1
|
Monteiro ÁB, Alves AF, Ribeiro Portela AC, Oliveira Pires HF, Pessoa de Melo M, Medeiros Vilar Barbosa NM, Bezerra Felipe CF. Pentylenetetrazole: A review. Neurochem Int 2024; 180:105841. [PMID: 39214154 DOI: 10.1016/j.neuint.2024.105841] [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: 06/13/2024] [Revised: 08/17/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Pentylenetetrazole (PTZ), a tetrazole derivative, is commonly used as a chemical agent to induce neurological disorders and replicate the characteristics of human epileptic seizures in animal models. This review offers a comprehensive analysis of the behavioral, neurophysiological, and neurochemical changes induced by PTZ. The epileptogenic and neurotoxic mechanisms of PTZ are associated with an imbalance between the GABAergic and glutamatergic systems. At doses exceeding 60 mg/kg, PTZ exerts its epileptic effects by non-competitively antagonizing GABAA receptors and activating NMDA receptors, resulting in an increased influx of cations such as Na+ and Ca2+. Additionally, PTZ promotes oxidative stress, microglial activation, and the synthesis of pro-inflammatory mediators, all of which are features characteristic of glutamatergic excitotoxicity. These mechanisms ultimately lead to epileptic seizures and neuronal cell death, which depend on the dosage and method of administration. The behavioral, electroencephalographic, and histological changes associated with PTZ further establish it as a valuable preclinical model for the study of epileptic seizures, owing to its simplicity, cost-effectiveness, and reproducibility.
Collapse
Affiliation(s)
- Álefe Brito Monteiro
- Laboratory of Psychopharmacology, Institute of Drugs and Medicines Research, Federal University of Paraíba, Brazil
| | - Alan Ferreira Alves
- Laboratory of Psychopharmacology, Institute of Drugs and Medicines Research, Federal University of Paraíba, Brazil
| | | | | | - Mayara Pessoa de Melo
- Laboratory of Psychopharmacology, Institute of Drugs and Medicines Research, Federal University of Paraíba, Brazil
| | | | | |
Collapse
|
2
|
Reddy DS, Vadassery A, Ramakrishnan S, Singh T, Clossen B, Wu X. Kindling Models of Epileptogenesis for Developing Disease-Modifying Drugs for Epilepsy. Curr Protoc 2024; 4:e70020. [PMID: 39436626 PMCID: PMC11498896 DOI: 10.1002/cpz1.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Kindling models are widely used animal models to study the pathobiology of epilepsy and epileptogenesis. These models exhibit distinctive features whereby sub-threshold stimuli instigate the initial induction of brief focal seizures. Over time, the severity and duration of these seizures progressively increase, leading to a fully epileptic state, which is marked by consistent development of generalized tonic-clonic seizures. Kindling involves focal stimulation via implanted depth electrodes or repeated administration of chemoconvulsants such as pentylenetetrazol. Comparative analysis of preclinical and clinical findings has confirmed a high predictive validity of fully kindled animals for testing novel antiseizure medications. Thus, kindling models remain an essential component of anticonvulsant drug development programs. This article provides a comprehensive guide to working protocols, testing of therapeutic drugs, outcome parameters, troubleshooting, and data analysis for various electrical and chemical kindling epileptogenesis models for new therapeutic development and optimization. The use of pharmacological agents or genetically modified mice in kindling experiments is valuable, offering insights into the impact of a specific target on various aspects of seizures, including thresholds, initiation, spread, termination, and the generation of a hyperexcitable network. These kindling epileptogenesis paradigms are helpful in identifying mechanisms and disease-modifying interventions for epilepsy. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Hippocampal kindling Basic Protocol 2: Amygdala kindling Basic Protocol 3: Rapid hippocampal kindling Basic Protocol 4: Chemical kindling.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
- Institute of Pharmacology and Neurotherapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Abhinav Vadassery
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Bryan Clossen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
3
|
Tang Y, Liu Y, Gong Y, Zhang S, Cui S, Wang Y, Chen Z, Xu C. Caspase-1 inhibitor CZL80 protects against acute seizures via amplifying the inhibitory neural transmission. Neurochem Int 2024; 179:105809. [PMID: 39047793 DOI: 10.1016/j.neuint.2024.105809] [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: 06/19/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Current anti-seizure medications (ASDs) primarily target ion channels or neurotransmissions; however, their practicability is limited by unwanted side-effects and pharmacoresistance. Cumulative evidence has proposed pro-inflammatory caspase-1 as a potential target for developing ASDs. In this study, we showed that the small-molecular caspase-1 inhibitor CZL80 can prevent seizures in various models including the maximal electroshock (MES), the pentylenetetrazol (PTZ), and the amygdaloid kindled models. Specifically, we discovered that CZL80 prevented death, reduced the duration of generalized seizures, and increased the threshold of generalized seizures in a dose-dependent manner in the MES model. In the PTZ model, CZL80 decreased the seizure stages, prolonged the latency to stage 4 seizures, and decreased the death rate. And in amygdaloid kindled rats, CZL80 inhibited the seizure stages, shortened the durations of both generalized seizures and after-discharges. And the anti-seizure efficacy of CZL80 was diminished in caspase-1 knockout mice. In vitro electrophysiology recordings revealed that CZL80 was able to decreased the excitability of glutamatergic pyramidal neurons, as denoted by reducing the spontaneous neuronal firings and increasing the rheobase injected currents to elicit action potentials. Furthermore, CZL80 was able to increase the amplitudes of inhibitory post-synaptic currents (IPSC), while the excitatory post-synaptic currents (EPSC) were not influenced. Lastly, daily administration of CZL80 for 3 weeks did not influence the normal locomotor functions in mice. In sum, our results highlighted CZL80 as a potential anti-seizure therapy with therapeutic significance.
Collapse
Affiliation(s)
- Yingying Tang
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yao Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yiwei Gong
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Shuo Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Sunliang Cui
- College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
4
|
Jakubiec M, Abram M, Zagaja M, Socała K, Panic V, Latacz G, Mogilski S, Szafarz M, Szala-Rycaj J, Saunders J, West PJ, Nieoczym D, Przejczowska-Pomierny K, Szulczyk B, Krupa A, Wyska E, Wlaź P, Metcalf CS, Wilcox K, Andres-Mach M, Kamiński RM, Kamiński K. Discovery and Profiling of New Multimodal Phenylglycinamide Derivatives as Potent Antiseizure and Antinociceptive Drug Candidates. ACS Chem Neurosci 2024; 15:3228-3256. [PMID: 39166702 PMCID: PMC11378297 DOI: 10.1021/acschemneuro.4c00438] [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] [Indexed: 08/23/2024] Open
Abstract
We developed a focused series of original phenyl-glycinamide derivatives which showed potent activity across in vivo mouse seizure models, namely, maximal electroshock (MES) and 6 Hz (using both 32 and 44 mA current intensities) seizure models. Following intraperitoneal (i.p.) administration, compound (R)-32, which was identified as a lead molecule, demonstrated potent protection against all seizure models with ED50 values of 73.9 mg/kg (MES test), 18.8 mg/kg (6 Hz, 32 mA test), and 26.5 mg/kg (6 Hz, 44 mA test). Furthermore, (R)-32 demonstrated efficacy in both the PTZ-induced kindling paradigm and the ivPTZ seizure threshold test. The expression of neurotrophic factors, such as mature brain-derived neurotrophic factor (mBDNF) and nerve growth factor (NGF), in the hippocampus and/or cortex of mice, and the levels of glutamate and GABA were normalized after PTZ-induced kindling by (R)-32. Importantly, besides antiseizure activity, (R)-32 demonstrated potent antinociceptive efficacy in formalin-induced pain, capsaicin-induced pain, as well as oxaliplatin- and streptozotocin-induced peripheral neuropathy in mice (i.p.). No influence on muscular strength and body temperature in mice was observed. Pharmacokinetic studies and in vitro ADME-Tox data (i.e., high metabolic stability in human liver microsomes, a weak influence on CYPs, no hepatotoxicity, satisfactory passive transport, etc.) proved favorable drug-like properties of (R)-32. Thermal stability of (R)-32 shown in thermogravimetry and differential scanning calorimetry gives the opportunity to develop innovative oral solid dosage forms loaded with this compound. The in vitro binding and functional assays indicated its multimodal mechanism of action. (R)-32, beyond TRPV1 antagonism, inhibited calcium and sodium currents at a concentration of 10 μM. Therefore, the data obtained in the current studies justify a more detailed preclinical development of (R)-32 for epilepsy and pain indications.
Collapse
Affiliation(s)
- Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Mirosław Zagaja
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin 20-950, Poland
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, Lublin 20-033, Poland
| | - Vanja Panic
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Szczepan Mogilski
- Department Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Joanna Szala-Rycaj
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin 20-950, Poland
| | - Jerry Saunders
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Peter J West
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Dorota Nieoczym
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, Lublin 20-033, Poland
| | - Katarzyna Przejczowska-Pomierny
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Bartłomiej Szulczyk
- Chair and Department of Pharmacotherapy and Pharmaceutical Care, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1B, Warsaw 02-097, Poland
| | - Anna Krupa
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, Lublin 20-033, Poland
| | - Cameron S Metcalf
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Karen Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Marta Andres-Mach
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin 20-950, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow 30-688, Poland
| |
Collapse
|
5
|
Barker-Haliski M, Hawkins NA. Innovative drug discovery strategies in epilepsy: integrating next-generation syndrome-specific mouse models to address pharmacoresistance and epileptogenesis. Expert Opin Drug Discov 2024; 19:1099-1113. [PMID: 39075876 PMCID: PMC11390315 DOI: 10.1080/17460441.2024.2384455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Although there are numerous treatment options already available for epilepsy, over 30% of patients remain resistant to these antiseizure medications (ASMs). Historically, ASM discovery has relied on the demonstration of efficacy through the use of 'traditional' acute in vivo seizure models (e.g. maximal electroshock, subcutaneous pentylenetetrazol, and kindling). However, advances in genetic sequencing technologies and remaining medical needs for people with treatment-resistant epilepsy or special patient populations have encouraged recent efforts to identify novel compounds in syndrome-specific models of epilepsy. Syndrome-specific models, including Scn1a variant models of Dravet syndrome and APP/PS1 mice associated with familial early-onset Alzheimer's disease, have already led to the discovery of two mechanistically novel treatments for developmental and epileptic encephalopathies (DEEs), namely cannabidiol and soticlestat, respectively. AREAS COVERED In this review, the authors discuss how it is likely that next-generation drug discovery efforts for epilepsy will more comprehensively integrate syndrome-specific epilepsy models into early drug discovery providing the reader with their expert perspectives. EXPERT OPINION The percentage of patients with pharmacoresistant epilepsy has remained unchanged despite over 30 marketed ASMs. Consequently, there is a high unmet need to reinvent and revise discovery strategies to more effectively address the remaining needs of patients with specific epilepsy syndromes, including drug-resistant epilepsy and DEEs.
Collapse
Affiliation(s)
| | - Nicole A Hawkins
- Feinberg School of Medicine Chicago, Northwestern University, Chicago, IL, USA
| |
Collapse
|
6
|
Hu L, Liu Y, Yuan Z, Guo H, Duan R, Ke P, Meng Y, Tian X, Xiao F. Glucose-6-phosphate dehydrogenase alleviates epileptic seizures by repressing reactive oxygen species production to promote signal transducer and activator of transcription 1-mediated N-methyl-d-aspartic acid receptors inhibition. Redox Biol 2024; 74:103236. [PMID: 38875958 PMCID: PMC11225908 DOI: 10.1016/j.redox.2024.103236] [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: 05/09/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
The pathogenesis of epilepsy remains unclear; however, a prevailing hypothesis suggests that the primary underlying cause is an imbalance between neuronal excitability and inhibition. Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme in the pentose phosphate pathway, which is primarily involved in deoxynucleic acid synthesis and antioxidant defense mechanisms and exhibits increased expression during the chronic phase of epilepsy, predominantly colocalizing with neurons. G6PD overexpression significantly reduces the frequency and duration of spontaneous recurrent seizures. Furthermore, G6PD overexpression enhances signal transducer and activator of transcription 1 (STAT1) expression, thus influencing N-methyl-d-aspartic acid receptors expression, and subsequently affecting seizure activity. Importantly, the regulation of STAT1 by G6PD appears to be mediated primarily through reactive oxygen species signaling pathways. Collectively, our findings highlight the pivotal role of G6PD in modulating epileptogenesis, and suggest its potential as a therapeutic target for epilepsy.
Collapse
Affiliation(s)
- Liqin Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Ziwei Yuan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Haokun Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Ran Duan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Pingyang Ke
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Yuan Meng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
Pérez-Pérez D, Monío-Baca C, von Rüden EL, Buchecker V, Wagner A, Schönhoff K, Zvejniece L, Klimpel D, Potschka H. Preclinical efficacy profiles of the sigma-1 modulator E1R and of fenfluramine in two chronic mouse epilepsy models. Epilepsia 2024; 65:2470-2482. [PMID: 39119787 DOI: 10.1111/epi.18037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Given its key homeostatic role affecting mitochondria, ionotropic and metabotropic receptors, and voltage-gated ion channels, sigma-1 receptor (Sig1R) represents an interesting target for epilepsy management. Antiseizure effects of the positive allosteric modulator E1R have already been reported in acute seizure models. Although modulation of serotonergic neurotransmission is considered the main mechanism of action of fenfluramine, its interaction with Sig1R may be of additional relevance. METHODS To further explore the potential of Sig1R as a target, we assessed the efficacy and tolerability of E1R and fenfluramine in two chronic mouse models, including an amygdala kindling paradigm and the intrahippocampal kainate model. The relative contribution of the interaction with Sig1R was analyzed using combination experiments with the Sig1R antagonist NE-100. RESULTS Whereas E1R exerted pronounced dose-dependent antiseizure effects at well-tolerated doses in fully kindled mice, only limited effects were observed in response to fenfluramine, without a clear dose dependency. In the intrahippocampal kainate model, E1R failed to influence electrographic seizure activity. In contrast, fenfluramine significantly reduced the frequency of electrographic seizure events and their cumulative duration. Pretreatment with NE-100 reduced the effects of E1R and fenfluramine in the kindling model. Surprisingly, pre-exposure to NE-100 in the intrahippocampal kainate model rather enhanced and prolonged fenfluramine's antiseizure effects. SIGNIFICANCE In conclusion, the kindling data further support Sig1R as an interesting target for novel antiseizure medications. However, it is necessary to further explore the preclinical profile of E1R in chronic epilepsy models with spontaneous seizures. Despite the rather limited effects in the kindling paradigm, the findings from the intrahippocampal kainate model suggest that it is of interest to further assess a possible broad-spectrum potential of fenfluramine.
Collapse
Affiliation(s)
- Daniel Pérez-Pérez
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Cristina Monío-Baca
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Verena Buchecker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Amelie Wagner
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katharina Schönhoff
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Liga Zvejniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Dennis Klimpel
- Department of Forensic and Clinical Toxicology, Medizinisches Versorgungszentrum Labor Krone, Bad Salzuflen, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
8
|
Paulussen I, Beckert H, Musial TF, Gschossmann LJ, Wolf J, Schmitt M, Clasadonte J, Mairet-Coello G, Wolff C, Schoch S, Dietrich D. SV2B defines a subpopulation of synaptic vesicles. J Mol Cell Biol 2024; 15:mjad054. [PMID: 37682518 PMCID: PMC11184983 DOI: 10.1093/jmcb/mjad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/03/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023] Open
Abstract
Synaptic vesicles can undergo several modes of exocytosis, endocytosis, and trafficking within individual synapses, and their fates may be linked to different vesicular protein compositions. Here, we mapped the intrasynaptic distribution of the synaptic vesicle proteins SV2B and SV2A in glutamatergic synapses of the hippocampus using three-dimensional electron microscopy. SV2B was almost completely absent from docked vesicles and a distinct cluster of vesicles found near the active zone. In contrast, SV2A was found in all domains of the synapse and was slightly enriched near the active zone. SV2B and SV2A were found on the membrane in the peri-active zone, suggesting the recycling from both clusters of vesicles. SV2B knockout mice displayed an increased seizure induction threshold only in a model employing high-frequency stimulation. Our data show that glutamatergic synapses generate molecularly distinct populations of synaptic vesicles and are able to maintain them at steep spatial gradients. The almost complete absence of SV2B from vesicles at the active zone of wildtype mice may explain why SV2A has been found more important for vesicle release.
Collapse
Affiliation(s)
- Isabelle Paulussen
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, Bonn 53127, Germany
- Synaptic Neuroscience Team, Department of Neuropathology, University Hospital Bonn, Bonn 53127, Germany
| | - Hannes Beckert
- Microscopy Core Facility, Medical Faculty, University of Bonn, Bonn 53127, Germany
| | - Timothy F Musial
- Microscopy Core Facility, Medical Faculty, University of Bonn, Bonn 53127, Germany
| | - Lena J Gschossmann
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, Bonn 53127, Germany
- Synaptic Neuroscience Team, Department of Neuropathology, University Hospital Bonn, Bonn 53127, Germany
| | - Julia Wolf
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, Bonn 53127, Germany
- Synaptic Neuroscience Team, Department of Neuropathology, University Hospital Bonn, Bonn 53127, Germany
| | | | | | | | | | - Susanne Schoch
- Synaptic Neuroscience Team, Department of Neuropathology, University Hospital Bonn, Bonn 53127, Germany
| | - Dirk Dietrich
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, Bonn 53127, Germany
| |
Collapse
|
9
|
Terman SW, Kirkpatrick L, Kerr WT, Akiyama LF, Baajour W, Atilgan D, Dorotan MKC, Choi HW, French JA. Challenges and directions in epilepsy diagnostics and therapeutics: Proceedings of the 17th Epilepsy Therapies and Diagnostics Development conference. Epilepsia 2024; 65:846-860. [PMID: 38135921 PMCID: PMC11018495 DOI: 10.1111/epi.17875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
Substantial efforts are underway toward optimizing the diagnosis, monitoring, and treatment of seizures and epilepsy. We describe preclinical programs in place for screening investigational therapeutic candidates in animal models, with particular attention to identifying and eliminating drugs that might paradoxically aggravate seizure burden. After preclinical development, we discuss challenges and solutions in the design and regulatory logistics of clinical trial execution, and efforts to develop disease biomarkers and interventions that may be not only seizure-suppressing, but also disease-modifying. As disease-modifying treatments are designed, there is clear recognition that, although seizures represent one critical therapeutic target, targeting nonseizure outcomes like cognitive development or functional outcomes requires changes to traditional designs. This reflects our increasing understanding that epilepsy is a disease with profound impact on quality of life for the patient and caregivers due to both seizures themselves and other nonseizure factors. This review examines selected key challenges and future directions in epilepsy diagnostics and therapeutics, from drug discovery to translational application.
Collapse
Affiliation(s)
- Samuel W Terman
- University of Michigan Department of Neurology, Ann Arbor, MI 48109, USA
| | - Laura Kirkpatrick
- University of Pittsburgh Department of Neurology, Pittsburgh, PA 15213, USA
- University of Pittsburgh Department of Pediatrics, Pittsburgh, PA 15213, USA
| | - Wesley T Kerr
- University of Michigan Department of Neurology, Ann Arbor, MI 48109, USA
- University of Pittsburgh Department of Neurology, Pittsburgh, PA 15213, USA
- University of Pittsburgh Department of Biomedical Informatics, Pittsburgh, PA 15213, USA
| | - Lisa F Akiyama
- University of Washington Department of Neurology, Seattle, WA 98105, USA
| | - Wadih Baajour
- University of Texas Health Science Center at Houston, Department of Neurology, Houston, TX 77030, USA
| | - Deniz Atilgan
- University of Texas Health Science Center at Houston, Department of Neurology, Houston, TX 77030, USA
| | | | - Hyoung Won Choi
- Emory University Department of Pediatrics, Division of Neurology, Atlanta, GA 30322
| | - Jacqueline A French
- NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
10
|
Yang Y, Chen Z, Zhou J, Jiang S, Wang G, Wan L, Yu J, Jiang M, Wang Y, Hu J, Liu X, Wang Y. Anti-PD-1 treatment protects against seizure by suppressing sodium channel function. CNS Neurosci Ther 2024; 30:e14504. [PMID: 37904722 PMCID: PMC11017438 DOI: 10.1111/cns.14504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/01/2023] Open
Abstract
AIMS Although programmed cell death protein 1 (PD-1) typically serves as a target for immunotherapies, a few recent studies have found that PD-1 is expressed in the nervous system and that neuronal PD-1 might play a crucial role in regulating neuronal excitability. However, whether brain-localized PD-1 is involved in seizures and epileptogenesis is still unknown and worthy of in-depth exploration. METHODS The existence of PD-1 in human neurons was confirmed by immunohistochemistry, and PD-1 expression levels were measured by real-time quantitative PCR (RT-qPCR) and western blotting. Chemoconvulsants, pentylenetetrazol (PTZ) and cyclothiazide (CTZ), were applied for the establishment of in vivo (rodents) and in vitro (primary hippocampal neurons) models of seizure, respectively. SHR-1210 (a PD-1 monoclonal antibody) and sodium stibogluconate (SSG, a validated inhibitor of SH2-containing protein tyrosine phosphatase-1 [SHP-1]) were administrated to investigate the impact of PD-1 pathway blockade on epileptic behaviors of rodents and epileptiform discharges of neurons. A miRNA strategy was applied to determine the impact of PD-1 knockdown on neuronal excitability. The electrical activities and sodium channel function of neurons were determined by whole-cell patch-clamp recordings. The interaction between PD-1 and α-6 subunit of human voltage-gated sodium channel (Nav1.6) was validated by performing co-immunostaining and co-immunoprecipitation (co-IP) experiments. RESULTS Our results reveal that PD-1 protein and mRNA levels were upregulated in lesion cores compared with perifocal tissues of surgically resected specimens from patients with intractable epilepsy. Furthermore, we show that anti-PD-1 treatment has anti-seizure effects both in vivo and in vitro. Then, we reveal that PD-1 blockade can alter the electrophysiological properties of sodium channels. Moreover, we reveal that PD-1 acts together with downstream SHP-1 to regulate sodium channel function and hence neuronal excitability. Further investigation suggests that there is a direct interaction between neuronal PD-1 and Nav1.6. CONCLUSION Our study reveals that neuronal PD-1 plays an important role in epilepsy and that anti-PD-1 treatment protects against seizures by suppressing sodium channel function, identifying anti-PD-1 treatment as a novel therapeutic strategy for epilepsy.
Collapse
Affiliation(s)
- Yuling Yang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zhiyun Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Shize Jiang
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Li Wan
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Min Jiang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yulong Wang
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jie Hu
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
11
|
Armstrong OJ, Neal ES, Vidovic D, Xu W, Borges K. Transient anticonvulsant effects of time-restricted feeding in the 6-Hz mouse model. Epilepsy Behav 2024; 151:109618. [PMID: 38184948 DOI: 10.1016/j.yebeh.2023.109618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/09/2024]
Abstract
INTRODUCTION Intermittent fasting enhances neural bioenergetics, is neuroprotective, and elicits antioxidant effects in various animal models. There are conflicting findings on seizure protection, where intermittent fasting regimens often cause severe weight loss resembling starvation which is unsustainable long-term. Therefore, we tested whether a less intensive intermittent fasting regimen such as time-restricted feeding (TRF) may confer seizure protection. METHODS Male CD1 mice were assigned to either ad libitum-fed control, continuous 8 h TRF, or 8 h TRF with weekend ad libitum food access (2:5 TRF) for one month. Body weight, food intake, and blood glucose levels were measured. Seizure thresholds were determined at various time points using 6-Hz and maximal electroshock seizure threshold (MEST) tests. Protein levels and mRNA expression of genes, enzyme activity related to glucose metabolism, as well as mitochondrial dynamics were assessed in the cortex and hippocampus. Markers of antioxidant defence were evaluated in the plasma, cortex, and liver. RESULTS Body weight gain was similar in the ad libitum-fed and TRF mouse groups. In both TRF regimens, blood glucose levels did not change between the fed and fasted state and were higher during fasting than in the ad libitum-fed groups. Mice in the TRF group had increased seizure thresholds in the 6-Hz test on day 15 and on day 19 in a second cohort of 2:5 TRF mice, but similar seizure thresholds at other time points compared to ad libitum-fed mice. Continuous TRF did not alter MEST seizure thresholds on day 28. Mice in the TRF group showed increased maximal activity of pyruvate dehydrogenase in the cortex, which was accompanied by increased protein levels of mitochondrial pyruvate carrier 1 in the cortex and hippocampus. There were no other major changes in protein or mRNA levels associated with energy metabolism and mitochondrial dynamics in the brain, nor markers of antioxidant defence in the brain, liver, or plasma. CONCLUSIONS Both continuous and 2:5 TRF regimens transiently increased seizure thresholds in the 6-Hz model at around 2 weeks, which coincided with stability of blood glucose levels during the fed and fasted periods. Our findings suggest that the lack of prolonged anticonvulsant effects in the acute electrical seizure models employed may be attributed to only modest metabolic and antioxidant adaptations found in the brain and liver. Our findings underscore the potential therapeutic value of TRF in managing seizure-related conditions.
Collapse
Affiliation(s)
- Oliver J Armstrong
- School of Biomedical Sciences, Skerman Building 65, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Elliott S Neal
- School of Biomedical Sciences, Skerman Building 65, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Diana Vidovic
- School of Biomedical Sciences, Medical Building 181, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Weizhi Xu
- School of Biomedical Sciences, Skerman Building 65, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Karin Borges
- School of Biomedical Sciences, Skerman Building 65, The University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
12
|
Leo A, Bosco F, Guarnieri L, De Sarro C, Rania V, Gallelli L, Citraro R, De Sarro G. Cenobamate enhances the anticonvulsant effect of other antiseizure medications in the DBA/2 mouse model of reflex epilepsy. Eur J Pharmacol 2024; 962:176222. [PMID: 38029871 DOI: 10.1016/j.ejphar.2023.176222] [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: 08/30/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Clinical studies documented that cenobamate (CNB) has a marked efficacy compared to other antiseizure medications (ASMs) in reducing focal seizures. To date, different aspects of CNB need to be clarified, including its efficacy against generalized seizures. Similarly, the pattern of drug-drug interactions between CNB and other ASMs also compels further investigation. This study aimed to detect the role of CNB on generalized seizures using the DBA/2 mouse model. We have also studied the effects of an adjunctive CNB treatment on the antiseizure properties of some ASMs against reflex seizures. The effects of this adjunctive treatment on motor performance, body temperature, and brain levels of ASMs were also evaluated. CNB was able to antagonize seizures in DBA/2 mice. CNB, at 5 mg/kg, enhanced the antiseizure activity of ASMs, such as diazepam, clobazam, levetiracetam, perampanel, phenobarbital, topiramate, and valproate. No synergistic effects were observed when CNB was co-administered with some Na+ channel blockers. The increase in antiseizure activity was associated with a comparable intensification in motor impairment; however, the therapeutic index of combined treatment of ASMs with CNB was more favorable than the combination with vehicle except for carbamazepine, phenytoin, and oxcarbazepine. Since CNB did not significantly influence the brain levels of the ASMs studied, we suggest that pharmacokinetic interactions seem not probable. Overall, this study shows the ability of CNB to counteract generalized reflex seizures in mice. Moreover, our data documented an evident synergistic antiseizure effect for the combination of CNB with ASMs including phenobarbital, benzodiazepines, valproate, perampanel, topiramate, and levetiracetam.
Collapse
Affiliation(s)
- Antonio Leo
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Francesca Bosco
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Lorenza Guarnieri
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Caterina De Sarro
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Vincenzo Rania
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Luca Gallelli
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Rita Citraro
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| | - Giovambattista De Sarro
- Department of Health Sciences, School of Medicine and Surgery, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
13
|
Yang CS, Wu MC, Lai MC, Wu SN, Huang CW. Identification of New Antiseizure Medication Candidates in Preclinical Animal Studies. Int J Mol Sci 2023; 24:13143. [PMID: 37685950 PMCID: PMC10487685 DOI: 10.3390/ijms241713143] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Epilepsy is a multifactorial neurologic disease that often leads to many devastating disabilities and an enormous burden on the healthcare system. Until now, drug-resistant epilepsy has presented a major challenge for approximately 30% of the epileptic population. The present article summarizes the validated rodent models of seizures employed in pharmacological researches and comprehensively reviews updated advances of novel antiseizure candidates in the preclinical phase. Newly discovered compounds that demonstrate antiseizure efficacy in preclinical trials will be discussed in the review. It is inspiring that several candidates exert promising antiseizure activities in drug-resistant seizure models. The representative compounds consist of derivatives of hybrid compounds that integrate multiple approved antiseizure medications, novel positive allosteric modulators targeting subtype-selective γ-Aminobutyric acid type A receptors, and a derivative of cinnamamide. Although the precise molecular mechanism, pharmacokinetic properties, and safety are not yet fully clear in every novel antiseizure candidate, the adapted approaches to design novel antiseizure medications provide new insights to overcome drug-resistant epilepsy.
Collapse
Affiliation(s)
- Chih-Sheng Yang
- Department of Neurology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung City 42743, Taiwan;
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien City 97004, Taiwan
| | - Man-Chun Wu
- Department of Family Medicine and Preventive Medicine Center, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung City 42743, Taiwan
| | - Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Medical Center, Tainan City 71004, Taiwan;
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan
| |
Collapse
|
14
|
Lehmann LM, Barker-Haliski M. Loss of normal Alzheimer's disease-associated Presenilin 2 function alters antiseizure medicine potency and tolerability in the 6-Hz focal seizure model. Front Neurol 2023; 14:1223472. [PMID: 37592944 PMCID: PMC10427874 DOI: 10.3389/fneur.2023.1223472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Patients with early-onset Alzheimer's disease (EOAD) experience seizures and subclinical epileptiform activity, which may accelerate cognitive and functional decline. Antiseizure medicines (ASMs) may be a tractable disease-modifying strategy; numerous ASMs are marketed with well-established safety. However, little information is available to guide ASM selection as few studies have rigorously quantified ASM potency and tolerability in traditional seizure models in rodents with EOAD-associated risk factors. Presenilin 2 (PSEN2) variants evoke EOAD, and these patients experience seizures. This study thus established the anticonvulsant profile of mechanistically distinct ASMs in the frontline 6-Hz limbic seizure test evoked in PSEN2-knockout (KO) mice to better inform seizure management in EOAD. Methods The median effective dose (ED50) of prototype ASMs was quantified in the 6-Hz test in male and female PSEN2-KO and wild-type (WT) C57BL/6J mice (3-4 months old). Minimal motor impairment (MMI) was assessed to estimate a protective index (PI). Immunohistological detection of cFos established the extent to which 6-Hz stimulation activates discrete brain regions in KO vs. WT mice. Results There were significant genotype-related differences in the potency and tolerability of several ASMs. Valproic acid and levetiracetam were significantly more potent in male KO than in WT mice. Additionally, high doses of valproic acid significantly worsened MMI in KO mice. Conversely, carbamazepine was significantly less potent in female KO vs. WT mice. In both male and female KO mice vs. WTs, perampanel and lamotrigine were equally potent. However, there were marked genotype-related shifts in PI of both carbamazepine and perampanel, with KO mice exhibiting less MMI at the highest doses tested. Gabapentin was ineffective against 6-Hz seizures in KO mice vs. WTs without MMI changes. Neuronal activation 90 min following 6-Hz stimulation was significantly increased in the posterior parietal association cortex overlying CA1 and in the piriform cortex of WT mice, while stimulation-induced increases in cFos immunoreactivity were absent in KO mice. Discussion Acute ASM potency and tolerability in the high-throughput 6-Hz test may be significantly altered with loss of normal PSEN2 function. Seizures in discrete EOAD populations may benefit from precisely selected medicines optimized for primary ASM pharmacological mechanisms.
Collapse
Affiliation(s)
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
Sullivan KA, Vitko I, Blair K, Gaykema RP, Failor MJ, San Pietro JM, Dey D, Williamson JM, Stornetta RL, Kapur J, Perez-Reyes E. Drug-Inducible Gene Therapy Effectively Reduces Spontaneous Seizures in Kindled Rats but Creates Off-Target Side Effects in Inhibitory Neurons. Int J Mol Sci 2023; 24:11347. [PMID: 37511107 PMCID: PMC10379297 DOI: 10.3390/ijms241411347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Over a third of patients with temporal lobe epilepsy (TLE) are not effectively treated with current anti-seizure drugs, spurring the development of gene therapies. The injection of adeno-associated viral vectors (AAV) into the brain has been shown to be a safe and viable approach. However, to date, AAV expression of therapeutic genes has not been regulated. Moreover, a common property of antiepileptic drugs is a narrow therapeutic window between seizure control and side effects. Therefore, a long-term goal is to develop drug-inducible gene therapies that can be regulated by clinically relevant drugs. In this study, a first-generation doxycycline-regulated gene therapy that delivered an engineered version of the leak potassium channel Kcnk2 (TREK-M) was injected into the hippocampus of male rats. Rats were electrically stimulated until kindled. EEG was monitored 24/7. Electrical kindling revealed an important side effect, as even low expression of TREK M in the absence of doxycycline was sufficient to cause rats to develop spontaneous recurring seizures. Treating the epileptic rats with doxycycline successfully reduced spontaneous seizures. Localization studies of infected neurons suggest seizures were caused by expression in GABAergic inhibitory neurons. In contrast, doxycycline increased the expression of TREK-M in excitatory neurons, thereby reducing seizures through net inhibition of firing. These studies demonstrate that drug-inducible gene therapies are effective in reducing spontaneous seizures and highlight the importance of testing for side effects with pro-epileptic stressors such as electrical kindling. These studies also show the importance of evaluating the location and spread of AAV-based gene therapies in preclinical studies.
Collapse
Affiliation(s)
- Kyle A Sullivan
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Iuliia Vitko
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | - Kathryn Blair
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | - Ronald P Gaykema
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | - Madison J Failor
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | | | - Deblina Dey
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | - John M Williamson
- Department of Neurology, University of Virginia, Charlottesville, VA 22980, USA
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22980, USA
- UVA Brain Institute, University of Virginia, Charlottesville, VA 22980, USA
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22980, USA
- UVA Brain Institute, University of Virginia, Charlottesville, VA 22980, USA
| |
Collapse
|
16
|
Mohamed AM, Ali DA, Kolieb E, Abdelaziz EZ. Ceftriaxone and selenium mitigate seizures and neuronal injury in pentylenetetrazole-kindled rats: Oxidative stress and inflammatory pathway. Int Immunopharmacol 2023; 120:110304. [PMID: 37224649 DOI: 10.1016/j.intimp.2023.110304] [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: 02/09/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Epilepsy is one of the most serious worldwide neurological disorders that lead to the cognitive-psychosocial insults in recurrent seizures. About one third of the patients are drug-resistant, so innovative drugs are needed to manage seizures to improve the quality of life. Ceftriaxone is a cephalosporin antibiotic that increases the expression of glutamate transporters-1 and improves the neurobehavioral effects caused by increased glutamate level in the CNS. Selenium is well known antioxidant. The present study aimed to investigate ceftriaxone and selenium therapeutic effects against epilepsy in rats. Epilepsy was induced by PTZ given at a dose (50 mg/kg I.P) on alternative days for 13 days. Eighty rats were randomly divided into 8 groups: Group1-2; normal and vehicle control, Group 3; PTZ group, Group 4-8; kindled rats received selenium, ceftriaxone100, ceftriaxone200, selenium + ceftriaxone100 and selenium + ceftriaxone200 mg/kg/day respectively for a week. At the end of the study, behavioral tests were performed. Oxidative stress, inflammatory markers, neurotransmitters and GLT-1 were measured in brain tissue homogenate. Brain histopathological investigation was also done. PTZ-kindled rats exhibited increased Racine score, besides behavioral tests and histopathological changes, significant elevation in oxidative stress and inflammatory markers, with decrease in serotonin, dopamine, GABA levels and GLT-1 expressions. Selenium and Ceftriaxone alone or combined treatment decreased Racine score with remarkable improvement in behavioral and histopathological changes. The antioxidant enzymes, neurotransmitters and GLT-1 expressions were increased, along with reduced TNF-α, IL-1 levels. Current study showed that selenium + ceftriaxone100 group represents a possible approach to improve epilepsy particularly through inhibiting oxidative stress and inflammation.
Collapse
Affiliation(s)
- Asmaa M Mohamed
- Department of Pharmacology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Dina A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Eman Kolieb
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Eman Z Abdelaziz
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
17
|
Łukawski K, Czuczwar SJ. Oxidative Stress and Neurodegeneration in Animal Models of Seizures and Epilepsy. Antioxidants (Basel) 2023; 12:antiox12051049. [PMID: 37237916 DOI: 10.3390/antiox12051049] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Free radicals are generated in the brain, as well as in other organs, and their production is proportional to the brain activity. Due to its low antioxidant capacity, the brain is particularly sensitive to free radical damage, which may affect lipids, nucleic acids, and proteins. The available evidence clearly points to a role for oxidative stress in neuronal death and pathophysiology of epileptogenesis and epilepsy. The present review is devoted to the generation of free radicals in some animal models of seizures and epilepsy and the consequences of oxidative stress, such as DNA or mitochondrial damage leading to neurodegeneration. Additionally, antioxidant properties of antiepileptic (antiseizure) drugs and a possible use of antioxidant drugs or compounds in patients with epilepsy are reviewed. In numerous seizure models, the brain concentration of free radicals was significantly elevated. Some antiepileptic drugs may inhibit these effects; for example, valproate reduced the increase in brain malondialdehyde (a marker of lipid peroxidation) concentration induced by electroconvulsions. In the pentylenetetrazol model, valproate prevented the reduced glutathione concentration and an increase in brain lipid peroxidation products. The scarce clinical data indicate that some antioxidants (melatonin, selenium, vitamin E) may be recommended as adjuvants for patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| |
Collapse
|
18
|
Zhuang XF, Liu YX, Yang ZH, Gao Q, Wang L, Ju C, Wang K. Attenuation of Epileptogenesis and Cognitive Deficits by a Selective and Potent Kv7 Channel Opener in Rodent Models of Seizures. J Pharmacol Exp Ther 2023; 384:315-325. [PMID: 36396352 DOI: 10.1124/jpet.122.001328] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/12/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
Targeting neuronal Kv7 channels by pharmacological activation has been proven to be an attractive therapeutic strategy for epilepsy. Here, we show that activation of Kv7 channels by an opener SCR2682 dose-dependently reduces seizure activity and severity in rodent models of epilepsy induced by a GABAa receptor antagonist pentylenetetrazole (PTZ), maximal electroshock, and a glutamate receptor agonist kainic acid (KA). Electroencephalographic recordings of rat cerebral cortex confirm that SCR2682 also decreases epileptiform discharges in KA-induced seizures. Nissl and neuronal nuclei staining further demonstrates that SCR2682 also protects neurons from injury induced by KA. In Morris water maze navigation and Y-maze tests, SCR2682 improves PTZ- and KA-induced cognitive impairment. Taken together, our findings demonstrate that pharmacological activation of Kv7 by novel opener SCR2682 may hold promise for therapy of epilepsy with cognitive impairment. SIGNIFICANCE STATEMENT: A neuronal Kv7 channel opener SCR2682 attenuates epileptogenesis and seizure-induced cognitive impairment in rodent models of seizures, thus possessing a developmental potential for effective therapy of epilepsy with cognitive impairment.
Collapse
Affiliation(s)
- Xiao-Fei Zhuang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - Yu-Xue Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - Zhi-Hong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - Qin Gao
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - Lei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College (X.-F.Z., Y.-X.L., Z.-H.Y., Q.G., L.W., C.J., K.W.) and Institute of Innovative Drugs, Qingdao University, Qingdao, China (K.W.)
| |
Collapse
|
19
|
Li J, Ma G, Yan M, Liu H, Zhang K, Lin R, Lü G. Subcutaneous infection mouse model could be applied into real time monitoring the efficacy of anti-cystic echinococcosis drug in vivo. Acta Trop 2023; 238:106802. [PMID: 36565837 DOI: 10.1016/j.actatropica.2022.106802] [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: 05/31/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Cystic echinococcosis (CE) is a zoonotic parasitic disease with a cosmopolitan distribution, and it is urgent to develop novel anti-helminthic agents. The intraperitoneal (ip) infection mice model was widely used to evaluate the efficacy of potential anti-CE compounds. Still, it's time-consuming, and the inability to achieve real-time monitoring hinders the development of potential anti-CE compounds. In this study, a CE mouse model was established by subcutaneous (sc) injection of protoscoleces of Echinococcus granulosus sensu stricto (E.granulosus s.s.) and used to assess the efficiency and efficacy of prospective anti-CE drugs. Compared to the ip infection CE mice model, the lesion volume of sc infection protoscoleces of E.granulosus s.s. (EgPSCs) could be measured by vernier caliper at week 6 post-infection. In contrast, the lesion volume of ip infection CE mice model was detected by ultrasound-assisted diagnosis at week 16 post-infection. Oral administration of albendazole (ABZ) could reduce cystic weight by 32.17% and 17.61%, the cystic number by 12.24% and 25.19%, and damage the ultrastructure of the cysts of E. granulosus s.s. in the sc and ip infection group, respectively. Furthermore, we found that the sc infection mice model could real-time monitor the lesion volume of E. granulosus s.s. during the ABZ and everolimus treatment. Therefore, we consider that the sc infection CE mice model is an assistant tool for screening and developing potential anti-CE compounds.
Collapse
Affiliation(s)
- Jintian Li
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; College of pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guizhi Ma
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; College of pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mingzhi Yan
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; College of pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hui Liu
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Kainan Zhang
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Renyong Lin
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Guodong Lü
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, China; Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; College of pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
20
|
Barker-Haliski M, Pitsch J, Galanopoulou AS, Köhling R. A companion to the preclinical common data elements for phenotyping seizures and epilepsy in rodent models. A report of the TASK3-WG1C: Phenotyping working group of the ILAE/AES joint translational task force. Epilepsia Open 2022. [PMID: 36461665 DOI: 10.1002/epi4.12676] [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: 11/18/2021] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Epilepsy is a heterogeneous disorder characterized by spontaneous seizures and behavioral comorbidities. The underlying mechanisms of seizures and epilepsy across various syndromes lead to diverse clinical presentation and features. Similarly, animal models of epilepsy arise from numerous dissimilar inciting events. Preclinical seizure and epilepsy models can be evoked through many different protocols, leaving the phenotypic reporting subject to diverse interpretations. Serendipity can also play an outsized role in uncovering novel drivers of seizures or epilepsy, with some investigators even stumbling into epilepsy research because of a new genetic cross or unintentional drug effect. The heightened emphasis on rigor and reproducibility in preclinical research, including that which is conducted for epilepsy, underscores the need for standardized phenotyping strategies. To address this goal as part of the TASK3-WG1C Working Group of the International League Against Epilepsy (ILAE)/American Epilepsy Society (AES) Joint Translational Task Force, we developed a case report form (CRF) to describe the common data elements (CDEs) necessary for the phenotyping of seizure-like behaviors in rodents. This companion manuscript describes the use of the proposed CDEs and CRF for the visual, behavioral phenotyping of seizure-like behaviors. These phenotyping CDEs and accompanying CRF can be used in parallel with video-electroencephalography (EEG) studies or as a first visual screen to determine whether a model manifests seizure-like behaviors before utilizing more specialized diagnostic tests, like video-EEG. Systematic logging of seizure-like behaviors may help identify models that could benefit from more specialized diagnostic tests to determine whether these are epileptic seizures, such as video-EEG.
Collapse
Affiliation(s)
- Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Julika Pitsch
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Isabelle Rapin Division of Child Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Rüdiger Köhling
- Oscar-Langendorff-Institut für Physiologie, Universitätsmedizin Rostock, Rostock, Germany
| |
Collapse
|
21
|
Birhan YS. Medicinal plants utilized in the management of epilepsy in Ethiopia: ethnobotany, pharmacology and phytochemistry. Chin Med 2022; 17:129. [PMCID: PMC9675240 DOI: 10.1186/s13020-022-00686-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022] Open
Abstract
Epilepsy is a common central nervous system (CNS) disorder that affects 50 million people worldwide. Patients with status epilepticus (SE) suffer from devastating comorbidities and a high incidence of mortalities. Antiepileptic drugs (AEDs) are the mainstream treatment options for the symptomatic relief of epilepsy. The incidence of refractory epilepsy and the dose-dependent neurotoxicity of AEDs such as fatigue, cognitive impairment, dizziness, attention-deficit behavior, and other side effects are the major bottlenecks in epilepsy treatment. In low- and middle-income countries (LMICs), epilepsy patients failed to adhere to the AEDs regimens and consider other options such as complementary and alternative medicines (CAMs) to relieve pain due to status epilepticus (SE). Plant-based CAMs are widely employed for the treatment of epilepsy across the globe including Ethiopia. The current review documented around 96 plant species (PS) that are often used for the treatment of epilepsy in Ethiopia. It also described the in vivo anticonvulsant activities and toxicity profiles of the antiepileptic medicinal plants (MPs). Moreover, the phytochemical constituents of MPs with profound anticonvulsant effects were also assessed. The result reiterated that a lot has to be done to show the association between herbal-based epilepsy treatment and in vivo pharmacological activities of MPs regarding their mechanism of action (MOA), toxicity profiles, and bioactive constituents so that they can advance into the clinics and serve as a treatment option for epilepsy.
Collapse
Affiliation(s)
- Yihenew Simegniew Birhan
- grid.449044.90000 0004 0480 6730Department of Chemistry, College of Natural and Computational Sciences, Debre Markos University, P.O. Box 269, Debre Markos, Ethiopia
| |
Collapse
|
22
|
Rodent Models of Audiogenic Epilepsy: Genetic Aspects, Advantages, Current Problems and Perspectives. Biomedicines 2022; 10:biomedicines10112934. [PMID: 36428502 PMCID: PMC9687921 DOI: 10.3390/biomedicines10112934] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Animal models of epilepsy are of great importance in epileptology. They are used to study the mechanisms of epileptogenesis, and search for new genes and regulatory pathways involved in the development of epilepsy as well as screening new antiepileptic drugs. Today, many methods of modeling epilepsy in animals are used, including electroconvulsive, pharmacological in intact animals, and genetic, with the predisposition for spontaneous or refractory epileptic seizures. Due to the simplicity of manipulation and universality, genetic models of audiogenic epilepsy in rodents stand out among this diversity. We tried to combine data on the genetics of audiogenic epilepsy in rodents, the relevance of various models of audiogenic epilepsy to certain epileptic syndromes in humans, and the advantages of using of rodent strains predisposed to audiogenic epilepsy in current epileptology.
Collapse
|
23
|
Neuroprotective effects of Lasmiditan and Sumatriptan in an experimental model of post-stroke seizure in mice: Higher effects with concurrent opioid receptors or K ATP channels inhibitors. Toxicol Appl Pharmacol 2022; 454:116254. [PMID: 36155770 DOI: 10.1016/j.taap.2022.116254] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/29/2022] [Accepted: 09/18/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Early post-stroke seizure frequently occurs in stroke survivors within the first few days and is associated with poor functional outcomes. Therefore, efficient treatments of such complications with less adverse effects are pivotal. In this study, we investigated the possible beneficial effects of lasmiditan and sumatriptan against post-stroke seizures in mice and explored underlying mechanisms in their effects. METHODS Stroke was induced by double ligation of the right common carotid artery in mice. Immediately after the ligation, lasmiditan (0.1 mg/kg, intraperitoneally [i.p.]) or sumatriptan (0.03 mg/kg, i.p.) were administered. Twenty-four hours after the stroke induction, seizure susceptibility was evaluated using the pentylenetetrazole (PTZ)-induced clonic seizure model. In separate experiments, naltrexone (a non-specific opioid receptor antagonist) and glibenclamide (a KATP channel blocker) were administered 15 min before lasmiditan or sumatriptan injection. To evaluate the underlying signaling pathways, ELISA analysis of inflammatory cytokines (TNF-α and IL-1β) and western blot analysis of anti- and pro-apoptotic markers (Bcl-2 and Bax) were performed on mice isolated brain tissues. RESULTS Lasmiditan (0.1 mg/kg, i.p.) and sumatriptan (0.03 mg/kg, i.p.) remarkably decreased seizure susceptibility in stroke animals by reducing inflammatory cytokines and neuronal apoptosis. Concurrent administration of naltrexone (10 mg/kg, i.p.) or glibenclamide (0.3 mg/kg, i.p.) with lasmiditan or sumatriptan resulted in a higher neuroprotection against clonic seizures and efficiently reduced the inflammatory and apoptotic markers. CONCLUSION Lasmiditan and sumatriptan significantly increased post-stroke seizure thresholds in mice by suppressing inflammatory cytokines and neuronal apoptosis. Lasmiditan and sumatriptan seem to exert higher effects on seizure threshold with concurrent administration of the opioid receptors or KATP channels modulators.
Collapse
|
24
|
Aboutabl ME, Elkhateeb WA, Masoud MA, Daba GM, Afifi AH, Hussein RA. HPLC and GC-MS based metabolic profiles and in vivo anticonvulsant, sedative, and antinociceptive potentials of truffles Tirmania nivea and Tirmania pinoyi hydromethanolic extracts in mice. Biomed Chromatogr 2022; 36:e5481. [PMID: 35971328 DOI: 10.1002/bmc.5481] [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: 06/05/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022]
Abstract
GC-MS and HPLC analyses of the hydromethanolic extracts of the truffles Tirmania nivea (TN) and Tirmania pinoyi (TP) revealed the presence of 18 metabolites and 11 polyphenols, respectively. In vivo, TP extract protected against subcutaneous pentylenetetrazole (scPTZ) and maximal electric shock (MES)-induced convulsions faster than TN. TP (100 and 300 mg/kg) showed 100% protection and longer duration than TN in the scPTZ test. Similarly, at 300 mg/kg, TP demonstrated a quicker start (75%) and longer duration of action (100%) than TN in MES test. In scPTZ test, ED50 of TP demonstrated greater anticonvulsant efficacy than TN. In mice given TP and TN treatments, the brain GABA levels were noticeably increased. TP (100 and 300mg/kg) produced a notable sedative effect in open field test, whereas TN (100 or 300 mg/kg) and TP (300 mg/kg) reduced sleep latency by 79, 52, and 45%, respectively. In writhing test, TN (100 or 300mg/kg) significantly enhanced analgesic efficacy by 50 and 87%, respectively. Comparatively, in formalin test, TP and TN at a dosage of 300 mg/kg decreased the length of the licking by 34 and 59%, respectively. For the first time, this study explains the anticonvulsant, sedative, central, and peripheral analgesic activities of truffle extracts.
Collapse
Affiliation(s)
- Mona E Aboutabl
- Medicinal and Pharmaceutical Chemistry Department (Pharmacology Group), Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Waill A Elkhateeb
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Marwa A Masoud
- Pharmacology Department, National Organization for Drug Control and Research, Egyptian Drug Authority (EDA), Giza, Egypt
| | - Ghoson M Daba
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Ahmed H Afifi
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Rehab A Hussein
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| |
Collapse
|
25
|
Mantegazza M, Auvin S, Barker-Haliski M, Katsarou AM, Kubova H, Galanopoulou AS, Semple B, Reid CA. A companion to the preclinical common data elements for rodent genetic epilepsy models. A report of the TASK3-WG1B: Paediatric and genetic models working group of the ILAE/AES joint translational TASK force. Epilepsia Open 2022. [PMID: 35951766 DOI: 10.1002/epi4.12642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/11/2022] [Indexed: 11/06/2022] Open
Abstract
Rodent models of epilepsy remain the cornerstone of research into the mechanisms underlying genetic epilepsy. Reproducibility of experiments using these rodent models, occurring across a diversity of laboratories and commercial vendors, remains an issue impacting the cost-effectiveness and scientific rigor of the studies performed. Here, we present two case report forms (CRFs) describing common data elements (CDE) for genetic rodent models, developed by the TASK3-WG1B Working Group of the International League Against Epilepsy (ILAE)/American Epilepsy Society (AES) Joint Translational Task Force. The first CRF relates to genetic rodent models that have been engineered based on variants described in epilepsy patients. The second CRF encompasses both spontaneous and inbred rodent models. This companion piece describes the elements and discusses the important factors to consider before documenting each required element. These CRFs provide tools that allow investigators to more uniformly describe core experimental data on different genetic models across laboratories, with the aim of improving experimental reproducibility and thus translational impact of such studies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Côte d'Azur, CNRS UMR7275, Inserm, LabEx ICST, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Stėphane Auvin
- Université de Paris, INSERM UMR 1141, Service de Neurologie Pédiatrique, Hôpital Robert-Debré, APHP, Paris, France
- Institut Universitaire de France (IUF), Paris, France
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Anna-Maria Katsarou
- Laboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Hana Kubova
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Aristea S Galanopoulou
- Laboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Laboratory of Developmental Epilepsy, Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, Dominique P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bridgette Semple
- Department of Neuroscience, Monash University, Prahran, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher A Reid
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
26
|
del Pozo A, Lehmann L, Knox KM, Barker-Haliski M. Can Old Animals Reveal New Targets? The Aging and Degenerating Brain as a New Precision Medicine Opportunity for Epilepsy. Front Neurol 2022; 13:833624. [PMID: 35572927 PMCID: PMC9096090 DOI: 10.3389/fneur.2022.833624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/07/2022] [Indexed: 02/02/2023] Open
Abstract
Older people represent the fastest growing group with epilepsy diagnosis. For example, cerebrovascular disease may underlie roughly 30-50% of epilepsy in older adults and seizures are also an underrecognized comorbidity of Alzheimer's disease (AD). As a result, up to 10% of nursing home residents may take antiseizure medicines (ASMs). Despite the greater incidence of epilepsy in older individuals and increased risk of comorbid seizures in people with AD, aged animals with seizures are strikingly underrepresented in epilepsy drug discovery practice. Increased integration of aged animals into preclinical epilepsy drug discovery could better inform the potential tolerability and pharmacokinetic interactions in aged individuals as the global population becomes increasingly older. Quite simply, the ASMs on the market today were brought forth based on efficacy in young adult, neurologically intact rodents; preclinical information concerning the efficacy and safety of promising ASMs is not routinely evaluated in aged animals. Integrating aged animals more often into basic epilepsy research may also uncover novel treatments for hyperexcitability. For example, cannabidiol and fenfluramine demonstrated clear efficacy in syndrome-specific pediatric models that led to a paradigm shift in the perceived value of pediatric models for ASM discovery practice; aged rodents with seizures or rodents with aging-related neuropathology represent an untapped resource that could similarly change epilepsy drug discovery. This review, therefore, summarizes how aged rodent models have thus far been used for epilepsy research, what studies have been conducted to assess ASM efficacy in aged rodent seizure and epilepsy models, and lastly to identify remaining gaps to engage aging-related neurological disease models for ASM discovery, which may simultaneously reveal novel mechanisms associated with epilepsy.
Collapse
Affiliation(s)
| | | | | | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| |
Collapse
|
27
|
Effect of caffeine on the anticonvulsant action of pregabalin against electroconvulsions in mice. Pharmacol Rep 2022; 74:431-437. [PMID: 35171455 DOI: 10.1007/s43440-022-00356-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/24/2022] [Accepted: 02/02/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Experimental data indicate that caffeine (CAF) can reduce the anticonvulsant activity of antiepileptic drugs (AEDs) in animal models of seizures. The purpose of the current study was to examine the effect of CAF on the protective action of pregabalin (PGB) against electroconvulsions in mice. METHODS Maximal electroshock seizure (MES) test was used in the current study. In addition, the combined treatment with CAF and PGB was assessed in the passive avoidance task (long-term memory) and the chimney test (motor coordination). Drugs were injected intraperitoneally (ip) as single injections. CAF was administered at doses reported to compromise the anticonvulsant action of AEDs in mice. RESULTS CAF at a dose of 23.1 mg/kg reduced the anticonvulsant action of PGB in the MES test. The brain concentration of PGB was not significantly changed by CAF and vice versa. In the chimney test, CAF (23.1 mg/kg) protected mice against PGB-induced motor coordination impairment. CONCLUSIONS Regarding seizure control, it might be suggested that patients with epilepsy treated with PGB should avoid taking CAF. The estimated total brain concentration of PGB and CAF does not suggest a pharmacokinetic interaction as an explanation for these results.
Collapse
|
28
|
Łukawski K, Czuczwar SJ. Emerging therapeutic targets for epilepsy: Preclinical insights. Expert Opin Ther Targets 2022; 26:193-206. [PMID: 35130119 DOI: 10.1080/14728222.2022.2039120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Around 30% of patients with epilepsy suffer from drug-resistant seizures. Drug-resistant seizures may have significant consequences such as sudden death in epilepsy, injuries, memory disturbances, and childhood learning and developmental problems. Conventional and newer available antiepileptic drugs (AEDs) work via numerous mechanisms - mainly through inhibition of voltage-operated Na+ and/or Ca2+ channels, excitation of K+ channels, enhancement of GABA-mediated inhibition and/or blockade of glutamate-produced excitation. However, the discovery and development of novel brain targets may improve the future pharmacological management of epilepsy and hence is of pivotal importance. AREAS COVERED This article examines novel drug targets such as brain multidrug efflux transporters and inflammatory pathways; it progresses to discuss possible strategies for the management of drug-resistant seizures. Reduction of the consequences of blood brain barrier dysfunction and enhancement of anti-oxidative defense are discussed. EXPERT OPINION Novel drug targets comprise brain multidrug efflux transporters, TGF-β, Nrf2-ARE or m-TOR signaling and inflammatory pathways. Gene therapy and antagomirs seem the most promising targets. Epileptic foci may be significantly suppressed by viral-vector-mediated gene transfer, leading to an increased in situ concentration of inhibitory factors (for instance, galanin). Also, antagomirs offer a promising possibility of seizure inhibition by silencing micro-RNAs involved in epileptogenesis and possibly in seizure generation.
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland.,Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Physiopathology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland.,Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| |
Collapse
|
29
|
Wang Y, Wei P, Yan F, Luo Y, Zhao G. Animal Models of Epilepsy: A Phenotype-oriented Review. Aging Dis 2022; 13:215-231. [PMID: 35111370 PMCID: PMC8782545 DOI: 10.14336/ad.2021.0723] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022] Open
Abstract
Epilepsy is a serious neurological disorder characterized by abnormal, recurrent, and synchronous discharges in the brain. Long-term recurrent seizure attacks can cause serious damage to brain function, which is usually observed in patients with temporal lobe epilepsy. Controlling seizure attacks is vital for the treatment and prognosis of epilepsy. Animal models, such as the kindling model, which was the most widely used model in the past, allow the understanding of the potential epileptogenic mechanisms and selection of antiepileptic drugs. In recent years, various animal models of epilepsy have been established to mimic different seizure types, without clear merits and demerits. Accordingly, this review provides a summary of the views mentioned above, aiming to provide a reference for animal model selection.
Collapse
Affiliation(s)
- Yilin Wang
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Penghu Wei
- 1Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| | - Feng Yan
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yumin Luo
- 2Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| | - Guoguang Zhao
- 1Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.,3Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,4Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Cho C, Zeigler M, Mizuno S, Morrison RS, Totah RA, Barker-Haliski M. Reductions in Hydrogen Sulfide and Changes in Mitochondrial Quality Control Proteins Are Evident in the Early Phases of the Corneally Kindled Mouse Model of Epilepsy. Int J Mol Sci 2022; 23:ijms23031434. [PMID: 35163358 PMCID: PMC8835945 DOI: 10.3390/ijms23031434] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Epilepsy is a heterogenous neurological disorder characterized by recurrent unprovoked seizures, mitochondrial stress, and neurodegeneration. Hydrogen sulfide (H2S) is a gasotransmitter that promotes mitochondrial function and biogenesis, elicits neuromodulation and neuroprotection, and may acutely suppress seizures. A major gap in knowledge remains in understanding the role of mitochondrial dysfunction and progressive changes in H2S levels following acute seizures or during epileptogenesis. We thus sought to quantify changes in H2S and its methylated metabolite (MeSH) via LC-MS/MS following acute maximal electroshock and 6 Hz 44 mA seizures in mice, as well as in the early phases of the corneally kindled mouse model of chronic seizures. Plasma H2S was acutely reduced after a maximal electroshock seizure. H2S or MeSH levels and expressions of related genes in whole brain homogenates from corneally kindled mice were not altered. However, plasma H2S levels were significantly lower during kindling, but not after established kindling. Moreover, we demonstrated a time-dependent increase in expression of mitochondrial membrane integrity-related proteins, OPA1, MFN2, Drp1, and Mff during kindling, which did not correlate with changes in gene expression. Taken together, short-term reductions in plasma H2S could be a novel biomarker for seizures. Future studies should further define the role of H2S and mitochondrial stress in epilepsy.
Collapse
Affiliation(s)
- Christi Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Maxwell Zeigler
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Stephanie Mizuno
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA;
| | | | - Rheem A. Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Melissa Barker-Haliski
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA;
- Correspondence: ; Tel.: +1-206-685-1783
| |
Collapse
|
31
|
Roy A, Han VZ, Bard AM, Wehle DT, Smith SEP, Ramirez JM, Kalume F, Millen KJ. Non-synaptic Cell-Autonomous Mechanisms Underlie Neuronal Hyperactivity in a Genetic Model of PIK3CA-Driven Intractable Epilepsy. Front Mol Neurosci 2021; 14:772847. [PMID: 34899181 PMCID: PMC8662737 DOI: 10.3389/fnmol.2021.772847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Patients harboring mutations in the PI3K-AKT-MTOR pathway-encoding genes often develop a spectrum of neurodevelopmental disorders including epilepsy. A significant proportion remains unresponsive to conventional anti-seizure medications. Understanding mutation-specific pathophysiology is thus critical for molecularly targeted therapies. We previously determined that mouse models expressing a patient-related activating mutation in PIK3CA, encoding the p110α catalytic subunit of phosphoinositide-3-kinase (PI3K), are epileptic and acutely treatable by PI3K inhibition, irrespective of dysmorphology. Here we report the physiological mechanisms underlying this dysregulated neuronal excitability. In vivo, we demonstrate epileptiform events in the Pik3ca mutant hippocampus. By ex vivo analyses, we show that Pik3ca-driven hyperactivation of hippocampal pyramidal neurons is mediated by changes in multiple non-synaptic, cell-intrinsic properties. Finally, we report that acute inhibition of PI3K or AKT, but not MTOR activity, suppresses the intrinsic hyperactivity of the mutant neurons. These acute mechanisms are distinct from those causing neuronal hyperactivity in other AKT-MTOR epileptic models and define parameters to facilitate the development of new molecularly rational therapeutic interventions for intractable epilepsy.
Collapse
Affiliation(s)
- Achira Roy
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, India
| | - Victor Z Han
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Biology, University of Washington, Seattle, WA, United States
| | - Angela M Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Devin T Wehle
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States.,Department of Neurological Surgery, University of Washington, Seattle, WA, United States.,Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - Franck Kalume
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Neurological Surgery, University of Washington, Seattle, WA, United States.,Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
32
|
Feja M, Meller S, Deking LS, Kaczmarek E, During MJ, Silverman RB, Gernert M. OV329, a novel highly potent γ-aminobutyric acid aminotransferase inactivator, induces pronounced anticonvulsant effects in the pentylenetetrazole seizure threshold test and in amygdala-kindled rats. Epilepsia 2021; 62:3091-3104. [PMID: 34617595 DOI: 10.1111/epi.17090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE An attractive target to interfere with epileptic brain hyperexcitability is the enhancement of γ-aminobutyric acidergic (GABAergic) inhibition by inactivation of the GABA-metabolizing enzyme GABA aminotransferase (GABA-AT). GABA-AT inactivators were designed to control seizures by raising brain GABA levels. OV329, a novel drug candidate for the treatment of epilepsy and addiction, has been shown in vitro to be substantially more potent as a GABA-AT inactivator than vigabatrin, an antiseizure drug approved as an add-on therapy for adult patients with refractory complex partial seizures and monotherapy for pediatric patients with infantile spasms. Thus, we hypothesized that OV329 should produce pronounced anticonvulsant effects in two different rat seizure models. METHODS We therefore examined the effects of OV329 (5, 20, and 40 mg/kg ip) on the seizure threshold of female Wistar Unilever rats, using the timed intravenous pentylenetetrazole (ivPTZ) seizure threshold model as a seizure test particularly sensitive to GABA-potentiating manipulations, and amygdala-kindled rats as a model of difficult-to-treat temporal lobe epilepsy. RESULTS GABA-AT inactivation by OV329 clearly increased the threshold of both ivPTZ-induced and amygdala-kindled seizures. OV329 further showed a 30-fold greater anticonvulsant potency on ivPTZ-induced myoclonic jerks and clonic seizures compared to vigabatrin investigated previously. Notably, all rats were responsive to OV329 in both seizure models. SIGNIFICANCE These results reveal an anticonvulsant profile of OV329 that appears to be superior in both potency and efficacy to vigabatrin and highlight OV329 as a highly promising candidate for the treatment of seizures and pharmacoresistant epilepsies.
Collapse
Affiliation(s)
- Malte Feja
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Sebastian Meller
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Lillian S Deking
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Edith Kaczmarek
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Richard B Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA.,Department of Pharmacology, Chemistry of Life Processes Institute, Northwestern University, Chicago, Illinois, USA
| | - Manuela Gernert
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
33
|
Łukawski K, Czuczwar SJ. Understanding mechanisms of drug resistance in epilepsy and strategies for overcoming it. Expert Opin Drug Metab Toxicol 2021; 17:1075-1090. [PMID: 34310255 DOI: 10.1080/17425255.2021.1959912] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The present evidence indicates that approximately 70% of patients with epilepsy can be successfully treated with antiepileptic drugs (AEDs). A significant proportion of patients are not under sufficient control, and pharmacoresistant epilepsy is clearly associated with poor quality of life and increased morbidity and mortality. There is a great need for newer therapeutic options able to reduce the percentage of drug-resistant patients. AREAS COVERED A number of hypotheses trying to explain the development of pharmacoresistance have been put forward. These include: target hypothesis (altered AED targets), transporter (overexpression of brain efflux transporters), pharmacokinetic (overexpression of peripheral efflux transporters in the intestine or kidneys), intrinsic severity (initial high seizure frequency), neural network (aberrant networks), and gene variant hypothesis (genetic polymorphisms). EXPERT OPINION A continuous search for newer AEDs or among non-AEDs (blockers of efflux transporters, interleukin antagonists, cyclooxygenase inhibitors, mTOR inhibitors, angiotensin II receptor antagonists) may provide efficacious drugs for the management of drug-resistant epilepsy. Also, combinations of AEDs exerting synergy in preclinical and clinical studies (for instance, lamotrigine + valproate, levetiracetam + valproate, topiramate + carbamazepine) might be of importance in this respect. Preclinically antagonistic combinations must be avoided (lamotrigine + carbamazepine, lamotrigine + oxcarbazepine).
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland.,Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
34
|
Barker‐Haliski M, Knox K, Zierath D, Koneval Z, Metcalf C, Wilcox KS, White HS. Development of an antiepileptogenesis drug screening platform: Effects of everolimus and phenobarbital. Epilepsia 2021; 62:1677-1688. [PMID: 34080183 PMCID: PMC8260451 DOI: 10.1111/epi.16955] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The kainic acid (KA)-induced status epilepticus (SE) model in rats is a well-defined model of epileptogenesis. This model closely recapitulates many of the clinical and pathological characteristics of human temporal lobe epilepsy (TLE) that arise following SE or another neurological insult. Spontaneous recurrent seizures (SRS) in TLE can present after a latent period following a neurological insult (traumatic brain injury, SE event, viral infection, etc.). Moreover, this model is suitable for preclinical studies to evaluate the long-term process of epileptogenesis and screen putative disease-modifying/antiepileptogenic agents. The burden of human TLE is highly variable, similar to the post-KA SE rat model. In this regard, this model may have broad translational relevance. This report thus details the pharmacological characterization and methodological refinement of a moderate-throughput drug screening program using the post-KA-induced SE model of epileptogenesis in male Sprague Dawley rats to identify potential agents that may prevent or modify the burden of SRS. Specifically, we sought to demonstrate whether our protocol could prevent the development of SRS or lead to a reduced frequency/severity of SRS. METHODS Rats were administered either everolimus (2-3 mg/kg po) beginning 1, 2, or 24 h after SE onset, or phenobarbital (60 mg/kg ip) beginning 1 h after SE onset. All treatments were administered once/day for 5-7 days. Rats in all studies (n = 12/treatment dose/study) were then monitored intermittently by video-electroencephalography (2 weeks on, 2 weeks off, 2 weeks on epochs) to determine latency to onset of SRS and disease burden. RESULTS Although no adverse side effects were observed in our studies, no treatment significantly modified disease or prevented the presentation of SRS by 6 weeks after SE onset. SIGNIFICANCE Neither phenobarbital nor everolimus administered at several time points after SE onset prevented the development of SRS. Nonetheless, we demonstrate a practical and moderate-throughput screen for potential antiepileptogenic agents in a rat model of TLE.
Collapse
Affiliation(s)
| | - Kevin Knox
- Department of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | | | - Zachery Koneval
- Department of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Cameron Metcalf
- Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUtahUSA
| | - Karen S. Wilcox
- Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUtahUSA
| | - H. Steve White
- Department of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
35
|
Lehmann L, Lo A, Knox KM, Barker-Haliski M. Alzheimer's Disease and Epilepsy: A Perspective on the Opportunities for Overlapping Therapeutic Innovation. Neurochem Res 2021; 46:1895-1912. [PMID: 33929683 PMCID: PMC8254705 DOI: 10.1007/s11064-021-03332-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023]
Abstract
Early-onset Alzheimer's disease (AD) is associated with variants in amyloid precursor protein (APP) and presenilin (PSEN) 1 and 2. It is increasingly recognized that patients with AD experience undiagnosed focal seizures. These AD patients with reported seizures may have worsened disease trajectory. Seizures in epilepsy can also lead to cognitive deficits, neuroinflammation, and neurodegeneration. Epilepsy is roughly three times more common in individuals aged 65 and older. Due to the numerous available antiseizure drugs (ASDs), treatment of seizures has been proposed to reduce the burden of AD. More work is needed to establish the functional impact of seizures in AD to determine whether ASDs could be a rational therapeutic strategy. The efficacy of ASDs in aged animals is not routinely studied, despite the fact that the elderly represents the fastest growing demographic with epilepsy. This leaves a particular gap in understanding the discrete pathophysiological overlap between hyperexcitability and aging, and AD more specifically. Most of our preclinical knowledge of hyperexcitability in AD has come from mouse models that overexpress APP. While these studies have been invaluable, other drivers underlie AD, e.g. PSEN2. A diversity of animal models should be more frequently integrated into the study of hyperexcitability in AD, which could be particularly beneficial to identify novel therapies. Specifically, AD-associated risk genes, in particular PSENs, altogether represent underexplored contributors to hyperexcitability. This review assesses the available studies of ASDs administration in clinical AD populations and preclinical studies with AD-associated models and offers a perspective on the opportunities for further therapeutic innovation.
Collapse
Affiliation(s)
- Leanne Lehmann
- Undergraduate Neuroscience Program, University of Washington, Seattle, WA, 98195, USA
| | - Alexandria Lo
- Department of Public Health-Global Health, School of Public Health, University of Washington, Seattle, WA, 98195, USA
| | - Kevin M Knox
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
36
|
Acutain MF, Griebler Luft J, Vazquez CA, Popik B, Cercato MC, Epstein A, Salvetti A, Jerusalinsky DA, de Oliveira Alvares L, Baez MV. Reduced Expression of Hippocampal GluN2A-NMDAR Increases Seizure Susceptibility and Causes Deficits in Contextual Memory. Front Neurosci 2021; 15:644100. [PMID: 33897358 PMCID: PMC8064689 DOI: 10.3389/fnins.2021.644100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
N-methyl-D-aspartate receptors are heterotetramers composed of two GluN1 obligatory subunits and two regulatory subunits. In cognitive-related brain structures, GluN2A and GluN2B are the most abundant regulatory subunits, and their expression is subjected to tight regulation. During development, GluN2B expression is characteristic of immature synapses, whereas GluN2A is present in mature ones. This change in expression induces a shift in GluN2A/GluN2B ratio known as developmental switch. Moreover, modifications in this relationship have been associated with learning and memory, as well as different pathologies. In this work, we used a specific shRNA to induce a reduction in GluN2A expression after the developmental switch, both in vitro in primary cultured hippocampal neurons and in vivo in adult male Wistar rats. After in vitro characterization, we performed a cognitive profile and evaluated seizure susceptibility in vivo. Our in vitro results showed that the decrease in the expression of GluN2A changes GluN2A/GluN2B ratio without altering the expression of other regulatory subunits. Moreover, rats expressing the anti-GluN2A shRNA in vivo displayed an impaired contextual fear-conditioning memory. In addition, these animals showed increased seizure susceptibility, in terms of both time and intensity, which led us to conclude that deregulation in GluN2A expression at the hippocampus is associated with seizure susceptibility and learning–memory mechanisms.
Collapse
Affiliation(s)
- Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | - Jordana Griebler Luft
- Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cecila Alejandra Vazquez
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | - Bruno Popik
- Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Magalí C Cercato
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | | | - Anna Salvetti
- International Center for Infectiology Research (CIRI), INSERM U1111, CNRS UMR5308, Université de Lyon (UCBL1), Lyon, France
| | - Diana A Jerusalinsky
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | | | - Maria Verónica Baez
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina.,1° U.A. Departamento de Histologia, Embriología, Biologia Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
37
|
Łukawski K, Czuczwar SJ. Developing precision treatments for epilepsy using patient and animal models. Expert Rev Neurother 2020; 21:1241-1250. [PMID: 33339471 DOI: 10.1080/14737175.2021.1866989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Phenytoin was the first antiepileptic drug (AED) discovered in an animal model of seizures whose clinical efficacy was subsequently confirmed. This clearly indicated that a search for other AEDs had to consider animal studies.Areas covered: Main seizure tests used for the evaluation of possible anticonvulsive activity of potential anticonvulsants and their predictive values have been reviewed. Procedures used for the estimation of antiepileptogenic effects have been also included.Expert opinion: First-line seizure models comprise maximal electroshock (MES)-, pentylenetetrazol (PTZ)- and kindling-induced convulsions in rodents. The MES test may be considered as a convenient and easy model of generalized tonic-clonic seizures, PTZ test - as a model of generalized myoclonic seizures and to a certain degree - absence seizures. Kindled seizures (for example, from amygdala) may be regarded as a model of focal seizures. Some tests have been suggested for the search of AEDs effective in drug-resistant seizures - for instance, 6 Hz (44 mA) test or intrahippocampal kainate model of mesial temporal lobe epilepsy. There are also recommendations from experimental epileptology on synergistic AED combinations for patients with drug-resistant seizures. The clinical evidence on this issue is scarce and favors a combined treatment with valproate + lamotrigine.
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland.,Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
38
|
Jones A, Barker-Haliski M, Ilie AS, Herd MB, Baxendale S, Holdsworth CJ, Ashton JP, Placzek M, Jayasekera BAP, Cowie CJA, Lambert JJ, Trevelyan AJ, Steve White H, Marson AG, Cunliffe VT, Sills GJ, Morgan A. A multiorganism pipeline for antiseizure drug discovery: Identification of chlorothymol as a novel γ-aminobutyric acidergic anticonvulsant. Epilepsia 2020; 61:2106-2118. [PMID: 32797628 PMCID: PMC10756143 DOI: 10.1111/epi.16644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Current medicines are ineffective in approximately one-third of people with epilepsy. Therefore, new antiseizure drugs are urgently needed to address this problem of pharmacoresistance. However, traditional rodent seizure and epilepsy models are poorly suited to high-throughput compound screening. Furthermore, testing in a single species increases the chance that therapeutic compounds act on molecular targets that may not be conserved in humans. To address these issues, we developed a pipeline approach using four different organisms. METHODS We sequentially employed compound library screening in the zebrafish, Danio rerio, chemical genetics in the worm, Caenorhabditis elegans, electrophysiological analysis in mouse and human brain slices, and preclinical validation in mouse seizure models to identify novel antiseizure drugs and their molecular mechanism of action. RESULTS Initially, a library of 1690 compounds was screened in an acute pentylenetetrazol seizure model using D rerio. From this screen, the compound chlorothymol was identified as an effective anticonvulsant not only in fish, but also in worms. A subsequent genetic screen in C elegans revealed the molecular target of chlorothymol to be LGC-37, a worm γ-aminobutyric acid type A (GABAA ) receptor subunit. This GABAergic effect was confirmed using in vitro brain slice preparations from both mice and humans, as chlorothymol was shown to enhance tonic and phasic inhibition and this action was reversed by the GABAA receptor antagonist, bicuculline. Finally, chlorothymol exhibited in vivo anticonvulsant efficacy in several mouse seizure assays, including the 6-Hz 44-mA model of pharmacoresistant seizures. SIGNIFICANCE These findings establish a multiorganism approach that can identify compounds with evolutionarily conserved molecular targets and translational potential, and so may be useful in drug discovery for epilepsy and possibly other conditions.
Collapse
Affiliation(s)
- Alistair Jones
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Andrei S. Ilie
- Institute of Neuroscience, University of Newcastle, Newcastle, UK
| | - Murray B. Herd
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sarah Baxendale
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | | | - John-Paul Ashton
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Marysia Placzek
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Bodiabaduge A. P. Jayasekera
- Institute of Neuroscience, University of Newcastle, Newcastle, UK
- Department of Neurosurgery, Royal Victoria Infirmary, Newcastle, UK
| | - Christopher J. A. Cowie
- Institute of Neuroscience, University of Newcastle, Newcastle, UK
- Department of Neurosurgery, Royal Victoria Infirmary, Newcastle, UK
| | - Jeremy J. Lambert
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | | - H. Steve White
- Department of Pharmacy, University of Washington, Seattle
| | - Anthony G. Marson
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Graeme J. Sills
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Alan Morgan
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
39
|
Koneval Z, Knox KM, Memon A, Zierath DK, White HS, Barker-Haliski M. Antiseizure drug efficacy and tolerability in established and novel drug discovery seizure models in outbred vs inbred mice. Epilepsia 2020; 61:2022-2034. [PMID: 32757210 PMCID: PMC7722177 DOI: 10.1111/epi.16624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Initial identification of new investigational drugs for the treatment of epilepsy is commonly conducted in well-established mouse acute and chronic seizure models: for example, maximal electroshock (MES), 6 Hz, and corneal kindling. Comparison of the median effective dose (ED50) of approved antiseizure drugs (ASDs) vs investigational agents in these models provides evidence of their potential for clinical efficacy. Inbred and outbred mouse strains exhibit differential seizure susceptibility. However, few comparisons exist of the ED50 or median behaviorally impairing dose (TD50) of prototype ASDs in these models in inbred C57Bl/6 vs outbred CF-1 mice, both of which are often used for ASD discovery. METHODS We defined the strain-related ED50s and TD50s of several mechanistically distinct ASDs across established acute seizure models (MES, 6 Hz, and corneal-kindled mouse). We further quantified the strain-related effect of the MES ED50 of each ASD on gross behavior in a locomotor activity assay. Finally, we describe a novel pharmacoresistant corneal-kindling protocol that is suitable for moderate-throughput ASD screening and demonstrates highly differentiated ASD sensitivity. RESULTS We report significant strain-related differences in the MES ED50 of valproic acid (CF-1 ED50: 90 mg/kg [95% confidence interval (CI) 165-214] vs C57Bl/6: 276 mg/kg [226-366]), as well as significant differences in the ED50 of levetiracetam in the pharmacoresistant 6 Hz test (CF-1: 22.5 mg/kg [14.7-30.2] vs C57Bl/6: >500 mg/kg [CI not defined]). There were no differences in the calculated TD50 of these ASDs between strains. Furthermore, the MES ED50 of phenobarbital significantly enhanced locomotor activity of outbred CF-1, but not C57Bl/6, mice. SIGNIFICANCE Altogether, this study provides strain-related information to differentiate investigational agents from ASD standards-of-care in commonly employed preclinical discovery models and describes a novel kindled seizure model to further explore the mechanisms of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Zachery Koneval
- Department of Pharmacy, School of Pharmacy, University of Washington
| | - Kevin M. Knox
- Department of Pharmacy, School of Pharmacy, University of Washington
| | - Ali Memon
- Neuroscience Undergraduate Program, University of Washington
| | | | - H. Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington
| | | |
Collapse
|
40
|
Victor TR, Tsirka SE. Microglial contributions to aberrant neurogenesis and pathophysiology of epilepsy. NEUROIMMUNOLOGY AND NEUROINFLAMMATION 2020; 7:234-247. [PMID: 33154976 PMCID: PMC7641338 DOI: 10.20517/2347-8659.2020.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Microglia are dynamic cells that constitute the brain's innate immune system. Recently, research has demonstrated microglial roles beyond immunity, which include homeostatic roles in the central nervous system. The function of microglia is an active area of study, with insights into changes in neurogenesis and synaptic pruning being discovered in both health and disease. In epilepsy, activated microglia contribute to several changes that occur during epileptogenesis. In this review, we focus on the effects of microglia on neurogenesis and synaptic pruning, and discuss the current state of anti-seizure drugs and how they affect microglia during these processes. Our understanding of the role of microglia post-seizure is still limited and may be pivotal in recognizing new therapeutic targets for seizure intervention.
Collapse
Affiliation(s)
- Tanya R Victor
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
41
|
Krivoshein AV. α-Substituted Lactams and Acetamides: Ion Channel Modulators that Show Promise in Treating Drug-resistant Epilepsy. Cent Nerv Syst Agents Med Chem 2020; 20:79-87. [PMID: 32386500 DOI: 10.2174/1871524920666200510005458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 11/22/2022]
Abstract
The two main problems in the pharmacotherapy of epilepsy are resistance to currently available first-line medications (which occurs in about one third of patients) and the high incidence of side effects. To address these two challenges, extensive efforts are being undertaken to design new, structurally distinct antiepileptic drugs with a broad spectrum of anticonvulsant activity. Tests in animal models of epilepsy indicate that α-substituted lactams and acetamides show a broad spectrum of anticonvulsant activity (including very promising activity in drug-resistant models) as well as an excellent safety profile. Limited clinical results confirm these preclinical findings. In the first part of this review, pharmacology and toxicology of α-substituted lactams and acetamides and their putative protein targets in the brain have been discussed. This is followed by a discussion of structure-activity relationships among α-alkyl-, α-aryl-, and α-aryl-α-alkyl-substituted derivatives. The most promising structures seem to be those related to 3-ethyl-3-phenylpyrrolidin-2-one, 2-phenylbutyramide, and 2- sec-butylvaleramide. The information presented in this review is expected to facilitate rational drug design and development efforts for α-substituted lactams and acetamides.
Collapse
Affiliation(s)
- Arcadius V Krivoshein
- Chemistry Program, University of Houston-Clear Lake, Houston, TX 77058, United States
| |
Collapse
|