PR China
| | - Ying-Ying Lin
- Department of Neurology, Tongji University Affiliated Tenth People's Hospital, 200072 Shanghai, PR China
| | - Xue-Yuan Liu
- Department of Neurology, Tongji University Affiliated Tenth People's Hospital, 200072 Shanghai, PR China.
| |
Collapse
|
30
|
Takechi K, Suemaru K, Kiyoi T, Tanaka A, Araki H. The α4β2 nicotinic acetylcholine receptor modulates autism-like behavioral and motor abnormalities in pentylenetetrazol-kindled mice. Eur J Pharmacol 2016; 775:57-66. [PMID: 26868186 DOI: 10.1016/j.ejphar.2016.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/20/2016] [Accepted: 02/04/2016] [Indexed: 01/22/2023]
Abstract
Epilepsy is associated with several psychiatric disorders, including cognitive impairment, autism and attention deficit/hyperactivity disorder (ADHD). However, the psychopathology of epilepsy is frequently unrecognized and untreated in patients. In the present study, we investigated the effects of ABT-418, a neuronal nicotinic acetylcholine receptor agonist, on pentylenetetrazol (PTZ)-kindled mice with behavioral and motor abnormalities. PTZ-kindled mice displayed impaired motor coordination (in the rotarod test), anxiety (in the elevated plus maze test) and social approach impairment (in the three-chamber social test) compared with control mice. ABT-418 treatment (0.05 mg/kg, intraperitoneally) alleviated these behavioral abnormalities in PTZ-kindled mice. Immunolabeling of tissue sections demonstrated that expression of the α4 nicotinic acetylcholine receptor subunit in the medial habenula was similar in control and PTZ-kindled mice. However, expression was significantly decreased in the piriform cortex in PTZ-kindled mice. In addition, we examined the expression of the synaptic adhesion molecule neuroligin 3 (NLG3). NLG3 expression in the piriform cortex was significantly higher in PTZ-kindled mice compared with control mice. Collectively, our findings suggest that ADHD-like or autistic-like behavioral abnormalities associated with epilepsy are closely related to the downregulation of the α4 nicotinic receptor and the upregulation of NLG3 in the piriform cortex. In summary, this study indicates that ABT-418 might have therapeutic potential for attentional impairment in epileptic patients with psychiatric disorders such as autism and ADHD.
Collapse
Affiliation(s)
- Kenshi Takechi
- Division of Pharmacy, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Katsuya Suemaru
- School of Pharmacy, Shujitsu University, 1-6-1 Nishikawara, Naka-ku, Okayama 703-8516, Japan
| | - Takeshi Kiyoi
- Integrated Center for Science, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Akihiro Tanaka
- Division of Pharmacy, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Hiroaki Araki
- Division of Pharmacy, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan
| |
Collapse
|
31
|
Navarro V, Dagron C, Elie C, Lamhaut L, Demeret S, Urien S, An K, Bolgert F, Tréluyer JM, Baulac M, Carli P. Prehospital treatment with levetiracetam plus clonazepam or placebo plus clonazepam in status epilepticus (SAMUKeppra): a randomised, double-blind, phase 3 trial. Lancet Neurol 2015; 15:47-55. [PMID: 26627366 DOI: 10.1016/s1474-4422(15)00296-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Generalised convulsive status epilepticus (GCSE) should be treated quickly. Benzodiazepines are the only drug treatment available so far that is effective before admission to hospital. We assessed whether addition of the antiepileptic drug levetiracetam to the benzodiazepine clonazepam would improve prehospital treatment of GCSE. METHODS We did a prehospital, randomised, double-blind, phase 3, placebo-controlled, superiority trial to determine the efficacy of adding intravenous levetiracetam (2.5 g) to clonazepam (1 mg) in treatment of GCSE in 13 emergency medical service centres and 26 hospital departments in France. Randomisation was done at the Paris Descartes Clinical Research Unit with a list of random numbers generated by computer. Adults with convulsions lasting longer than 5 min were randomly assigned (1:1) by prehospital physicians to receive levetiracetam or placebo in combination with clonazepam. All physicians and paramedics were masked to group assignments. If the status epilepticus lasted beyond 5 min after drug injection, a second dose of 1 mg clonazepam was given. The primary outcome was cessation of convulsions within 15 min of drug injection. We analysed the modified intention-to-treat population that had received at least one injection of clonazepam and levetiracetam or placebo, excluding patients without valid consent and those randomised more than once. The trial is registered at EudraCT, number 2007-005782-35. FINDINGS Between July 20, 2009, and Dec 15, 2012, 107 patients were randomly assigned to receive placebo and 96 were assigned to receive levetiracetam. The trial was discontinued on Dec 15, 2012 when interim analysis showed no evidence of a treatment difference, and 68 patients in each group were included in the modified intention-to-treat analysis. Convulsions stopped at 15 min of drug injection in 57 of 68 patients (84%) receiving clonazepam and placebo and in 50 of 68 patients (74%) receiving clonazepam and levetiracetam (percentage difference -10.3%, 95% CI -24.0 to 3.4). Three deaths, 19 of 47 (40 %) serious adverse events, and 90 of 197 (46%) non-serious events were reported in the levetiracetam group, and four deaths, 28 of 47 (60%) serious events, and 107 of 197 (54%) non-serious events were reported in the placebo group. INTERPRETATION The addition of levetiracetam to clonazepam treatment presented no advantage over clonazepam treatment alone in the control of GCSE before admission to hospital. Future prehospital trials could assess the efficacy of clonazepam alone as a first-line treatment in status epilepticus and the efficacy of a second injection of clonazepam with another antiepileptic drug as second-line treatment. FUNDING UCB Pharma.
Collapse
Affiliation(s)
- Vincent Navarro
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Epilepsy Unit, and Brain and Spine Institute, Pitié-Salpêtrière Hospital and Université Pierre et Marie Curie, Paris, France.
| | - Christelle Dagron
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Caroline Elie
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Lionel Lamhaut
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Sophie Demeret
- AP-HP, Pitié-Salpêtrière Hospital, Neurological Intensive Care Unit, Paris, France
| | - Saïk Urien
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Kim An
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | - Francis Bolgert
- AP-HP, Pitié-Salpêtrière Hospital, Neurological Intensive Care Unit, Paris, France
| | - Jean-Marc Tréluyer
- AP-HP, Paris Descartes Clinical Research Unit/Clinical Investigation Centre and Université Paris Descartes, France
| | - Michel Baulac
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Epilepsy Unit, and Brain and Spine Institute, Pitié-Salpêtrière Hospital and Université Pierre et Marie Curie, Paris, France
| | - Pierre Carli
- AP-HP, Necker-Enfants Malades Hospital, SAMU 75, and Université Paris Descartes, Paris, France
| | | |
Collapse
|
32
|
Klee R, Töllner K, Rankovic V, Römermann K, Schidlitzki A, Bankstahl M, Löscher W. Network pharmacology for antiepileptogenesis: Tolerability of multitargeted drug combinations in nonepileptic vs. post-status epilepticus mice. Epilepsy Res 2015; 118:34-48. [PMID: 26600369 DOI: 10.1016/j.eplepsyres.2015.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/31/2015] [Accepted: 11/05/2015] [Indexed: 12/30/2022]
Abstract
Prevention of symptomatic epilepsy ("antiepileptogenesis") in patients at risk is a major unmet clinical need. Several drugs underwent clinical trials for epilepsy prevention, but none of the drugs tested was effective. Similarly, most previous preclinical attempts to develop antiepileptogenic strategies failed. In the majority of studies, drugs were given as monotherapy. However, epilepsy is a complex network phenomenon, so that it is unlikely that a single drug can halt epileptogenesis. We recently proposed multitargeted approaches ("network pharmacology") to interfere with epileptogenesis. One strategy, which, if effective, would allow a relatively rapid translation into the clinic, is developing novel combinations of clinically used drugs with diverse mechanisms that are potentially relevant for antiepileptogenesis. In order to test this strategy preclinically, we developed an algorithm for testing such drug combinations, which was inspired by the established drug development phases in humans. As a first step of this algorithm, tolerability of four rationally chosen, repeatedly administered drug combinations was evaluated by a large test battery in mice: A, levetiracetam and phenobarbital; B, valproate, losartan, and memantine; C, levetiracetam and topiramate; and D, levetiracetam, parecoxib, and anakinra. As in clinical trials, tolerability was separately evaluated before starting efficacy experiments to identify any adverse effects of the combinations that may critically limit the successful translation of preclinical findings to the clinic. Except combination B, all drug cocktails were relatively well tolerated. Based on previous studies, we expected that tolerability would be lower in the latent and chronic phases following status epilepticus in mice, but, except combinations C and D, no significant differences were determined between nonepileptic and post-status epilepticus animals. As a next step, the rationally chosen drug combinations will be evaluated for antiepileptogenic activity in mouse and rat models of symptomatic epilepsy.
Collapse
Affiliation(s)
- Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Vladan Rankovic
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
33
|
Bedner P, Dupper A, Hüttmann K, Müller J, Herde MK, Dublin P, Deshpande T, Schramm J, Häussler U, Haas CA, Henneberger C, Theis M, Steinhäuser C. Astrocyte uncoupling as a cause of human temporal lobe epilepsy. Brain 2015; 138:1208-22. [PMID: 25765328 DOI: 10.1093/brain/awv067] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
Glial cells are now recognized as active communication partners in the central nervous system, and this new perspective has rekindled the question of their role in pathology. In the present study we analysed functional properties of astrocytes in hippocampal specimens from patients with mesial temporal lobe epilepsy without (n = 44) and with sclerosis (n = 75) combining patch clamp recording, K(+) concentration analysis, electroencephalography/video-monitoring, and fate mapping analysis. We found that the hippocampus of patients with mesial temporal lobe epilepsy with sclerosis is completely devoid of bona fide astrocytes and gap junction coupling, whereas coupled astrocytes were abundantly present in non-sclerotic specimens. To decide whether these glial changes represent cause or effect of mesial temporal lobe epilepsy with sclerosis, we developed a mouse model that reproduced key features of human mesial temporal lobe epilepsy with sclerosis. In this model, uncoupling impaired K(+) buffering and temporally preceded apoptotic neuronal death and the generation of spontaneous seizures. Uncoupling was induced through intraperitoneal injection of lipopolysaccharide, prevented in Toll-like receptor4 knockout mice and reproduced in situ through acute cytokine or lipopolysaccharide incubation. Fate mapping confirmed that in the course of mesial temporal lobe epilepsy with sclerosis, astrocytes acquire an atypical functional phenotype and lose coupling. These data suggest that astrocyte dysfunction might be a prime cause of mesial temporal lobe epilepsy with sclerosis and identify novel targets for anti-epileptogenic therapeutic intervention.
Collapse
Affiliation(s)
- Peter Bedner
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Alexander Dupper
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Kerstin Hüttmann
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Julia Müller
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Michel K Herde
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Pavel Dublin
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany Current address: Institute of Neurobiology, University of Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Tushar Deshpande
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Johannes Schramm
- 2 Department of Neurosurgery, Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Ute Häussler
- 3 Experimental Epilepsy Research, Department of Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Carola A Haas
- 3 Experimental Epilepsy Research, Department of Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Christian Henneberger
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany 4 UCL Institute of Neurology, UCL, London WC1N 3BG, UK
| | - Martin Theis
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Christian Steinhäuser
- 1 Institute of Cellular Neurosciences and Medical Faculty, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| |
Collapse
|
34
|
Itoh K, Inamine M, Oshima W, Kotani M, Chiba Y, Ueno M, Ishihara Y. Prevention of status epilepticus-induced brain edema and neuronal cell loss by repeated treatment with high-dose levetiracetam. Brain Res 2015; 1608:225-34. [PMID: 25770058 DOI: 10.1016/j.brainres.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 11/28/2022]
Abstract
The management of status epilepticus (SE) is important to prevent mortality and the development of post-SE symptomatic epilepsy. Acquired epilepsy after an initial brain insult by SE can be experimentally reproduced in the murine model of SE induced by pilocarpine. In the present study, we evaluated the possibility of treatment with a high-dose of levetiracetam in this model. Repeated treatment with high-dose levetiracetam after termination of SE by diazepam significantly prevented the incidence of spontaneous recurrent seizures and mortality for at least 28 days. To determine the brain alterations after SE, magnetic resonance imaging was performed. Both T2-weighted imaging and diffusion-weighted imaging showed changes in the limbic regions. These changes in the limbic regions demonstrated the development of cytotoxic edema three hours after SE, followed by the development of vasogenic edema two days after SE. In the pilocarpine-SE model, the incidence of spontaneous recurrent seizures after SE was strongly associated with neuronal damage within a few hours to days after SE by the development of vasogenic edema via the breakdown of the blood-brain barrier in the limbic regions. High-dose levetiracetam significantly suppressed the parameters in the limbic areas. These data indicate that repeated treatment with high-dose levetiracetam for at least two days after SE termination by diazepam is important for controlling the neuronal damage by preventing brain edema. Therefore, these findings suggest that early treatment with high-dose levetiracetam after SE termination by diazepam may protect against adverse sequelae via the inhibition of neurotoxicity induced by brain edema events.
Collapse
Affiliation(s)
- Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan.
| | - Moriyoshi Inamine
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Wataru Oshima
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Masaharu Kotani
- Department of Molecular and Cellular Biology, Faculty of Pharmaceutical Science, Ohu University, Koriyama, Fukushima 963-8611, Japan
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima 739-8521, Japan
| |
Collapse
|
35
|
Radzik I, Miziak B, Dudka J, Chrościńska-Krawczyk M, Czuczwar SJ. Prospects of epileptogenesis prevention. Pharmacol Rep 2015; 67:663-8. [PMID: 25933984 DOI: 10.1016/j.pharep.2015.01.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 01/13/2023]
Abstract
Epilepsy is a common neurologic disease, affecting about 1-2% of the population. In around 30% of patients with epilepsy, their seizures are not satisfactorily controlled and drug-resistant epilepsy constitutes a real therapeutic challenge. Consequently, there are efforts aimed at the inhibition of epileptogenesis, a process of converting a normal into an epileptic brain. Data on this problem have been mainly obtained in post-status epilepticus rodent models in which spontaneous seizure activity and behavioral disturbances develop over time. Among antiepileptic drugs, diazepam at high dose of 20mg/kg given during status epilepticus, significantly inhibited the development of spontaneous seizures and also, a strong neuroprotective effect was evident. Also gabapentin and valproate (over a period of 40 days) proved effective in the inhibition of spontaneous seizure activity and reduction of behavioral deficit. However, there are also data that valproate (over 28 days) significantly improved the behavioral performance without affecting the occurrence of spontaneous seizures. A number of antiepileptic drugs, carbamazepine, lamotrigine, levetiracetam, phenobarbital, and topiramate were completely ineffective. Among non-antiepileptic drugs, some promise show rapamycin, losartan and combinations of anti-inflammatory drugs, targeting different inflammatory pathways. Inhibition of epileptogenesis may become a valuable therapeutic approach provided that there are reliable markers of this process. Actually, such markers begin to emerge.
Collapse
Affiliation(s)
- Iwona Radzik
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Medical University of Lublin, Lublin, Poland; Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Magdalena Chrościńska-Krawczyk
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Department of Pediatrics, Endocrinology and Neurology, Medical University of Lublin, Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Department of Physiopathology, Institute of Rural Health, Lublin, Poland.
| |
Collapse
|
36
|
Kaminski RM, Rogawski MA, Klitgaard H. The potential of antiseizure drugs and agents that act on novel molecular targets as antiepileptogenic treatments. Neurotherapeutics 2014; 11:385-400. [PMID: 24671870 PMCID: PMC3996125 DOI: 10.1007/s13311-014-0266-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A major goal of contemporary epilepsy research is the identification of therapies to prevent the development of recurrent seizures in individuals at risk, including those with brain injuries, infections, or neoplasms; status epilepticus; cortical dysplasias; or genetic epilepsy susceptibility. In this review we consider the evidence largely from preclinical models for the antiepileptogenic activity of a diverse range of potential therapies, including some marketed antiseizure drugs, as well as agents that act by immune and inflammatory mechanisms; reduction of oxidative stress; activation of the mammalian target of rapamycin or peroxisome proliferator-activated receptors γ pathways; effects on factors related to thrombolysis, hematopoesis, and angiogenesis; inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reducatase; brain-derived neurotrophic factor signaling; and blockade of α2 adrenergic and cannabinoid receptors. Antiepileptogenesis refers to a therapy of which the beneficial action is to reduce seizure frequency or severity outlasting the treatment period. To date, clinical trials have failed to demonstrate that antiseizure drugs have such disease-modifying activity. However, studies in animal models with levetiracetam and ethosuximide are encouraging, and clinical trials with these agents are warranted. Other promising strategies are inhibition of interleukin 1β signaling by drugs such as VX-765; modulation of sphingosine 1-phosphate signaling by drugs such as fingolimod; activation of the mammalian target of rapamycin by drugs such as rapamycin; the hormone erythropoietin; and, paradoxically, drugs such as the α2 adrenergic receptor antagonist atipamezole and the CB1 cannabinoid antagonist SR141716A (rimonabant) with proexcitatory activity. These approaches could lead to a new paradigm in epilepsy drug therapy where treatment for a limited period prevents the occurrence of spontaneous seizures, thus avoiding lifelong commitment to symptomatic treatment.
Collapse
Affiliation(s)
| | - Michael A. Rogawski
- />Department of Neurology, University of California, Davis School of Medicine, Sacramento, CA USA
| | | |
Collapse
|
37
|
Deshpande LS, Delorenzo RJ. Mechanisms of levetiracetam in the control of status epilepticus and epilepsy. Front Neurol 2014; 5:11. [PMID: 24550884 PMCID: PMC3907711 DOI: 10.3389/fneur.2014.00011] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/17/2014] [Indexed: 11/27/2022] Open
Abstract
Status epilepticus (SE) is a major clinical emergency that is associated with high mortality and morbidity. SE causes significant neuronal injury and survivors are at a greater risk of developing acquired epilepsy and other neurological morbidities, including depression and cognitive deficits. Benzodiazepines and some anticonvulsant agents are drugs of choice for initial SE management. Despite their effectiveness, over 40% of SE cases are refractory to the initial treatment with two or more medications. Thus, there is an unmet need of developing newer anti-SE drugs. Levetiracetam (LEV) is a widely prescribed anti-epileptic drug that has been reported to be used in SE cases, especially in benzodiazepine-resistant SE or where phenytoin cannot be used due to allergic side-effects. Levetiracetam’s non-classical anti-epileptic mechanisms of action, favorable pharmacokinetic profile, general lack of central depressant effects, and lower incidence of drug interactions contribute to its use in SE management. This review will focus on LEV’s unique mechanism of action that makes it a viable candidate for SE treatment.
Collapse
Affiliation(s)
| | - Robert J Delorenzo
- Department of Neurology, Virginia Commonwealth University , Richmond, VA , USA ; Department of Pharmacology and Toxicology, Virginia Commonwealth University , Richmond, VA , USA ; Department of Biochemistry, Virginia Commonwealth University , Richmond, VA , USA
| |
Collapse
|
38
|
|
39
|
Shetty AK. Prospects of levetiracetam as a neuroprotective drug against status epilepticus, traumatic brain injury, and stroke. Front Neurol 2013; 4:172. [PMID: 24204362 PMCID: PMC3816384 DOI: 10.3389/fneur.2013.00172] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/21/2013] [Indexed: 01/08/2023] Open
Abstract
Levetiracetam (LEV) is an anti-epileptic drug commonly used for the treatment of partial onset and generalized seizures. In addition to its neuromodulatory and neuroinhibitory effects via its binding to the synaptic vesicle protein SV2A, multiple studies have suggested neuroprotective properties for LEV in both epileptic and non-epileptic conditions. The purpose of this review is to discuss the extent of LEV-mediated protection seen in different neurological conditions, the potential of LEV for easing epileptogenesis, and the possible mechanisms that underlie the protective properties of LEV. LEV has been found to be particularly beneficial for restraining seizures in animal models of spontaneous epilepsy, acute seizures, and status epilepticus (SE). However, its ability for easing epileptogenesis and cognitive dysfunction following SE remains controversial with some studies implying favorable outcomes and others reporting no beneficial effects. Efficacy of LEV as a neuroprotective drug against traumatic brain injury (TBI) has received much attention. While animal studies in TBI models have showed significant neuroprotection and improvements in motor and memory performance with LEV treatment, clinical studies suggest that LEV has similar efficacy as phenytoin in terms of its ability to prevent post-traumatic epilepsy. LEV treatment for TBI is also reported to have fewer adverse effects and monitoring considerations but electroencephalographic recordings suggest the presence of increased seizure tendency. Studies on stroke imply that LEV is a useful alternative to carbamazepine for preventing post-stroke seizures in terms of efficacy and safety. Thus, LEV treatment has promise for restraining SE-, TBI-, or stroke-induced chronic epilepsy. Nevertheless, additional studies are needed to ascertain the most apt dose, timing of intervention, and duration of treatment after the initial precipitating injury and the mechanisms underlying LEV-mediated beneficial effects.
Collapse
Affiliation(s)
- Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White , Temple, TX , USA ; Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System , Temple, TX , USA ; Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine , College Station, TX , USA
| |
Collapse
|
40
|
Faure JB, Akimana G, Carneiro JEM, Cosquer B, Ferrandon A, Geiger K, Koning E, Penazzi L, Cassel JC, Nehlig A. A comprehensive behavioral evaluation in the lithium-pilocarpine model in rats: Effects of carisbamate administration during status epilepticus. Epilepsia 2013; 54:1203-13. [DOI: 10.1111/epi.12219] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Jean-Baptiste Faure
- Faculty of Medicine; INSERM U 666; Strasbourg France
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | - Gladys Akimana
- Faculty of Medicine; INSERM U 666; Strasbourg France
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | - José E. M. Carneiro
- Faculty of Medicine; INSERM U 666; Strasbourg France
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | - Brigitte Cosquer
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | | | - Karin Geiger
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | | | - Lorène Penazzi
- Laboratory of Cognitive and Adaptive Neuroscience; CNRS-UDS UMR 7364; Strasbourg France
| | | | - Astrid Nehlig
- Faculty of Medicine; INSERM U 666; Strasbourg France
| |
Collapse
|
41
|
Kwon YS, Pineda E, Auvin S, Shin D, Mazarati A, Sankar R. Neuroprotective and antiepileptogenic effects of combination of anti-inflammatory drugs in the immature brain. J Neuroinflammation 2013; 10:30. [PMID: 23442201 PMCID: PMC3599749 DOI: 10.1186/1742-2094-10-30] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/15/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Inflammatory signaling elicited by prolonged seizures can be contributory to neuronal injury as well as adverse plasticity leading to the development of spontaneous recurrent seizures (epilepsy) and associated co-morbidities. In this study, developing rat pups were subjected to lithium-pilocarpine status epilepticus (SE) at 2 and 3 weeks of age to study the effect of anti-inflammatory drugs (AID) on SE-induced hippocampal injury and the development of spontaneous seizures. FINDINGS We selected AIDs directed against interleukin-1 receptors (IL-1ra), a cyclooxygenase-2 (COX-2) inhibitor (CAY 10404), and an antagonist of microglia activation of caspase-1 (minocycline). Acute injury after SE was studied in the 2-week-old rats 24 h after SE. Development of recurrent spontaneous seizures was studied in 3-week-old rats subjected to SE 4 months after the initial insult.None of those AIDs were effective in attenuating CA1 injury in the 2-week-old pups or in limiting the development of spontaneous seizures in 3-week-old pups when administered individually. When empiric binary combinations of these drugs were tried, the combined targeting of IL-1r and COX-2 resulted in attenuation of acute CA1 injury, as determined 24 h after SE, in those animals. The same combination administered for 10 days following SE in 3-week-old rats, reduced the development of spontaneous recurrent seizures and limited the extent of mossy fiber sprouting. CONCLUSIONS Deployment of an empirically designed 'drug cocktail' targeting multiple inflammatory signaling pathways for a limited duration after an initial insult like SE may provide a practical approach to neuroprotection and anti-epileptogenic therapy.
Collapse
Affiliation(s)
- Young Se Kwon
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Pediatrics, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Eduardo Pineda
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Stéphane Auvin
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Pediatric Neurology, Hôpital Robert Debré, INSERM U676, Paris, 75019, France
| | - Don Shin
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Andrey Mazarati
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Raman Sankar
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
Low-intensity physical training recovers object recognition memory impairment in rats after early-life induced Status epilepticus. Int J Dev Neurosci 2013; 31:196-201. [PMID: 23318691 DOI: 10.1016/j.ijdevneu.2013.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 01/02/2013] [Accepted: 01/03/2013] [Indexed: 12/18/2022] Open
Abstract
When it occurs early in life, Status epilepticus (SE) can cause behavioural and cognitive impairments in adulthood. Here, we evaluated the putative benefits of low-intensity treadmill training on long-standing cognitive impairment in rats submitted to SE early in life. Wistar rats were submitted to LiCl-pilocarpine-induced SE at P16. Animals from the trained group underwent a low-intensity treadmill protocol for 5 days per week for 4 weeks. At adulthood, rats subjected to early-life SE displayed impairment in long-term memory in an object recognition task, while the training protocol completely reversed this deficit. This result was associated with neither locomotor alterations nor changes in emotional behaviour; there were no differences between groups in the distance travelled, grooming or rearing in the open field test; there were also no differences between groups in the number of risk assessment, time spent in open arms in an elevated plus maze and number of entries into the open arms. These data suggest that physical exercise can ameliorate the long-standing recognition memory deficit induced by early-life SE, suggesting that it may be useful as a putative intervention for patients who suffered SE during infancy.
Collapse
|
43
|
Rattka M, Brandt C, Löscher W. The intrahippocampal kainate model of temporal lobe epilepsy revisited: epileptogenesis, behavioral and cognitive alterations, pharmacological response, and hippoccampal damage in epileptic rats. Epilepsy Res 2012. [PMID: 23196211 DOI: 10.1016/j.eplepsyres.2012.09.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Systemic or intracerebral (e.g., intrahippocampal or intraamygdalar) administration of kainate, a potent neurotoxic analog of glutamate, is widely used to induce status epilepticus (SE) and subsequent development of epilepsy in rats. However, in apparent contrast to systemic administration, following intracerebral injection the proportion of rats that have been observed to generate spontaneous recurrent seizures (SRS) and the frequency of the SRS are comparatively low. More recently, it has been shown that these problems can be resolved by injecting kainate into the dorsal hippocampus of awake rats, thus avoiding the insult-modifying effects of anesthesia, which had often been used for intracerebral injection of this convulsant in previous studies. For further characterization of this model, we injected kainate (0.4 μg) unilaterally into the CA3 of the posterior hippocampus in awake rats, which induced limbic SE (ranging from 4 to 20 h) in all rats without mortality. Repeated video-EEG monitoring (24h/day, 7 days/week) for periods of 1-2.5 weeks from 1 to 8 months after SE demonstrated that 91% of the rats developed epilepsy, and that seizure frequency significantly increased over the course of the disease. Epilepsy was associated with increased behavioral excitability and impaired learning and memory in a water maze, most likely as a result of hippocampal pathology, which was characterized by extensive neuronal loss in CA3 and dentate hilus and dispersion of granule cells in the ipsilateral hippocampus. A drug trial with phenobarbital showed that all epileptic rats used in this trial responded to treatment with suppression of SRS. The data substantiate that intrahippocampal kainate injection in awake rats offers an excellent model of human temporal lobe epilepsy and indicate that this model may have particular advantages for studying mechanisms of injury-induced epilepsy and comorbidities as targets for antiepileptic and antiepileptogenic therapies.
Collapse
Affiliation(s)
- Marta Rattka
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | | | | |
Collapse
|
44
|
Samba Reddy D, Ramanathan G. Finasteride inhibits the disease-modifying activity of progesterone in the hippocampus kindling model of epileptogenesis. Epilepsy Behav 2012; 25:92-7. [PMID: 22835430 PMCID: PMC3444667 DOI: 10.1016/j.yebeh.2012.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/24/2012] [Accepted: 05/26/2012] [Indexed: 11/19/2022]
Abstract
Progesterone (P) plays an important role in seizure susceptibility in women with epilepsy. Preclinical and experimental studies suggest that P appears to interrupt epileptogenesis, which is a process whereby a normal brain becomes progressively susceptible to recurrent, unprovoked seizures due to precipitating risk factors. Progesterone has not been investigated widely for its potential disease-modifying activity in epileptogenic models. Recently, P has been shown to exert disease-modifying effects in the kindling model of epileptogenesis. However, the mechanisms underlying the protective effects of P against epileptogenesis remain unclear. In this study, we investigated the role of P-derived neurosteroids in the disease-modifying activity of P. It is hypothesized that 5α-reductase converts P to allopregnanolone and related neurosteroids that retard epileptogenesis in the brain. To test this hypothesis, we utilized the mouse hippocampus kindling model of epileptogenesis and investigated the effect of finasteride, a 5α-reductase and neurosteroid synthesis inhibitor. Progesterone markedly retarded the development of epileptogenesis and inhibited the rate of kindling acquisition to elicit stage 5 seizures. Pretreatment with finasteride led to complete inhibition of the P-induced retardation of the limbic epileptogenesis in mice. Finasteride did not significantly influence the acute seizure expression in fully kindled mice expressing stage 5 seizures. Thus, neurosteroids that potentiate phasic and tonic inhibition in the hippocampus, such as allopregnanolone, may mediate the disease-modifying effect of P, indicating a new role of neurosteroids in acquired limbic epileptogenesis and temporal lobe epilepsy.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| | | |
Collapse
|
45
|
Positive impact of levetiracetam on emotional learning and memory in naive mice. Life Sci 2012; 90:185-9. [DOI: 10.1016/j.lfs.2011.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 11/01/2011] [Accepted: 11/05/2011] [Indexed: 11/20/2022]
|
46
|
Langer M, Brandt C, Löscher W. Marked strain and substrain differences in induction of status epilepticus and subsequent development of neurodegeneration, epilepsy, and behavioral alterations in rats. Epilepsy Res 2011; 96:207-24. [DOI: 10.1016/j.eplepsyres.2011.06.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 05/30/2011] [Accepted: 06/04/2011] [Indexed: 10/18/2022]
|
47
|
Russo E, Citraro R, Scicchitano F, De Fazio S, Perrota I, Di Paola ED, Constanti A, De Sarro G. Effects of early long-term treatment with antiepileptic drugs on development of seizures and depressive-like behavior in a rat genetic absence epilepsy model. Epilepsia 2011; 52:1341-50. [DOI: 10.1111/j.1528-1167.2011.03112.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Gajda Z, Török R, Horváth Z, Szántai-Kis C, Orfi L, Kéri G, Szente M. Protein kinase inhibitor as a potential candidate for epilepsy treatment. Epilepsia 2011; 52:579-88. [PMID: 21371019 DOI: 10.1111/j.1528-1167.2011.02979.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Effects of the "VID-82925" kinase inhibitor molecule were investigated both during the developing phase as well as during the stable phase of the focus with spontaneous recurrent seizures using the 4-AP-induced in vivo epilepsy model in anesthetized rats. METHODS In electrophysiologic experiments, VID-82925 (0.85 mg/kg) was injected intravenously either before the induction (pretreatment) or after the development of the stable focus (treatment). Reference drugs carbamazepine (4.8 mg/kg) and levetiracetam (50 mg/kg) were employed using the same experimental paradigm. The antiepileptic effect of VID-82925 was also compared to those of the broad-spectrum gap junction channel blocker carbenoxolone (10 mm). KEY FINDINGS Pretreatment with VID-82925 revealed an antiepileptogenic effect as it suppressed significantly the manifestation of the epileptiform activity not only during the developing phase, but also for a considerable long period during the stable phase of the focus. The current data do not allow us to differentiate an antiictal treatment effect from an antiepileptogenic effect of the compound during the stable phase of the focus. Treatment with VID-82925 was also effective against ictogenesis during the stable phase of the focus. Pretreatment with levetiracetam failed to exert any antiepileptogenic effect. The antiepileptic effects of VID-82925 and of the reference drugs on the epileptiform activity of the stable focus were comparable in intensity; however, the effect of VID-82925 was 2-3 times longer. The effects of VID-82925 and of carbenoxolone overlapped one another to some extent, suggesting that VID-82925 may exert its effects at least partially through blocking of gap junctional communication. SIGNIFICANCE Our results indicate that inhibition of protein kinases may also provide an effective strategy for the development of a drug that is not only antiepileptic but also depresses the course of epileptogenesis.
Collapse
Affiliation(s)
- Zita Gajda
- Department of Comparative Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Prevention of epileptogenesis after brain trauma is an unmet medical challenge. Recent molecular profiling studies have provided an insight into molecular changes that contribute to formation of ictogenic neuronal networks, including genes regulating synaptic or neuronal plasticity, cell death, proliferation, and inflammatory or immune responses. These mechanisms have been targeted to prevent epileptogenesis in animal models. Favourable effects have been obtained using immunosuppressants, antibodies blocking adhesion of leucocytes to endothelial cells, gene therapy driving expression of neurotrophic factors, pharmacological neurostimulation, or even with conventional antiepileptic drugs by administering them before the appearance of genetic epilepsy. Further studies are needed to clarify the optimum time window and aetiological specificity of treatments. Questions related to adverse events also need further consideration. Encouragingly, the recent experimental studies emphasise that the complicated process of epileptogenesis can be favourably modified, and that antiepileptogenesis as a treatment indication might not be an impossible mission.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, A I Virtanen Institute for Molecular Sciences, University of Eastern Finland, and Department of Neurology, Kuopio University Hospital, Kuopio, Finland.
| | | |
Collapse
|
50
|
Löscher W, Brandt C. Prevention or modification of epileptogenesis after brain insults: experimental approaches and translational research. Pharmacol Rev 2010; 62:668-700. [PMID: 21079040 PMCID: PMC3014230 DOI: 10.1124/pr.110.003046] [Citation(s) in RCA: 305] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Diverse brain insults, including traumatic brain injury, stroke, infections, tumors, neurodegenerative diseases, and prolonged acute symptomatic seizures, such as complex febrile seizures or status epilepticus (SE), can induce "epileptogenesis," a process by which normal brain tissue is transformed into tissue capable of generating spontaneous recurrent seizures. Furthermore, epileptogenesis operates in cryptogenic causes of epilepsy. In view of the accumulating information about cellular and molecular mechanisms of epileptogenesis, it should be possible to intervene in this process before the onset of seizures and thereby either prevent the development of epilepsy in patients at risk or increase the potential for better long-term outcome, which constitutes a major clinical need. For identifying pharmacological interventions that prevent, interrupt or reverse the epileptogenic process in people at risk, two groups of animal models, kindling and SE-induced recurrent seizures, have been recommended as potentially useful tools. Furthermore, genetic rodent models of epileptogenesis are increasingly used in assessing antiepileptogenic treatments. Two approaches have been used in these different model categories: screening of clinically established antiepileptic drugs (AEDs) for antiepileptogenic or disease-modifying potential, and targeting the key causal mechanisms that underlie epileptogenesis. The first approach indicated that among various AEDs, topiramate, levetiracetam, carisbamate, and valproate may be the most promising. On the basis of these experimental findings, two ongoing clinical trials will address the antiepileptogenic potential of topiramate and levetiracetam in patients with traumatic brain injury, hopefully translating laboratory discoveries into successful therapies. The second approach has highlighted neurodegeneration, inflammation and up-regulation of immune responses, and neuronal hyperexcitability as potential targets for antiepileptogenesis or disease modification. This article reviews these areas of progress and discusses the challenges associated with discovery of antiepileptogenic therapies.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Bünteweg 17, Hannover, Germany.
| | | |
Collapse
|