1
|
Dell'Osso L, Nardi B, Massoni L, Gravina D, Benedetti F, Cremone IM, Carpita B. Neuroprotective Properties of Antiepileptics: What are the Implications for Psychiatric Disorders? Curr Med Chem 2024; 31:3447-3472. [PMID: 37226791 DOI: 10.2174/0929867330666230523155728] [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: 12/31/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/26/2023]
Abstract
Since the discovery of the first antiepileptic compound, increasing attention has been paid to antiepileptic drugs (AEDs), and recently, with the understanding of the molecular mechanism underlying cells death, a new interest has revolved around a potential neuroprotective effect of AEDs. While many neurobiological studies in this field have focused on the protection of neurons, growing data are reporting how exposure to AEDs can also affect glial cells and the plastic response underlying recovery; however, demonstrating the neuroprotective abilities of AEDs remains a changeling task. The present work aims to summarize and review the literature available on the neuroprotective properties of the most commonly used AEDs. Results highlighted how further studies should investigate the link between AEDs and neuroprotective properties; while many studies are available on valproate, results for other AEDs are very limited and the majority of the research has been carried out on animal models. Moreover, a better understanding of the biological basis underlying neuro-regenerative defects may pave the way for the investigation of further therapeutic targets and eventually lead to an improvement in the actual treatment strategies.
Collapse
Affiliation(s)
- Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Benedetta Nardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Leonardo Massoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Davide Gravina
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Francesca Benedetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Ivan Mirko Cremone
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Barbara Carpita
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| |
Collapse
|
2
|
Ouédraogo O, Balthazard R, Mamane VH, Jamann H, Millette F, Daigneault A, Arbour N, Larochelle C. Investigating anti-inflammatory and immunomodulatory properties of brivaracetam and lacosamide in experimental autoimmune encephalomyelitis (EAE). Epilepsy Res 2023; 192:107125. [PMID: 36963302 DOI: 10.1016/j.eplepsyres.2023.107125] [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: 12/24/2022] [Revised: 02/20/2023] [Accepted: 03/13/2023] [Indexed: 03/15/2023]
Abstract
PURPOSE Inflammation plays a role in drug-resistant epilepsy (DRE). We have previously reported an increased proportion of CD4 T cells displaying a pro-inflammatory profile in the peripheral blood of adults with DRE. Specific anti-epileptic drugs (AEDs) exhibit immunomodulatory properties that could increase the risk of infections but also contribute to their beneficial impact on DRE and other neurological diseases. The impact of novel generation AEDs on the profile of immune cells and on neuroinflammatory processes remains unclear. METHODS We compared the influence of brivaracetam and lacosamide on the activation of human and murine peripheral immune cells in vitro and in vivo in active experimental autoimmune encephalomyelitis (EAE), a common mouse model of central nervous system inflammation. RESULTS We found that brivaracetam and lacosamide at 2.5 μg/ml did not impair the survival and activation of human immune cells, but a higher dose of 25 μg/ml decreased mitogen-induced proliferation of CD8 T cells in vitro. Exposure to high doses of brivaracetam, and to a lesser extent lacosamide, reduced the proportion of CD25+ and CD107a+ CD8+ human T cells in vitro, and the frequency of CNS-infiltrating CD8+ T cells at EAE onset and CD11b+ myeloid cells at peak in vivo. Prophylactic administration of brivaracetam or lacosamide did not delay EAE onset but significantly improved the clinical course in the chronic phase of EAE compared to control. CONCLUSION Novel generation AEDs do not impair the response to immunization with MOG peptide but improve the course of EAE, possibly through a reduction of neuroaxonal damage.
Collapse
Affiliation(s)
- Oumarou Ouédraogo
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Renaud Balthazard
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Victoria Hannah Mamane
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Hélène Jamann
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Florence Millette
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Audrey Daigneault
- Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada
| | - Catherine Larochelle
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada; Research Center of the University of Montreal Hospital Center, Montreal, QC, Canada.
| |
Collapse
|
3
|
Riikonen R. Biochemical mechanisms in pathogenesis of infantile epileptic spasm syndrome. Seizure 2023; 105:1-9. [PMID: 36634586 DOI: 10.1016/j.seizure.2023.01.004] [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: 09/22/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanisms leading to infantile epileptic spasm syndrome (IESS) remain obscure. The only common factor seems to be that the spasms are restricted to a limited period of infancy, during a certain maturational state. Here the current literature regarding the biochemical mechanisms of brain maturation in IESS is reviewed, and various hypotheses of the pathophysiology are put together. They include: (1) imbalance of inhibitory (NGF, IGF-1, ACTH, GABA) and excitatory factors (glutamate, nitrites) which distinguishes the different etiological subgroups, (2) abnormality of the hypothalamic pituitary adrenal (HPA) axis linking insults and early life stress, (3) inflammation (4) yet poorly known genetic and epigenetic factors, and (5) glucocorticoid and vigabatrin action on brain development, pinpointing at molecular targets of the pathophysiology from another angle. An altered maturational process may explain why so many, seemingly independent etiological factors lead to the same clinical syndrome and frequently to developmental delay. Understanding these factors can provide ideas for novel therapies.
Collapse
Affiliation(s)
- Raili Riikonen
- Children's Hospital, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
4
|
Zagaja M, Zagaja A, Szala-Rycaj J, Szewczyk A, Lemieszek MK, Raszewski G, Andres-Mach M. Influence of Umbelliferone on the Anticonvulsant and Neuroprotective Activity of Selected Antiepileptic Drugs: An In Vivo and In Vitro Study. Int J Mol Sci 2022; 23:ijms23073492. [PMID: 35408852 PMCID: PMC8999126 DOI: 10.3390/ijms23073492] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Umbelliferone (7-hydroxycoumarin; UMB) is a coumarin with many biological properties, including antiepileptic activity. This study evaluated the effect of UMB on the ability of classical and novel antiepileptic drugs (e.g., lacosamide (LCM), levetiracetam (LEV), phenobarbital (PB) and valproate (VPA)) to prevent seizures evoked by the 6-Hz corneal-stimulation-induced seizure model. The study also evaluated the influence of this coumarin on the neuroprotective properties of these drugs in two in vitro models of neurodegeneration, including trophic stress and excitotoxicity. The results indicate that UMB (100 mg/kg, i.p.) significantly enhanced the anticonvulsant action of PB (p < 0.01) and VPA (p < 0.05), but not that of LCM orLEV, in the 6-Hz test. Whether alone or in combination with other anticonvulsant drugs (at their ED50 values from the 6-Hz test), UMB (100 mg/kg) did not affect motor coordination; skeletal muscular strength and long-term memory, as determined in the chimney; grip strength; or passive avoidance tests, respectively. Pharmacokinetic characterization revealed that UMB had no impact on total brain concentrations of PB or VPA in mice. The in vitro study indicated that UMB has neuroprotective properties. Administration of UMB (1 µg/mL), together with antiepileptic drugs, mitigated their negative impact on neuronal viability. Under trophic stress (serum deprivation) conditions, UMB enhanced the neurotrophic abilities of all the drugs used. Moreover, this coumarin statistically enhanced the neuroprotective effects of PB (p < 0.05) and VPA (p < 0.001) in the excitotoxicity model of neurodegeneration. The obtained results clearly indicate a positive effect of UMB on the anticonvulsant and neuroprotective properties of the selected drugs.
Collapse
Affiliation(s)
- Mirosław Zagaja
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
- Correspondence: (M.Z.); (A.Z.); Tel.: +48-81-718-4549 (M.Z.); +48-81-448-6850 (A.Z.)
| | - Anna Zagaja
- Chair and Department of Humanities and Social Medicine, Medical University of Lublin, Chodźki 7, 20-090 Lublin, Poland
- Correspondence: (M.Z.); (A.Z.); Tel.: +48-81-718-4549 (M.Z.); +48-81-448-6850 (A.Z.)
| | - Joanna Szala-Rycaj
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| | - Aleksandra Szewczyk
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| | - Marta Kinga Lemieszek
- Department of Medical Biology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland;
| | - Grzegorz Raszewski
- Department of Toxicology and Food Protection, Institute of Rural Health, Jaczewskiego 2, 20-950 Lublin, Poland;
| | - Marta Andres-Mach
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| |
Collapse
|
5
|
Löscher W, Klein P. New approaches for developing multi-targeted drug combinations for disease modification of complex brain disorders. Does epilepsy prevention become a realistic goal? Pharmacol Ther 2021; 229:107934. [PMID: 34216705 DOI: 10.1016/j.pharmthera.2021.107934] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
Over decades, the prevailing standard in drug discovery was the concept of designing highly selective compounds that act on individual drug targets. However, more recently, multi-target and combinatorial drug therapies have become an important treatment modality in complex diseases, including neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The development of such network-based approaches is facilitated by the significant advance in our understanding of the pathophysiological processes in these and other complex brain diseases and the adoption of modern computational approaches in drug discovery and repurposing. However, although drug combination therapy has become an effective means for the symptomatic treatment of many complex diseases, the holy grail of identifying clinically effective disease-modifying treatments for neurodegenerative and other brain diseases remains elusive. Thus, despite extensive research, there remains an urgent need for novel treatments that will modify the progression of the disease or prevent its development in patients at risk. Here we discuss recent approaches with a focus on multi-targeted drug combinations for prevention or modification of epilepsy. Over the last ~10 years, several novel promising multi-targeted therapeutic approaches have been identified in animal models. We envision that synergistic combinations of repurposed drugs as presented in this review will be demonstrated to prevent epilepsy in patients at risk within the next 5-10 years.
Collapse
Affiliation(s)
- Wolfgang Löscher
- 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
| |
Collapse
|
6
|
C-11, a New Antiepileptic Drug Candidate: Evaluation of the Physicochemical Properties and Impact on the Protective Action of Selected Antiepileptic Drugs in the Mouse Maximal Electroshock-Induced Seizure Model. Molecules 2021; 26:molecules26113144. [PMID: 34074008 PMCID: PMC8197310 DOI: 10.3390/molecules26113144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/17/2022] Open
Abstract
C-11 is a hybrid compound derived from 2-(2,5-dioxopyrrolidin-1-yl) propanamide, with a wide spectrum of anticonvulsant activity and low neurotoxicity. The aim of this study was to determine the effects of C-11 on the protective action of various antiepileptic drugs (i.e., carbamazepine CBZ, lacosamide LCM, lamotrigine LTG, and valproate VPA) against maximal electroshock-induced seizures (MES) in mice, as well as its neuroprotective and physicochemical/pharmacokinetic properties. Results indicate that C-11 (30 mg/kg, i.p.) significantly enhanced the anticonvulsant action of LCM (p < 0.001) and VPA (p < 0.05) but not that of CBZ and LTG in the MES test. Neither C-11 (30 mg/kg) alone nor its combination with other anticonvulsant drugs (at their ED50 values from the MES test) affected motor coordination; skeletal muscular strength and long-term memory, as determined in the chimney; grip strength and passive avoidance tests, respectively. Pharmacokinetic characterization revealed that C-11 had no impact on total brain concentrations of LCM or VPA in mice. Qualitative analysis of neuroprotective properties of C-11, after a single administration of pilocarpine, revealed no protective effect of this substance in the tested animals. Determination of physicochemical descriptors showed that C-11 meets the drug-likeness requirements resulting from Lipinski and Veber’s rules and prediction of gastrointestinal absorption and brain penetration, which is extremely important for the CNS-active compounds.
Collapse
|
7
|
Martin-Batista E, Maglio LE, Armas-Capote N, Hernández G, Alvarez de la Rosa D, Giraldez T. SGK1.1 limits brain damage after status epilepticus through M current-dependent and independent mechanisms. Neurobiol Dis 2021; 153:105317. [PMID: 33639207 DOI: 10.1016/j.nbd.2021.105317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022] Open
Abstract
Epilepsy is a neurological condition associated to significant brain damage produced by status epilepticus (SE) including neurodegeneration, gliosis and ectopic neurogenesis. Reduction of these processes constitutes a useful strategy to improve recovery and ameliorate negative outcomes after an initial insult. SGK1.1, the neuronal isoform of the serum and glucocorticoids-regulated kinase 1 (SGK1), has been shown to increase M-current density in neurons, leading to reduced excitability and protection against seizures. For this study, we used 4-5 months old male transgenic C57BL/6 J and FVB/NJ mice expressing near physiological levels of a constitutively active form of the kinase controlled by its endogenous promoter. Here we show that SGK1.1 activation potently reduces levels of neuronal death (assessed using Fluoro-Jade C staining) and reactive glial activation (reported by GFAP and Iba-1 markers) in limbic regions and cortex, 72 h after SE induced by kainate, even in the context of high seizure activity. This neuroprotective effect is not exclusively through M-current activation but is also directly linked to decreased apoptosis levels assessed by TUNEL assays and quantification of Bim and Bcl-xL by western blot of hippocampal protein extracts. Our results demonstrate that this newly described antiapoptotic role of SGK1.1 activation acts synergistically with the regulation of cellular excitability, resulting in a significant reduction of SE-induced brain damage in areas relevant to epileptogenesis.
Collapse
Affiliation(s)
- Elva Martin-Batista
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| | - Laura E Maglio
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| | - Natalia Armas-Capote
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| | - Guadalberto Hernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| | - Teresa Giraldez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Campus de Ciencias de la Salud sn, 38200 San Cristobal de La Laguna, Spain; Instituto de Tecnologías Biomédicas (ITB), Campus de Ciencias de la Salud sn, 38071 San Cristobal de La Laguna, Spain.
| |
Collapse
|
8
|
Wiciński M, Puk O, Malinowski B. Cenobamate: Neuroprotective Potential of a New Antiepileptic Drug. Neurochem Res 2020; 46:439-446. [PMID: 33252771 DOI: 10.1007/s11064-020-03188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/31/2022]
Abstract
Central nervous system (CNS) injuries annually afflict approximately 2.7 million people in United States only, inflicting costs of nearly 100 billion US dollars. The gravity of this problem is a consequence of severe and prolonged disability of patients due to a scarce regeneration of CNS, along with the lack of efficient neuroprotective and neuroregenrative therapies. Therefore, the first and most important task in managing the CNS injury is reduction of the damaged area, and apoptosis of neurons occurs not only during the trauma, but in great extent within the following minutes and hours. This process, called secondary injury phase, is a result of trauma-induced metabolic changes in nervous tissue and neuron apoptosis. Cenobamate is a new antiepileptic drug approved by FDA on November 21, 2019. Regardless of its primary purpose, cenobamate, as a blocker of voltage-gated sodium channels and positive modulator of GABAa receptors, it appears to be a promising neuroprotective agent. Moreover, through activation of PI3K/Akt-CREB-BDNF pathway, it leads to the increase of anti-apoptotic factor levels and the decrease of pro-apoptotic factor levels, which induce inhibition of apoptosis and increase neuron survival. Similarly to riluzole, cenobamate could be an important part of a perioperative procedure in neurosurgery, decreasing the occurrence of neurological deficits. Provided that cenobamate will be effective in aforementioned conditions, it could improve treatment outcomes of millions of patients every year, thereby an extensive investigation of its efficacy as a neuroprotective treatment after central nervous system trauma should follow.
Collapse
Affiliation(s)
- Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland
| | - Oskar Puk
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland.
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland
| |
Collapse
|
9
|
Role of Innate Immune Receptor TLR4 and its endogenous ligands in epileptogenesis. Pharmacol Res 2020; 160:105172. [PMID: 32871246 DOI: 10.1016/j.phrs.2020.105172] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/22/2022]
Abstract
Understanding the interplay between the innate immune system, neuroinflammation, and epilepsy might offer a novel perspective in the quest of exploring new treatment strategies. Due to the complex pathology underlying epileptogenesis, no disease-modifying treatment is currently available that might prevent epilepsy after a plausible epileptogenic insult despite the advances in pre-clinical and clinical research. Neuroinflammation underlies the etiopathogenesis of epilepsy and convulsive disorders with Toll-like receptor (TLR) signal transduction being highly involved. Among TLR family members, TLR4 is an innate immune system receptor and lipopolysaccharide (LPS) sensor that has been reported to contribute to epileptogenesis by regulating neuronal excitability. Herein, we discuss available evidence on the role of TLR4 and its endogenous ligands, the high mobility group box 1 (HMGB1) protein, the heat shock proteins (HSPs) and the myeloid related protein 8 (MRP8), in epileptogenesis and post-traumatic epilepsy (PTE). Moreover, we provide an account of the promising findings of TLR4 modulation/inhibition in experimental animal models with therapeutic impact on seizures.
Collapse
|
10
|
Tedrus GMAS, Passos MLGA, Vargas LM, Menezes LEFJ. Cognition and epilepsy: Cognitive screening test. Dement Neuropsychol 2020; 14:186-193. [PMID: 32595889 PMCID: PMC7304275 DOI: 10.1590/1980-57642020dn14-020013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cognitive deficits often occur in people with epilepsy (PWE). However, in Brazil, PWE might not undergo neurocognitive evaluation due to the low number of validated tests available and lack of multidisciplinary teams in general epilepsy outpatient clinics.
Collapse
Affiliation(s)
| | | | - Letícia Muniz Vargas
- Undergraduate Student - Faculty of Medicine, Pontifical Catholic University of Campinas, Campinas, SP, Brazil
| | | |
Collapse
|
11
|
Kern DM, Cepeda MS, Lovestone S, Seabrook GR. Aiding the discovery of new treatments for dementia by uncovering unknown benefits of existing medications. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:862-870. [PMID: 31872043 PMCID: PMC6909196 DOI: 10.1016/j.trci.2019.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction There is a significant need for disease-modifying therapies to treat and prevent dementia, including Alzheimer's disease. Availability of real-world observational information and new analytic techniques to analyze large volumes of data can provide a path to aid drug discovery. Methods Using a self-controlled study design, we examined the association between 2181 medications and incidence of dementia across four US insurance claims databases. Medications associated with ≥50% reduction in risk of dementia in ≥2 databases were examined. Results A total of 117,015,066 individuals were included in the analysis. Seventeen medications met our threshold criteria for a potential protective effect on dementia and fell into five classes: catecholamine modulators, anticonvulsants, antibiotics/antivirals, anticoagulants, and a miscellaneous group. Discussion The biological pathways of the medications identified in this analysis may be targets for further research and may aid in discovering novel therapeutic approaches to treat dementia. These data show association not causality.
Collapse
Affiliation(s)
- David M Kern
- Janssen Research & Development, Epidemiology, Titusville, NJ, USA
| | - M Soledad Cepeda
- Janssen Research & Development, Epidemiology, Titusville, NJ, USA
| | - Simon Lovestone
- Janssen Research & Development, Neuroscience, Beerse, Belgium
| | - Guy R Seabrook
- Johnson & Johnson, Scientific Innovation, South San Francisco, CA, USA
| |
Collapse
|
12
|
Kubová H, Folbergrová J, Rejchrtová J, Tsenov G, Pařízková M, Burchfiel J, Mikulecká A, Mareš P. The Free Radical Scavenger N-Tert-Butyl-α-Phenylnitrone (PBN) Administered to Immature Rats During Status Epilepticus Alters Neurogenesis and Has Variable Effects, Both Beneficial and Detrimental, on Long-Term Outcomes. Front Cell Neurosci 2018; 12:266. [PMID: 30210297 PMCID: PMC6121067 DOI: 10.3389/fncel.2018.00266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Status epilepticus (SE), especially in immature animals, is known to produce recurrent spontaneous seizures and behavioral comorbidities later in life. The cause of these adverse long-term outcomes is unknown, but it has been hypothesized that free radicals produced by SE may play a role. We tested this hypothesis by treating immature (P25) rats with the free radical scavenger N-tert-butyl-α-phenylnitrone (PBN) at the time of lithium chloride (LiCl)/pilocarpine (PILO)-induced SE. Later, long-term outcomes were assessed. Cognitive impairment (spatial memory) was tested in the Morris water maze (MWM). Emotional disturbances were assessed by the capture test (aggressiveness) and elevated plus maze's (EPM) test (anxiety). Next, the presence and severity of spontaneous seizures were assessed by continuous video/EEG monitoring for 5 days. Finally, immunochemistry, stereology and morphology were used to assess the effects of PBN on hippocampal neuropathology and neurogenesis. PBN treatment modified the long-term effects of SE in varying ways, some beneficial and some detrimental. Beneficially, PBN protected against severe anatomical damage in the hippocampus and associated spatial memory impairment. Detrimentally, PBN treated animals had more severe seizures later in life. PBN also made animals more aggressive and more anxious. Correlating with these detrimental long-term outcomes, PBN significantly modified post-natal neurogenesis. Treated animals had significantly increased numbers of mature granule cells (GCs) ectopically located in the dentate hilus (DH). These results raise the possibility that abnormal neurogenesis may significantly contribute to the development of post-SE epilepsy and behavioral comorbidities.
Collapse
Affiliation(s)
- Hana Kubová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jaroslava Folbergrová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Rejchrtová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Grygoriy Tsenov
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martina Pařízková
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - James Burchfiel
- Strong Epilepsy Center, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Anna Mikulecká
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Pavel Mareš
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Helmstaedter C, Beghi E, Elger CE, Kälviäinen R, Malmgren K, May TW, Perucca E, Trinka E, Witt JA. No Evidence of a Causal Role of Antiepileptic Drug Treatment with Regard to the Development of Dementia. J Am Geriatr Soc 2018; 66:1850-1852. [PMID: 30094813 DOI: 10.1111/jgs.15467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 11/28/2022]
Affiliation(s)
| | - Ettore Beghi
- Department of Neuroscience, IRCCS Istituto Mario Negri, Milano, Italy
| | - Christian E Elger
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Reetta Kälviäinen
- Kuopio Epilepsy Center, Neurocenter, Kuopio University Hospital, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kristina Malmgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Neuro Healthcare, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Theodor W May
- Society for Epilepsy Research e.V., Epilepsy Center Bethel, Bielefeld, Germany
| | - Emilio Perucca
- Division of Clinical and Experimental Pharmacology, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.,Clinical Trial Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Eugen Trinka
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, Salzburg, Austria.,Centre of Neuroscience, Christian Doppler University Hospital, Salzburg, Austria.,Department of Public Health and Health Technology Assessment, University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Juri-Alexander Witt
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
14
|
Helmstaedter C, Beghi E, Elger CE, Kälviäinen R, Malmgren K, May TW, Perucca E, Trinka E, Witt JA. No proof of a causal relationship between antiepileptic drug treatment and incidence of dementia. Comment on: Use of antiepileptic drugs and dementia risk-An analysis of Finnish health register and German health insurance data. Epilepsia 2018; 59:1303-1306. [PMID: 29806877 DOI: 10.1111/epi.14432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2018] [Indexed: 11/27/2022]
Affiliation(s)
| | - Ettore Beghi
- Department of Neuroscience, IRCCS Istituto Mario Negri, Milan, Italy
| | - Christian E Elger
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Reetta Kälviäinen
- Kuopio Epilepsy Center, Neurocenter, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kristina Malmgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Neuro Healthcare, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Theodor W May
- Society for Epilepsy Research, Epilepsy Center Bethel Bielefeld, Bielefeld, Germany
| | - Emilio Perucca
- Division of Clinical and Experimental Pharmacology, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.,Clinical Trial Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Eugen Trinka
- Department of Neurology, Christian Doppler University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria.,Center of Neuroscience, Christian Doppler University Hospital Salzburg, Salzburg, Austria.,Department of Public Health and Health Technology Assessment, UMIT-University for Health Sciences, Medical Informatics, and Technology, Hall in Tirol, Austria
| | - Juri-Alexander Witt
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
15
|
Mazhar F, Malhi SM, Simjee SU. Comparative studies on the effects of clinically used anticonvulsants on the oxidative stress biomarkers in pentylenetetrazole-induced kindling model of epileptogenesis in mice. J Basic Clin Physiol Pharmacol 2017; 28:31-42. [PMID: 27658141 DOI: 10.1515/jbcpp-2016-0034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Oxidative stress plays a key role in the pathogenesis of epilepsy and contributes in underlying epileptogenesis process. Anticonvulsant drugs targeting the oxidative stress domain of epileptogenesis may provide better control of seizure. The present study was carried out to investigate the effect of clinically used anti-epileptic drugs (AEDs) on the course of pentylenetetrazole (PTZ)-induced kindling and oxidative stress markers in mice. METHODS Six mechanistically heterogeneous anticonvulsants: phenobarbital, phenytoin, levetiracetam, pregabalin, topiramate, and felbamate were selected and their redox profiles were determined. Diazepam was used as a drug control for comparison. Kindling was induced by repeated injections of a sub-convulsive dose of PTZ (50 mg/kg, s.c.) on alternate days until seizure score 5 was evoked in the control kindled group. Anticonvulsants were administered daily. Following PTZ kindling, oxidative stress biomarkers were assessed in homogenized whole brain samples and estimated for the levels of nitric oxide, peroxide, malondialdehyde, protein carbonyl, reduced glutathione, and activities of nitric oxide synthase and superoxide dismutase. RESULTS Biochemical analysis revealed a significant increase in the levels of reactive oxygen species with a parallel decrease in endogenous anti-oxidants in PTZ-kindled control animals. Daily treatment with levetiracetam and felbamate significantly decreased the PTZ-induced seizure score as well as the levels of nitric oxide (p<0.001), nitric oxide synthase activity (p<0.05), peroxide levels (p<0.05), and malondialdehyde (p<0.05). Levetiracetam and felbamate significantly decreased lipid and protein peroxidation whereas topiramate was found to reduce lipid peroxidation only. CONCLUSIONS An AED that produces anticonvulsant effect by the diversified mechanism of action such as levetiracetam, felbamate, and topiramate exhibited superior anti-oxidative stress activity in addition to their anticonvulsant activity.
Collapse
|
16
|
Differential effects of lacosamide, phenytoin and topiramate on peripheral nerve excitability: An ex vivo electrophysiological study. Neurotoxicology 2015; 52:57-63. [PMID: 26542247 DOI: 10.1016/j.neuro.2015.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/10/2015] [Accepted: 10/29/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Antiepileptic drugs (AEDs) are mainly used to control cortical hyperexcitability. Some of them (e.g. phenytoin (PHT) and topiramate (TPM)) have also effects on the peripheral nervous system (PNS). Lacosamide (LCM) is a novel AED that stabilizes hyperexcitable neuronal membranes by selectively enhancing the slow inactivation of voltage-gated sodium channels (VGSCs). Although the mechanism of action of LCM is fairly well understood, there are no in vitro data available regarding any possible PNS effects of LCM. OBJECTIVE To investigate, in vitro, the effects of LCM on peripheral nerve excitability in comparison with PHT and TPM, two AEDs that act, in part, by stabilizing the fast inactivation state of VGSCs. METHODS Experiments were conducted on the isolated sciatic nerve of the adult rat using standard electrophysiological methods. The effects of LCM on the amplitude and latency of the evoked compound action potential (CAP) during a 48h period of drug exposure were recorded and compared with the effects of PHT and TPM. RESULTS LCM produced inhibitory effects on CAP at concentrations significantly higher than the therapeutic levels (>25μg/ml). At these concentrations (62.57-125.15μg/ml), an acute and immediate increment of the latency and decrement of the amplitude of the CAP were observed. In contrast to LCM, PHT caused an acute decrement in the amplitude as well as an increment in the latency of the CAP even at subtherapeutic levels (5μg/ml). With regard to TPM, the amplitude of the CAP was not affected at the supratherapeutic concentrations but at the therapeutic concentration of 33.94μg/ml a reduced decrement of the CAP amplitude compared to the controls was observed. CONCLUSIONS LCM, PHT and TPM exert differential effects on peripheral nerve excitability. PHT inhibited the sciatic nerve CAP even at subtherapeutic levels whereas LCM was safe within the therapeutic concentration range. TPM did not affect the CAP amplitude even at high supratherapeutic concentrations whereas in the therapeutic range a neuroprotective effect was observed. Possible underlying mechanisms and the clinical implications of these findings are discussed.
Collapse
|
17
|
Abstract
Epileptogenesis is a chronic process that can be triggered by genetic or acquired factors, and that can continue long after epilepsy diagnosis. In 2015, epileptogenesis is not a treatment indication, and there are no therapies available in clinic to treat individuals at risk of epileptogenesis. However, thanks to active research, a large number of animal models have become available for search of molecular mechanisms of epileptogenesis. The first glimpses of treatment targets and biomarkers that could be developed to become useful in clinic are in sight. However, the heterogeneity of the epilepsy condition, and the dynamics of molecular changes over the course of epileptogenesis remain as challenges to overcome.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland Department of Neurology, Kuopio University Hospital, FI-70211 Kuopio, Finland
| | - Katarzyna Lukasiuk
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah 84108
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
18
|
Pitkänen A, Huusko N, Ndode-Ekane XE, Kyyriäinen J, Lipponen A, Lipsanen A, Sierra A, Bolkvadze T. Gender issues in antiepileptogenic treatments. Neurobiol Dis 2014; 72 Pt B:224-32. [PMID: 24912075 DOI: 10.1016/j.nbd.2014.05.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 05/23/2014] [Accepted: 05/29/2014] [Indexed: 12/12/2022] Open
Abstract
Disease modification of epilepsy refers to the alleviation of epileptogenesis or comorbidities after genetic or acquired epileptogenic brain insults. There are currently 30 proof-of-concept experimental pharmacologic studies that have demonstrated some beneficial disease-modifying effects. None of these studies, however, has yet passed from the laboratory to the clinic. The International League Against Epilepsy and American Epilepsy Society working groups on antiepileptogenic (AEG) therapies recently released recommendations for conducting preclinical AEG studies, taking into account many of the critiques raised by previous study designs. One of the issues relates to the lack of analysis of AEG efficacy in both sexes. A review of the literature reveals that most of the preclinical studies have been performed using male rodents, whereas clinical study cohorts include both males and females. Therefore, it is important to determine whether sex differences should be taken into account to a greater extent than they have been historically at different phases of experimental studies. Here we address the following questions based on analysis of available experimental AEG studies: (a) whether sex differences should be considered when searching for novel AEG targets, (b) how sex differences can affect the preclinical AEG study designs and analysis of outcome measures, and (c) what factors should be considered when examining the effect of sex on outcome of clinical AEG trials or the clinical use of AEGs.
Collapse
Affiliation(s)
- Asla Pitkänen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, PO Box 1777, FIN-70211 Kuopio, Finland.
| | - Noora Huusko
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Xavier Ekolle Ndode-Ekane
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Jenni Kyyriäinen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Anssi Lipponen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Anu Lipsanen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Alejandra Sierra
- Biomedical Imaging Unit, A. I. Virtanen Institute for Molecular Sciences, University of Eastern, Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Tamuna Bolkvadze
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| |
Collapse
|
19
|
Effect of lacosamide on structural damage and functional recovery after traumatic brain injury in rats. Epilepsy Res 2014; 108:653-65. [PMID: 24636248 DOI: 10.1016/j.eplepsyres.2014.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 01/31/2014] [Accepted: 02/01/2014] [Indexed: 11/23/2022]
Abstract
In a subgroup of patients, traumatic brain injury (TBI) results in the occurrence of acute epileptic seizures or even status epilepticus, which are treated with antiepileptic drugs (AEDs). Recent experimental data, however, suggest that administration of AEDs at the early post-injury phase can compromise the recovery process. The present study was designed to assess the profile of a novel anticonvulsant, lacosamide (Vimpat) on post-TBI structural, motor and cognitive outcomes. Moderate TBI was induced by lateral fluid-percussion injury in adult rats. Treatment with 0.9% saline or lacosamide (30 mg/kg, i.p.) was started at 30 min post-injury and continued at 8h intervals for 3d (total daily dose 90 mg/kg/d). Rats were randomly assigned to 4 treatment groups: sham-operated controls treated with vehicle (Sham-Veh) or lacosamide (Sham-LCM) and injured animals treated with vehicle (TBI-Veh) or lacosamide (TBI-LCM). As functional outcomes we tested motor recovery with composite neuroscore and beam-walking at 2, 7, and 15 d post-injury. Cognitive recovery was tested with the Morris water-maze at 12-14 d post-TBI. To assess the structural outcome, animals underwent magnetic resonance imaging (MRI) at 2 d post-TBI. At 16d post-TBI, rats were perfused for histology to analyze cortical and hippocampal neurodegeneration and axonal damage. Our data show that at 2 d post-TBI, both the TBI-Veh and TBI-LCM groups were equally impaired in neuroscore. Thereafter, motor recovery occurred similarly during the first week. At 2 wk post-TBI, recovery of the TBI-LCM group lagged behind that in the TBI-VEH group (p<0.05). Performance in beam-walking did not differ between the TBI-Veh and TBI-LCM groups. Both TBI groups were similarly impaired in the Morris water-maze at 2 wk post-TBI. MRI and histology did not reveal any differences in the cortical or hippocampal damage between the TBI-Veh and TBI-LCM groups. Taken together, acute treatment with LCM had no protective effects on post-TBI structural or functional impairment. Composite neuroscore in the TBI-LCM group lagged behind that in the TBI-Veh group at 15 d post-injury, but no compromise was found in other indices of post-TBI recovery in the LCM treated animals.
Collapse
|
20
|
|
21
|
Cell death and survival mechanisms are concomitantly active in the hippocampus of patients with mesial temporal sclerosis. Neuroscience 2013; 237:56-65. [DOI: 10.1016/j.neuroscience.2013.01.050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/18/2013] [Accepted: 01/19/2013] [Indexed: 11/24/2022]
|
22
|
Gabapentin is neuroprotective through glutamate receptor-independent mechanisms in staurosporine-induced apoptosis of cultured rat cerebellar neurons. Transl Neurosci 2013. [DOI: 10.2478/s13380-013-0139-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe anticonvulsants that are currently available modulate the activity of neuronal receptors and ion channels, which are equally involved in apoptotic pathways. We investigated the hypothesis that gabapentin (GP), an anticonvulsant without effect on glutamate receptors acting as GABA analog, has neuroprotective properties. For comparison, we chose topiramate (TPM), which has been reported to be neuroprotective via AMPA receptors blockade. For this purpose, we used rat cerebellar granule neuron (CGN) cultures and we triggered apoptosis independent of glutamate receptors with staurosporine, a broad-spectrum protein kinase inhibitor. GP at therapeutic range concentration significantly increased cell viability in CGN cultures maintained in physiological KCl concentration and reversed apoptosis induced by staurosporine. Blockade of NMDA or AMPA receptors by MK801 or NBQX, respectively, did not alter GP neuroprotection, which was reversed instead by GABA. In contrast, protective effect of TPM on STS-treated CGN cultures was annihilated by NBQX, and not altered by MK801 or GABA. Treatments with neuroprotective concentrations of GP or TPM did not modify the expression of neuronal cell adhesion molecule or synaptophysin or the morphological aspect of neuronal endings. In summary, we report that GP is neuroprotective through glutamate-receptor independent mechanisms and without alteration of neuronal plasticity markers, which makes it a possible candidate for clinical neuroprotection trials.
Collapse
|
23
|
Ono T, Galanopoulou AS. Epilepsy and epileptic syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 724:99-113. [PMID: 22411237 DOI: 10.1007/978-1-4614-0653-2_8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epilepsy is one of the most common neurological disorders. In most patients with epilepsy, seizures respond to available medications. However, a significant number of patients, especially in the setting of medically-intractable epilepsies, may experience different degrees of memory or cognitive impairment, behavioral abnormalities or psychiatric symptoms, which may limit their daily functioning. As a result, in many patients, epilepsy may resemble a neurodegenerative disease. Epileptic seizures and their potential impact on brain development, the progressive nature of epileptogenesis that may functionally alter brain regions involved in cognitive processing, neurodegenerative processes that relate to the underlying etiology, comorbid conditions or epigenetic factors, such as stress, medications, social factors, may all contribute to the progressive nature of epilepsy. Clinical and experimental studies have addressed the pathogenetic mechanisms underlying epileptogenesis and neurodegeneration.We will primarily focus on the findings derived from studies on one of the most common causes of focal onset epilepsy, the temporal lobe epilepsy, which indicate that both processes are progressive and utilize common or interacting pathways. In this chapter we will discuss some of these studies, the potential candidate targets for neuroprotective therapies as well as the attempts to identify early biomarkers of progression and epileptogenesis, so as to implement therapies with early-onset disease-modifying effects.
Collapse
Affiliation(s)
- Tomonori Ono
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
24
|
Sloviter RS. Progress on the issue of excitotoxic injury modification vs. real neuroprotection; implications for post-traumatic epilepsy. Neuropharmacology 2011; 61:1048-50. [PMID: 21839755 DOI: 10.1016/j.neuropharm.2011.07.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 07/27/2011] [Indexed: 11/17/2022]
Affiliation(s)
- Robert S Sloviter
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724-5050, USA.
| |
Collapse
|
25
|
François J, Germe K, Ferrandon A, Koning E, Nehlig A. Carisbamate has powerful disease-modifying effects in the lithium-pilocarpine model of temporal lobe epilepsy. Neuropharmacology 2011; 61:313-28. [PMID: 21539848 DOI: 10.1016/j.neuropharm.2011.04.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 04/13/2011] [Accepted: 04/15/2011] [Indexed: 11/30/2022]
Abstract
Lithium-pilocarpine, a relevant model of temporal lobe epilepsy was used to test the neuroprotective and antiepileptogenic effects of carisbamate. Status epilepticus (SE) was induced in adult rats by lithium and pilocarpine. Carisbamate (30, 60, 90, and 120 mg/kg) was injected at 1 and 9 h after SE onset and continued twice daily for 6 additional days. The reference groups received diazepam instead of carisbamate. Neuroprotection was assessed during the first 24 h of SE with Fluoro-Jade B and after 14 days with thionine staining. SE severity and epileptic outcome were assessed by video, and surface and depth electroencephalographic recordings. At the two highest doses, carisbamate treatment reduced SE severity; produced strong neuroprotection of hippocampus, ventral cortices, thalamus, and amygdala; prevented mossy fiber sprouting in the dentate gyrus of the hippocampus; and delayed or suppressed the occurrence of spontaneous motor seizures. Rats with no spontaneous motor seizures displayed spike-and-wave discharges that share all the characteristics of absence seizures. In conclusion, carisbamate is able to induce strong neuroprotection and affect the nature of epileptogenic events occurring during and after lithium-pilocarpine status epilepticus, reflecting marked insult- and disease-modifying effects.
Collapse
Affiliation(s)
- Jennifer François
- InsermU666, University Louis Pasteur; Faculty of Medicine, 11 rue Humann, 67085 Strasbourg Cedex, France.
| | | | | | | | | |
Collapse
|
26
|
Kouzounias K, Kimiskidis VK, Siozos T, Violaris K, Kostomitsopoulos N, Karayannakos PE, Sotirakoglou K, Nanassis K. Topiramate promotes neurological recovery in a new model of traumatic brain injury in rats. Neuroscience 2011; 183:171-7. [PMID: 21496474 DOI: 10.1016/j.neuroscience.2011.03.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/22/2011] [Accepted: 03/30/2011] [Indexed: 12/31/2022]
Abstract
The aim of this study is to investigate the neuroprotective effects of the anticonvulsant topiramate in a new model of traumatic brain injury in rats. A new model of traumatic brain injury, based on the weight-drop technique, was developed for the purpose of this study. Seventy-five male Wistar rats weighing 320-470 g were studied. All rats were anesthetized, subsequently submitted to a round craniectomy in the left parietal region and a weight of 50 g was used for the production of a cortical contusion. In study I, 44 rats were randomized in three groups to receive either topiramate 40 mg/kg (n=13), topiramate 60 mg/kg (n=14), or water for injection (n=17) i.p. 30 min after the injury and every 12 h thereafter for 3 days. The rats were tested clinically 24 h, 72 h, 10 days and 20 days after the injury. On day 21 the animals were sacrificed and the brains were removed and prepared for histopathological analysis. In study II, 19 rats were randomized to receive either topiramate 60 mg/kg (n=10) or water for injection (n=9) i.p. 30 min after the injury and every 12 h (four doses in total). 48 h after the injury the animals were sacrificed and the brains were rapidly removed and analyzed for water content with the dry-wet weight technique. The animals that received topiramate performed significantly better in neurological tests compared to the animals that received vehicle ten (P<0.05) and 20 (P<0.001) days after the injury. There was no difference between the high and the low dose of the drug. Topiramate had no effect on the anatomic volume of the lesion. The animals that received topiramate had a tendency to present with less cerebral edema formation, but the difference was not statistically significant (P>0.05). These findings suggest that topiramate promotes neurological recovery in rats after traumatic brain injury without affecting the final size of the traumatic lesion and that it might play a role in the reduction of post-traumatic cerebral edema.
Collapse
Affiliation(s)
- K Kouzounias
- Department of Neurosurgery, Aristotle University of Thessaloniki, Ippokrateio Hospital, Thessaloniki, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
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
|
28
|
Rattka M, Brandt C, Bankstahl M, Bröer S, Löscher W. Enhanced susceptibility to the GABA antagonist pentylenetetrazole during the latent period following a pilocarpine-induced status epilepticus in rats. Neuropharmacology 2011; 60:505-12. [DOI: 10.1016/j.neuropharm.2010.11.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/01/2010] [Accepted: 11/02/2010] [Indexed: 01/04/2023]
|
29
|
Simonato M, Zucchini S. Are the neurotrophic factors a suitable therapeutic target for the prevention of epileptogenesis? Epilepsia 2010; 51 Suppl 3:48-51. [DOI: 10.1111/j.1528-1167.2010.02609.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
30
|
Abstract
Prevention of epileptogenesis is an unmet need in medicine. During the last 3 years, however, several preclinical studies have demonstrated remarkable favorable effects of novel treatments on genetic and acquired epileptogenesis. These include the use of immunosuppressants and treatments that modify cellular adhesion, proliferation, and/or plasticity. In addition, the use of antiepileptic drugs in rats with genetic epilepsy or proconvulsants in acquired epilepsy models has provided somewhat unexpected favorable effects. This review summarizes these studies, and introduces some caveats when interpreting the data. In particular, the effect of genetic background, the severity of epileptogenic insult, the method and duration of seizure monitoring, and size of animal population are discussed. Furthermore, a novel scheme for defining epileptogenesis-related terms is presented.
Collapse
Affiliation(s)
- Asla Pitkänen
- Epilepsy Research Laboratory, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
31
|
The COX-2 inhibitor parecoxib is neuroprotective but not antiepileptogenic in the pilocarpine model of temporal lobe epilepsy. Exp Neurol 2010; 224:219-33. [PMID: 20353773 DOI: 10.1016/j.expneurol.2010.03.014] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 03/17/2010] [Accepted: 03/20/2010] [Indexed: 11/23/2022]
Abstract
The enzyme cyclooxygenase-2 (COX-2), which catalyzes the production of pro-inflammatory prostaglandins, is induced in the brain after various insults, thus contributing to brain inflammatory processes involved in the long-term consequences of such insults. Mounting evidence supports that inflammation may contribute to epileptogenesis and neuronal injury developing after brain insults. Anti-inflammatory treatments, such as selective COX-2 inhibitors, may thus constitute a novel approach for anti-epileptogenesis or disease-modification after brain injuries such as head trauma, cerebral ischemia or status epilepticus (SE). However, recent rat experiments with prophylactic administration of two different COX-2 inhibitors after SE resulted in conflicting results. In the present study, we evaluated whether treatment with parecoxib, a pro-drug of the highly potent and selective COX-2 inhibitor valdecoxib, alters the long-term consequences of a pilocarpine-induced SE in rats. Parecoxib was administered twice daily at 10 mg/kg for 18 days following SE. Five weeks after termination of treatment, spontaneous recurrent seizures were recorded by continuous video/EEG monitoring. Prophylactic treatment with parecoxib prevented the SE-induced increase in prostaglandin E(2) and reduced neuronal damage in the hippocampus and piriform cortex. However, the incidence, frequency or duration of spontaneous seizures developing after SE or the behavioral and cognitive alterations associated with epilepsy were not affected by parecoxib. Only the severity of spontaneous seizures was reduced, indicating a disease-modifying effect. These results substantiate that COX-2 contributes to neuronal injury developing after SE, but inhibition of COX-2 is no effective means to modify epileptogenesis.
Collapse
|
32
|
HAMED SHERIFAA. THE RATIONALE FOR NEUROPROTECTION IN EPILEPSY: STEPS FORWARD FOR NEW THERAPEUTIC AND PREVENTIVE STRATEGIES. J Integr Neurosci 2010. [DOI: 10.1142/s0219635210002378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
33
|
Abstract
In the last fifteen years, new antiepileptic medications have been offered for the treatment of patients with epilepsy. Nevertheless, despite optimal medical treatment, up to 30% of patients still experience recurrent seizures and the challenge for new, more efficacious and better-tolerated drugs continues. New antiepileptic drugs include the evolution of pre-existing drugs and new compounds identified through the investigation of additional molecular targets, such as SV2A synaptic vesicle protein, voltage-gated potassium channels, ionotropic and metabotropic glutamate receptors, and gap junctions. This paper reviews the available information on various classes of molecules that are in the pipeline as well as on the innovative approaches to the treatment of epilepsy.
Collapse
Affiliation(s)
- Pasquale Striano
- Muscular & Neurodegenerative Diseases Unit, Institute G. Gaslini, Genova, Italy
| | | |
Collapse
|
34
|
Mechanistic biomarkers for autism treatment. Med Hypotheses 2009; 73:950-4. [PMID: 19619951 DOI: 10.1016/j.mehy.2009.06.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Accepted: 06/13/2009] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Autism is a syndrome with a number of etiologies with differing mechanisms that lead to abnormal development. This review highlights the need to identify autism subgroups as they each might require unique approaches for prevention or treatment. METHODS Targeting treatments to specific mechanisms and utilizing biomarkers can more rapidly advance our understanding of how to classify and treat autism subgroups based on translational mechanisms. We illustrate this approach using mechanisms that may influence the course of autism and provide rationale for selected biomarkers that could guide treatments targeted anywhere from DNA to symptom expression. CONCLUSIONS The use of potential biomarkers that point to specific mechanisms of disordered neurodevelopment will help identify meaningful subtypes of autism and will help tailor treatment or prevention strategies for each mechanism rather than solely to a symptom category.
Collapse
|
35
|
Farooq MU, Bhatt A, Majid A, Gupta R, Khasnis A, Kassab MY. Levetiracetam for managing neurologic and psychiatric disorders. Am J Health Syst Pharm 2009; 66:541-61. [PMID: 19265183 DOI: 10.2146/ajhp070607] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The role of levetiracetam in different epileptic, nonepileptic, neurologic, and psychiatric disorders is discussed. SUMMARY Levetiracetam, an antiepileptic drug (AED), was first approved as an adjunctive therapy for the treatment of partial epilepsy in adults. It is currently being used in the treatment of multiple seizure disorders, including generalized tonic-clonic; absence; myoclonic, especially juvenile myoclonic; Lennox-Gastaut syndrome; and refractory epilepsy in children and adults. Data are emerging on possible uses of levetiracetam outside the realm of epilepsy because of its unique mechanisms of action. There is preliminary evidence about the efficacy of levetiracetam in the treatment of different psychiatric disorders, including anxiety, panic, stress, mood and bipolar, autism, and Tourette's syndrome. The most serious adverse effects associated with levetiracetam use are behavioral in nature and might be more common in patients with a history of psychiatric and neurobehavioral problems. CONCLUSION Levetiracetam is an effective AED with potential benefits in other neurologic and psychiatric disorders. The benefit-risk ratio in an individual patient with a specific condition should be used to determine its optimal use. Levetiracetam's use in nonepileptic conditions is not recommended until more data become available from larger trials.
Collapse
Affiliation(s)
- Muhammad U Farooq
- Department of Neurology and Ophthalmology, Michigan State University, 138 Service Road, East Lansing, MI 48824, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Molecular and cellular basis of epileptogenesis in symptomatic epilepsy. Epilepsy Behav 2009; 14 Suppl 1:16-25. [PMID: 18835369 DOI: 10.1016/j.yebeh.2008.09.023] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/18/2008] [Accepted: 09/18/2008] [Indexed: 12/21/2022]
Abstract
Epileptogenesis refers to a process in which an initial brain-damaging insult triggers a cascade of molecular and cellular changes that eventually lead to the occurrence of spontaneous seizures. Cellular alterations include neurodegeneration, neurogenesis, axonal sprouting, axonal injury, dendritic remodeling, gliosis, invasion of inflammatory cells, angiogenesis, alterations in extracellular matrix, and acquired channelopathies. Large-scale molecular profiling of epileptogenic tissue has provided information about the molecular pathways that can initiate and maintain cellular alterations. Currently we are learning how these pathways contribute to postinjury epileptogenesis and recovery process and whether they could be used as treatment targets.
Collapse
|
37
|
Ben-Menachem E, Sander JW, Stefan H, Schwalen S, Schäuble B. Topiramate monotherapy in the treatment of newly or recently diagnosed epilepsy. Clin Ther 2008; 30:1180-95. [PMID: 18691980 DOI: 10.1016/s0149-2918(08)80045-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The efficacy of topiramate (TPM) as an adjunctive treatment for epilepsy has been established in placebo-controlled clinical trials. Clinical trials of antiepileptic monotherapy usually evaluate low and high doses of study drug or compare study drug with another active agent. OBJECTIVE This article reviews available evidence for the use of TPM as monotherapy in patients with newly or recently diagnosed epilepsy. METHODS A search of MEDLINE, EMBASE, BIOSIS, SCISEARCH, and the Cochrane Database of Systematic Reviews (all years) for reports of controlled trials of TPM monotherapy in patients with recently diagnosed (within the previous 3 years) epilepsy was conducted in January 2008 using the terms topiramate, epilepsy, newly diagnosed, recently diagnosed, and monotherapy. Identified trials were included in the review if they were published in peer-reviewed journals and enrolled > or = 20 patients. RESULTS Three randomized, double-blind, controlled trials met the criteria for inclusion in the review. In a comparison of TPM 50 and 500 mg/d, the higher dose was associated with significantly greater freedom from seizures at 6 months compared with the lower dose (54% vs 39%, respectively; P = 0.02). The time to first seizure was significantly associated with mean plasma TPM concentrations (P = 0.015). In a comparison of TPM 50 and 400 mg/d, the time to first seizure was significantly longer with the higher dose compared with the lower dose (P<0.001, Kaplan-Meier analysis), and the probability of 12-month seizure freedom was significantly higher (76% vs 59%, respectively; P = 0.001). Again, the time to first seizure was significantly associated with mean plasma TPM concentrations (P = 0.029). In a comparative study of TPM 100 and 200 mg/d, carbamazepine 600 mg/d, and valproate 1250 mg/d, there was no significant difference in rates of 6-month seizure freedom with TPM 100 and 200 mg/d (49% and 44%, respectively), carbamazepine (44%), and valproate (44%). Adverse events in the 3 studies were similar between TPM dose groups, although the incidence generally increased with increasing doses, occurred early in treatment, and decreased with prolonged therapy. In a pooled analysis of the 3 trials, the most commonly occurring adverse events during dose titration were paresthesia (25%), fatigue (16%), dizziness (13%), somnolence (13%), and nausea (10%); the most frequent adverse events during maintenance therapy were headache (20%), decreased appetite (11%), and weight loss (11%). CONCLUSION In the 3 studies reviewed, TPM monotherapy was effective and generally well tolerated in patients with newly diagnosed epilepsy.
Collapse
Affiliation(s)
- Elinor Ben-Menachem
- Department of Clinical Neuroscience, Section of Neurology, Sahlgren University Hospital, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
38
|
Neurodevelopmental delay in children exposed to antiepileptic drugs in utero: a critical review directed at structural study-bias. J Neurol Sci 2008; 271:1-14. [PMID: 18479711 DOI: 10.1016/j.jns.2008.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 03/13/2008] [Accepted: 03/18/2008] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The general issue whether in utero exposure to antiepileptic drugs (AEDs) causes congenital malformations (teratogenicity) was raised as early as 1968. The 'congenital hydantoin syndrome' after intrauterine exposure to phenytoin (PHT) was first described in 1975. In 1984, DiLiberti proposed the label 'Fetal Valproate Syndrome' (FVS) for children with a cluster of minor congenital anomalies in the form of dysmorphic facial appearances with or without major abnormalities after intra-uterine exposure to valproate (VPA). Later, also the presence of central nervous system (CNS) dysfunction became part of the description. The question whether developmental delay, educational impairment, or behavioural disorders are also a characteristic of intrauterine exposure to AEDs and especially VPA, is of major importance to many women with epilepsy, parents and physicians involved. METHODS Literature was searched using MEDLINE and other relevant databases: 56 studies were identified and interpreted. RESULTS The identified studies do not allow definite conclusions. The possibility of neurodevelopmental delay, behavioural disorders, or learning disabilities as an outcome of in utero exposure to AEDs and especially VPA, needs to be considered seriously. The literature however does not provide evidence for a valid risk estimate. Moreover the evidence found for a specific increased risk for VPA could be structurally biased. DISCUSSION The major problem in this field is the methodology and in particular the existence of important confounding factors that complicate any attempt to correlate intra-uterine exposure to AEDs with neurodevelopmental delay. We propose a number of guidelines for studies on behavioural teratogenicity.
Collapse
|
39
|
Nehlig A. What is animal experimentation telling us about new drug treatments of status epilepticus? Epilepsia 2008; 48 Suppl 8:78-81. [PMID: 18330008 DOI: 10.1111/j.1528-1167.2007.01358.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Basic research is mostly focused on the consequences of status epilepticus (SE) in terms of neuronal loss, behavior, epileptogenesis or disease-modifying effects such as preventing epilepsy or reducing seizure severity. Among the drugs tested, several were able to trigger neuroprotection but only a few had disease-modifying effects. At this point, many data are still missing, namely which drugs could efficiently stop SE or which mechanisms of action should be searched for to prevent the harmful consequences of SE.
Collapse
Affiliation(s)
- Astrid Nehlig
- INSERM U 666, Faculty of Medicine, Strasbourg, France.
| |
Collapse
|
40
|
Cell and gene therapies in epilepsy – promising avenues or blind alleys? Trends Neurosci 2008; 31:62-73. [DOI: 10.1016/j.tins.2007.11.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 11/28/2007] [Accepted: 11/30/2007] [Indexed: 11/23/2022]
|
41
|
Karceski S. Electrical stimulation devices in the treatment of epilepsy. ACTA NEUROCHIRURGICA. SUPPLEMENT 2007; 97:247-59. [PMID: 17691311 DOI: 10.1007/978-3-211-33081-4_28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Over the last ten years there has been a progressively increasing interest in the research and clinical application of implantable electrical brain stimulation devices in the treatment of drug-resistant epilepsy. The concept is not new, but the efforts were strengthened and accelerated after the efficacy of vagus nerve stimulation in controlling epilepsy was first demonstrated in the early 1990s and gained subsequently the approval of the USA Food and Drug Administration in 1997. This chapter reviews the progress made in this field. Special emphasis is given to the most important available evidence from animal and human studies, the neuroanatomical pathways and the role of the relevant neurotransmitters, the stimulation devices and the significance of correct programming of the stimulation parameters. The chapter also examines the antiepileptic efficacy of stimulation in all the known targets including vagus nerve, cerebellum, thalamus, subthalamic nucleus, locus ceruleus, and epileptogenic cortex. On the basis of the current evidence, the future directions of this exciting field are described.
Collapse
Affiliation(s)
- S Karceski
- Department of Neurology, New York Presbyterian Hospital, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
42
|
Cunha AOS, Mortari MR, Carolino ROG, Coutinho-Netto J, Dos Santos WF. Glutamate binding is altered in hippocampus and cortex of Wistar rats after pilocarpine-induced Status Epilepticus. Neurosci Lett 2007; 424:51-4. [PMID: 17709190 DOI: 10.1016/j.neulet.2007.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 06/25/2007] [Accepted: 07/12/2007] [Indexed: 11/17/2022]
Abstract
Several evidences have pointed to biochemical alterations in some brain structures after experimental Status Epilepticus (SE). Thus, the effects of pilocarpine-induced SE on the glutamate binding in the hippocampus and cortex of Wistar rats were evaluated. Groups of animals were submitted to a 3h SE induced by intrahippocampal microinjection of pilocarpine, which was interrupted by the administration of sodium thiopental. Two weeks later the animals were sacrificed and had their cerebral cortices and hippocampi removed in order to perform the binding experiments. The results show that the pilocarpine-induced SE provoked an increase in 2.5-fold in the B(max) values for glutamate binding in the cortex, but not in the hippocampus. Moreover, we observed a 4-fold increase for the Kd values in the hippocampus and a 2-fold increase in the cortex. These findings might indicate that the epileptogenesis involves alterations in the glutamate receptors that are not restricted to the limbic system. Moreover, changes in these receptors are not exclusively of number, but rather involve the affinity for their ligands.
Collapse
Affiliation(s)
- Alexandra Olimpio Siqueira Cunha
- Neurobiology and Venoms Laboratory, Department of Biology, Faculty of Philosophy, Sciences and Literature, University of São Paulo, Brazil
| | | | | | | | | |
Collapse
|
43
|
Vajda FJE. Pharmacotherapy of epilepsy: new armamentarium, new issues. J Clin Neurosci 2007; 14:813-23. [PMID: 17618119 DOI: 10.1016/j.jocn.2007.02.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 01/29/2007] [Accepted: 02/04/2007] [Indexed: 10/23/2022]
Abstract
Since 1990 there have been over ten antiepileptic drugs (AEDs) approved for the therapy of epilepsy. These agents have a new spectrum of efficacy and novel adverse effects, some totally unexpected. They also represent an enormous escalation of costs. Few have been subjected to head-to-head comparisons in monotherapy against established AEDs. The aim of therapy is to eliminate rather than to reduce seizure manifestations. Many traditional agents have been phased out due to poor tolerability. New epilepsy syndromes and genetic contributions to epilepsy have been refined. Special considerations apply to various classes of sufferers such as the elderly, women of childbearing age, and sufferers with concomitant disorders, treated with medications capable of drug interactions. There is a recognition of the value of slow introduction, a preference for monotherapy, recognition of the effects of AEDs on hormones and reproductive function and effects on the fetus exposed to AEDs in utero, comprising physical malformations and effects on cognitive development. A balance between efficacy and safety is pivotal, as every preference about the initial pharmacotherapy of epilepsy and subsequent polytherapy has its protagonists. With improvement in diagnostic techniques and new therapeutic modalities it is likely that in the future, pharmacogenomics and an understanding of pharmacoresistance may influence drug selection for individual patients with epilepsy.
Collapse
Affiliation(s)
- Frank J E Vajda
- Department of Medicine, Monash University and Medical Centre, 246 Clayton Road, Clayton 3168, Victoria, and Department of Medicine, St Vincents Hospital, Melbourne, Australia.
| |
Collapse
|
44
|
Pitkänen A. New data suggest that discontinuation of status epilepticus is not necessary for antiepileptogenic effect in immature brain. Epilepsy Curr 2007; 6:170-2. [PMID: 17260049 PMCID: PMC1783477 DOI: 10.1111/j.1535-7511.2006.00135.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Treatment of Experimental Status Epilepticus in Immature Rats: Dissociation between Anticonvulsant and Antiepileptogenic Effects Suchomelova L, Baldwin RA, Kubova H, Thompson KW, Sankar R, Wasterlain CG Pediatr Res 2006;59:237–243 We studied the effects of treating status epilepticus (SE) induced by lithium and pilocarpine at postnatal day 15 (P15) or 28 (P28), on the severity of acute SE and of SEinduced epileptogenesis. Rats received topiramate (10 or 50 mg/kg, IP) or diazepam (5 mg/kg, IP) 20, 40 or 70 min after pilocarpine, and three months after SE 24-h video/EEG recordings were obtained for one (P28) or two weeks (P15) continuously. In P15 rats, topiramate did not modify the course of SE, yet treatment at 20 or 40 min completely prevented the development of spontaneous recurrent seizures (SRS) while later treatment (70 min) was partially effective in reducing the severity and frequency of SRS. Diazepam was effective against acute SE at all time points tested. Early (20 min) but not late treatment with diazepam had the effect of reducing the frequency and severity of SRS. In P28 rats, both drugs reduced the cumulative seizure time. Early treatment (20 min) with either drug reduced the incidence of chronic epilepsy. Late treatment (40/70 min) did not alter the incidence of SRS, but decreased their frequency. This study demonstrates that, in the treatment of SE, anticonvulsant and antiepileptogenic effects can be dissociated in a development-specific manner: topiramate was antiepileptogenic without being an effective anticonvulsant in P15 animals at the doses tested. Diazepam, on the other hand, was a better anticonvulsant than an antiepileptogenic agent in the P15 animals at the dose tested. Such effects were not seen in the older animals.
Collapse
|
45
|
Brandt C, Heile A, Potschka H, Stoehr T, Löscher W. Effects of the novel antiepileptic drug lacosamide on the development of amygdala kindling in rats. Epilepsia 2007; 47:1803-9. [PMID: 17116018 DOI: 10.1111/j.1528-1167.2006.00818.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE The current treatment of epilepsy focuses exclusively on the prophylaxis or suppression of seizures and thus provides merely a symptomatic treatment, without clear influence on the course of the disease. There is a need for new drugs that act at different molecular targets than currently available antiepileptic drugs (AEDs) and for new therapies designed to block the process of epileptogenesis. In recent years, different research lines have examined the epileptogenic process in order to understand the different stages in this process, and with the hope that early recognition and intervention could prevent the development or progression of epilepsy. In animals, acquired epilepsy is studied most commonly with the kindling model and status epilepticus models. In the present study, we used the kindling model to evaluate whether the novel AED lacosamide affects kindling-induced epileptogenesis. This drug does not seem to act by any of the mechanisms of currently available AEDs, but the exact molecular mechanisms of action of lacosamide have not yet been clarified. METHODS Groups of 9-10 rats were treated with either vehicle or different doses of lacosamide (3, 10, or 30 mg/kg/day) over 22-23 days during amygdala kindling. RESULTS Daily administration of lacosamide during kindling acquisition produced a dose-dependent effect on kindling development. While the drug was inactive at 3 mg/kg/day, significant retardation of kindling was observed at 10 mg/kg/day, by which the average number of stimulations to reach kindling criterion was increased by >90%. A significant inhibitory effect on kindling acquisition was also observed with 30 mg/kg/day, but this dose of lacosamide was associated with adverse effects. CONCLUSIONS The present data demonstrate that lacosamide, in addition to exerting anticonvulsant activity, has the potential to retard kindling-induced epileptogenesis. Whether this indicates that lacosamide possesses antiepileptogenic or disease-modifying potential needs to be further evaluated, including studies in other models of acquired epilepsy.
Collapse
Affiliation(s)
- Claudia Brandt
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | | | | | | | | |
Collapse
|
46
|
François J, Koning E, Ferrandon A, Nehlig A. The combination of topiramate and diazepam is partially neuroprotective in the hippocampus but not antiepileptogenic in the lithium-pilocarpine model of temporal lobe epilepsy. Epilepsy Res 2006; 72:147-63. [PMID: 16945504 DOI: 10.1016/j.eplepsyres.2006.07.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/19/2006] [Accepted: 07/21/2006] [Indexed: 11/29/2022]
Abstract
Lithium-pilocarpine induces status epilepticus (SE), leading to extensive damage and spontaneous recurrent seizures (SRS). Neuroprotective and antiepileptogenic effects of topiramate (TPM) associated with diazepam (DZP) were investigated in this model. SE was induced by LiCl and pilocarpine. TPM (10, 30 or 60 mg/kg) was injected at the onset of SE and 10h later and DZP (2.5 and 1.25mg/kg) at 2 and 10h after SE. TPM treatment was continued twice daily for 6 days. Other rats received two injections of DZP on the day of SE. Cell counting was performed on thionine-stained sections 14 days after SE and after 2 months of epilepsy. Occurrence and frequency of SRS were video-recorded. The MRI T2-weighted signal was quantified in hippocampus and ventral cortices. DZP-TPM treatment induced partial neuroprotection in CA1 and hilus, and tended to increase the percentage of rats with protected neurons in layer III/IV of the ventral entorhinal cortex. The latency to and frequency of SRS were not modified by DZP-TPM. T2-weighted signal was decreased in hippocampus 3 days after SE at all TPM doses and in ventral hippocampus after epilepsy onset. In conclusion, although DZP-TPM treatment was able to partially protect two areas critical for epileptogenesis, the hippocampus and ventral entorhinal cortex, it was not sufficient to prevent epileptogenesis.
Collapse
Affiliation(s)
- Jennifer François
- INSERM U666, Faculty of Medicine, 11 rue Humann, 67085 Strasbourg Cedex, France.
| | | | | | | |
Collapse
|
47
|
Frisch C, Kudin AP, Elger CE, Kunz WS, Helmstaedter C. Amelioration of water maze performance deficits by topiramate applied during pilocarpine-induced status epilepticus is negatively dose-dependent. Epilepsy Res 2006; 73:173-80. [PMID: 17084066 DOI: 10.1016/j.eplepsyres.2006.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Revised: 10/02/2006] [Accepted: 10/02/2006] [Indexed: 11/24/2022]
Abstract
Temporal lobe epilepsy is characterized by a progressive loss of memory capacities, due to sclerosis and functional impairment of mesiotemporal brain areas. We have shown recently that topiramate (TPM) dose-dependently protects hippocampal CA1 and CA3 neurons during initial status epilepticus in the rat pilocarpine model of temporal lobe epilepsy by inhibition of mitochondrial transition pore opening. In the present study, in order to evaluate possible positive effects of the treatment on learning and memory, we investigated water maze performance of rats receiving different dosages of TPM (20 and 100 mg/kg) after 40 min and 4 mg/kg diazepam after 160 min of pilocarpine-induced status epilepticus in relation to performance of animals receiving 4 mg/kg diazepam after 40 min of SE, and to performance of sham-treated control animals. Unexpectedly, 20 but not 100 mg/kg TPM significantly extenuated short-term memory deficits. While neuroprotective effects of TPM were observed in hippocampal CA subfields of animals treated with 100 mg/kg TPM, cell loss in rats treated with 20 mg/kg TPM was indistinguishable from animals receiving diazepam only. The present results indicate a negative dose-dependency of memory-saving effects of TPM applied during status epilepticus apparently dissociated from hippocampal neuroprotection.
Collapse
Affiliation(s)
- Christian Frisch
- Department of Epileptology, University of Bonn Medical Center, Sigmund Freud-Strasse 25, 53105 Bonn, Germany.
| | | | | | | | | |
Collapse
|
48
|
Dichter MA. Models of epileptogenesis in adult animals available for antiepileptogenesis drug screening. Epilepsy Res 2006; 68:31-5. [PMID: 16377136 DOI: 10.1016/j.eplepsyres.2005.09.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Accepted: 09/14/2005] [Indexed: 11/24/2022]
Abstract
Epileptogenesis is the process by which parts of a normal brain are converted to a hyperexcitable brain, often after an injury. Researchers must understand this process before they know where and how to change it. Animal models are used to evaluate the process of epileptogenesis by studing status epelepticus, electrical kindling, or other methods that provoke injuries. All are associated with neuronal loss to more or less degree, synaptic reorganization, axon sprouting, neurogenesis, gliosis, and changes in gene expression in neurons and astrocytes. He describes several types of animal models and how they might be useful in developing effective strategies for preventing epilepsy.
Collapse
|
49
|
Abstract
As in Clark and Prout's classic work, we identify three phases of generalised convulsive status epilepticus, which we call impending, established, and subtle. We review physiological and subcellular changes that might play a part in the transition from single seizures to status epilepticus and in the development of time-dependent pharmacoresistance. We review the principles underlying the treatment of status epilepticus and suggest that prehospital treatment is beneficial, that therapeutic drugs should be used in rapid sequence according to a defined protocol, and that refractory status epilepticus should be treated with general anaesthesia. We comment on our preference for drugs with a short elimination half-life and discuss some therapeutic choices.
Collapse
Affiliation(s)
- James W Y Chen
- Department of Neurology and Brain Research Institute, Geffen School of Medicine at UCLA, and VA Greater Los Angeles Health Care System, Los Angeles, CA 90073, USA
| | | |
Collapse
|
50
|
Abstract
The efficacy of antiepileptic drugs (AEDs) and psychotropic medications in children with autism is limited to the treatment of seizures or to specific behaviors such as irritability, impulsivity, hyperactivity, repetitive behaviors, or aggression. The reliability and value of the available data--to determine the efficacy of these medications in autism--are limited by lack of controlled clinical trials, the small number of subjects, the heterogeneity of the population studied, and the brief duration of most drug trials. Indeed, few controlled clinical trials using AEDs in autism, with or without seizures, have been conducted. Because some AEDs also have a positive effect on mood, the benefits that children with autism sometimes obtain from these medications may not be due to the treatment of the abnormal electrical activity or the seizures per se but to an effect on common neuronal systems responsible for both behavior and epilepsy. The relationship between epilepsy and autism, and specifically the effects that abnormal electrical activity may have on the developing brain, may provide some valuable insights into the type of studies that are needed to help us understand the pathophysiology of autism.
Collapse
Affiliation(s)
- Roberto Tuchman
- Department of Neurology, Dan Marino Center, Miami Children's Hospital, University of Miami, Miami, Florida, USA.
| |
Collapse
|