1
|
Al-Beltagi M, Saeed NK, Bediwy AS, Elbeltagi R. Unraveling the nutritional challenges in epilepsy: Risks, deficiencies, and management strategies: A systematic review. World J Exp Med 2025; 15:104328. [DOI: 10.5493/wjem.v15.i2.104328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Malnutrition and epilepsy share a complex bidirectional relationship, with malnutrition serving as a potential risk factor for epilepsy development, while epilepsy, in turn, often exerts profound effects on nutritional status. Nutritional interventions have emerged as a critical adjunctive approach in epilepsy management.
AIM To explore the multifaceted associations between malnutrition and epilepsy, structured into three primary sections: (1) Elucidating the impact of malnutrition as a risk factor for epilepsy onset; (2) Examining the reciprocal influence of epilepsy on nutritional status, and (3) Evaluating diverse nutritional interventions in the management of epilepsy.
METHODS A systematic search was conducted across PubMed, Scopus, and Web of Science databases utilizing defined keywords related to malnutrition, epilepsy, and nutritional interventions. Inclusion criteria encompassed various study types, including clinical trials, animal models, cohort studies, case reports, meta-analyses, systematic reviews, guidelines, editorials, and review articles. Four hundred sixteen pertinent references were identified, with 198 review articles, 153 research studies, 21 case reports, 24 meta-analyses, 14 systematic reviews, 4 guidelines, and 2 editorials meeting the predefined criteria.
RESULTS The review revealed the intricate interplay between malnutrition and epilepsy, highlighting malnutrition as a potential risk factor in epilepsy development and elucidating how epilepsy often leads to nutritional deficiencies. Findings underscored the importance of nutritional interventions in managing epilepsy, showing their impact on seizure frequency, neuronal function, and overall brain health.
CONCLUSION This systematic review emphasizes the bidirectional relationship between malnutrition and epilepsy while emphasizing the critical role of nutritional management in epilepsy treatment. The multifaceted insights underscore the need for a holistic approach to addressing nutritional aspects alongside conventional epilepsy management strategies.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatrics, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Governmental Hospitals, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, The Royal College of Surgeons in Ireland, Busaiteen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Reem Elbeltagi
- Medicine, Royal College of Surgeons in Ireland, Medical University of Bahrain, Busaiteen 15503, Muharraq, Bahrain
| |
Collapse
|
2
|
Li C, Wang X, Deng M, Luo Q, Yang C, Gu Z, Lin S, Luo Y, Chen L, Li Y, He B. Antiepileptic Drug Combinations for Epilepsy: Mechanisms, Clinical Strategies, and Future Prospects. Int J Mol Sci 2025; 26:4035. [PMID: 40362274 PMCID: PMC12071858 DOI: 10.3390/ijms26094035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Epilepsy is a chronic neurological disorder characterized by abnormal neuronal discharge, leading to recurrent and unpredictable disruptions in brain function. Despite over 30 antiepileptic drugs (AEDs), 30% of patients develop drug-resistant epilepsy, requiring combination therapy. This review explores epilepsy's pathogenesis, including neuronal hyperexcitability, neurotransmitter imbalances, and ion channel dysfunction, alongside genetic, inflammatory, immune, and oxidative stress factors. AEDs are classified by mechanisms like voltage-gated ion channel modulation and GABA/glutamate regulation, tracing their evolution from traditional (e.g., phenobarbital) to modern therapies (e.g., lamotrigine). Combination therapy, using complementary mechanisms (e.g., lacosamide with levetiracetam), enhances efficacy but poses risks like drug interactions and cognitive impairment. Integrating molecular biology and pharmacology advances, this review highlights the need for rational drug selection and individualized strategies to improve epilepsy treatment outcomes and patient quality of life. Future directions include personalized treatments, optimized dosage forms, novel drug targets, and multi-target drugs.
Collapse
Affiliation(s)
- Cunjiang Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Xingyu Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Mingzhenlong Deng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Qinggen Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Chaoxing Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Zhicheng Gu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Shuxian Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Yongxiang Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Lei Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| | - Yan Li
- School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China
| | - Bin He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, Guizhou Medical University, Guiyang 550004, China; (C.L.); (X.W.); (M.D.); (Q.L.); (C.Y.); (Z.G.); (S.L.); (Y.L.); (L.C.)
| |
Collapse
|
3
|
Garcia JD, Wang C, Alexander RPD, Banks E, Fenton T, DeKeyser JM, Abramova TV, George AL, Ben-Shalom R, Hackos DH, Bender KJ. Differential roles of Na V 1.2 and Na V 1.6 in neocortical pyramidal cell excitability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.17.629038. [PMID: 40235970 PMCID: PMC11996326 DOI: 10.1101/2024.12.17.629038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Mature neocortical pyramidal cells functionally express two sodium channel (Na V ) isoforms: Na V 1.2 and Na V 1.6. These isoforms are differentially localized to pyramidal cell compartments, and as such are thought to contribute to different aspects of neuronal excitability. But determining their precise roles in pyramidal cell excitability has been hampered by a lack of tools that allow for selective, acute block of each isoform individually. Here, we leveraged aryl sulfonamide-based molecule (ASC) inhibitors of Na V channels that exhibit state-dependent block of both Na V 1.2 and Na V 1.6, along with knock-in mice with changes in Na V 1.2 or Na V 1.6 structure that prevents ASC binding. This allowed for acute, potent, and reversible block of individual isoforms that permitted dissection of the unique contributions of Na V 1.2 and Na V 1.6 in pyramidal cell excitability. Remarkably, block of each isoform had contrasting-and in some situations, opposing-effects on neuronal action potential output, with Na V 1.6 block decreasing and Na V 1.2 block increasing output. Thus, Na V isoforms have unique roles in regulating different aspects of pyramidal cell excitability, and our work may help guide development of therapeutics designed to temper hyperexcitability through selective Na V isoform blockade.
Collapse
|
4
|
Dhureja M, Chaturvedi P, Choudhary A, Kumar P, Munshi A. Molecular Insights of Drug Resistance in Epilepsy: Multi-omics Unveil. Mol Neurobiol 2025; 62:1-17. [PMID: 38753128 DOI: 10.1007/s12035-024-04220-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/03/2024] [Indexed: 06/12/2024]
Abstract
Epilepsy is a devastating neurological disorder mainly associated with impaired synchronic discharge that leads to sensory, motor, and psychomotor impairments. Till now, about 30 anti-seizure medications (ASMs) have been approved for the management of epilepsy, yet one-third of individuals still have uncontrollable epilepsy and develop resistance. Drug resistance epilepsy (DRE) is defined as the condition where two ASMs fail to control the seizure in epileptic patients. The leading cause of the resistance was the extended use of ASMs. According to various studies, alterations in some genes and their expressions, along with specific metabolic impairments, are suggested to be associated with ASMs resistance and DRE pathophysiology. Several factors aid in the pathophysiology of DRE, such as alterations in protein-encoding genes such as neurotransmitter receptors, drug transporters, ion channels, and drug targets. Furthermore, the altered metabolite levels of metabolites implicated in neurotransmitter signaling, energetic pathways, oxidative stress, and neuroinflammatory signaling differentiate the epileptic patient from the DRE patient. Various DRE biomarkers can be identified using the "integrated omics approach," which includes the study of genomics, transcriptomics, and metabolomics. The current review has been compiled to understand the pathophysiological mechanisms of DRE by focusing on genomics, transcriptomics, and metabolomics. An effort has also been made to identify the therapeutic targets based on identifying significant markers by a multi-omics approach. This has the potential to develop novel therapeutic interventions in the future.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Pragya Chaturvedi
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India
| | - Anita Choudhary
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicines, Central University of Punjab, Bathinda, India.
| |
Collapse
|
5
|
Morris-Schaffer K, Higgins L, Kocabas NA, Faulhammer F, Cordova A, Freeman E, Kamp H, Nahar M, Richmond E, Rooseboom M. A weight of evidence review on the mode of action, adversity, and the human relevance of xylene's observed thyroid effects in rats. Crit Rev Toxicol 2025; 55:1-26. [PMID: 39785829 DOI: 10.1080/10408444.2024.2422890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 01/12/2025]
Abstract
Xylene substances have wide industrial and consumer uses and are currently undergoing dossier and substance evaluation under Registration, Evaluation, Authorization and Restriction of Chemicals (REACH) for further toxicological testing including consideration of an additional neurotoxicological testing cohort to an extended one-generation reproduction toxicity (EOGRT) study. New repeated dose study data on xylenes identify the thyroid as a potential target tissue, and therefore a weight of evidence review is provided to investigate whether or not xylene-mediated changes on the hypothalamus-pituitary-thyroid (HPT) axis are secondary to liver enzymatic induction and are of a magnitude that is relevant for neurological human health concerns. Multiple published studies confirm xylene-mediated increases in liver weight, hepatocellular hypertrophy, and liver enzymatic induction via the oral or inhalation routes, including an increase in uridine 5'-diphospho-glucuronosyltransferase (UDP-GT) activity, the key step in thyroid hormone metabolism in rodents. Only minimal to slight increases in thyroid follicular cell hypertrophy have been observed in some xylene repeated dose studies, with no associated robust or consistent perturbance of thyroid hormone changes across the studies or carried through to offspring indicating adaptive homeostatic maintenance of the HPT axis. Also importantly, in vitro human cell line data from the United States Environmental Protection Agency (US EPA) Toxicity Forecasting (ToxCast) provides supporting evidence of xylene's inability to directly perturb thyroidal functionality. A further supplemental in-depth metabolomics analysis (MetaMap®Tox) of xylene showed a tentative match to compounds that also demonstrate extra-thyroidal effects on the HPT axis as a consequence of liver enzyme induction. Lastly, the slight HPT axis changes mediated by xylene were well-below the published literature thresholds for developmental neurotoxicological outcomes established for thyroidal changes in animals and humans. In summary, the data and various lines of scientific evidence presented herein individually and collectively demonstrate that xylene's mediated changes in the HPT axis, via a secondary extra-thyroidal MOA (i.e. liver enzyme induction), do not raise a human health concern with regards to developmental neurotoxicity. As such, the available toxicological data do not support the classification of xylene as a known or suspected endocrine disruptor, specifically through the thyroid modality, per Regulations Commission Delegated Regulation (EU) 2023/707 of 19 December 2022 amending Regulation (EC) No 1272/2008 and do not support the need for a neurotoxicological cohort evaluation in any subsequent EOGRTS.
Collapse
Affiliation(s)
| | - Larry Higgins
- Scientific Services, Penman Consulting bvba, Brussels, Belgium
| | | | - Frank Faulhammer
- Global Toxicology & Ecotoxicology, BASF SE, Ludwigshafen, Germany
| | - Alexandra Cordova
- Environmental & Earth Sciences, Exponent Incorporated, Austin, TX, USA
| | - Elaine Freeman
- Exponent Incorporated, Chemical Regulation and Food Safety, Washington, D.C., USA
| | | | - Muna Nahar
- Exponent Incorporated, Chemical Regulation and Food Safety, Washington, D.C., USA
| | - Emily Richmond
- Chemical Regulation and Food Safety, Exponent International, UK
| | - Martijn Rooseboom
- Product Stewardship, Science & Regulatory, Shell Global Solutions International B.V. The Hague, the Netherlands
| |
Collapse
|
6
|
Goodchild SJ, Shuart NG, Williams AD, Ye W, Parrish RR, Soriano M, Thouta S, Mezeyova J, Waldbrook M, Dean R, Focken T, Ghovanloo MR, Ruben PC, Scott F, Cohen CJ, Empfield J, Johnson JP. Molecular Pharmacology of Selective Na V1.6 and Dual Na V1.6/Na V1.2 Channel Inhibitors that Suppress Excitatory Neuronal Activity Ex Vivo. ACS Chem Neurosci 2024; 15:1169-1184. [PMID: 38359277 PMCID: PMC10958515 DOI: 10.1021/acschemneuro.3c00757] [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: 11/21/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Voltage-gated sodium channel (NaV) inhibitors are used to treat neurological disorders of hyperexcitability such as epilepsy. These drugs act by attenuating neuronal action potential firing to reduce excitability in the brain. However, all currently available NaV-targeting antiseizure medications nonselectively inhibit the brain channels NaV1.1, NaV1.2, and NaV1.6, which potentially limits the efficacy and therapeutic safety margins of these drugs. Here, we report on XPC-7724 and XPC-5462, which represent a new class of small molecule NaV-targeting compounds. These compounds specifically target inhibition of the NaV1.6 and NaV1.2 channels, which are abundantly expressed in excitatory pyramidal neurons. They have a > 100-fold molecular selectivity against NaV1.1 channels, which are predominantly expressed in inhibitory neurons. Sparing NaV1.1 preserves the inhibitory activity in the brain. These compounds bind to and stabilize the inactivated state of the channels thereby reducing the activity of excitatory neurons. They have higher potency, with longer residency times and slower off-rates, than the clinically used antiseizure medications carbamazepine and phenytoin. The neuronal selectivity of these compounds is demonstrated in brain slices by inhibition of firing in cortical excitatory pyramidal neurons, without impacting fast spiking inhibitory interneurons. XPC-5462 also suppresses epileptiform activity in an ex vivo brain slice seizure model, whereas XPC-7224 does not, suggesting a possible requirement of Nav1.2 inhibition in 0-Mg2+- or 4-AP-induced brain slice seizure models. The profiles of these compounds will facilitate pharmacological dissection of the physiological roles of NaV1.2 and NaV1.6 in neurons and help define the role of specific channels in disease states. This unique selectivity profile provides a new approach to potentially treat disorders of neuronal hyperexcitability by selectively downregulating excitatory circuits.
Collapse
Affiliation(s)
- Samuel J. Goodchild
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Noah Gregory Shuart
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Aaron D. Williams
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Wenlei Ye
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - R. Ryley Parrish
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Maegan Soriano
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Samrat Thouta
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Janette Mezeyova
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Matthew Waldbrook
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Richard Dean
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Thilo Focken
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Mohammad-Reza Ghovanloo
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department
of Neurology, Yale University, New Haven, Connecticut 06519, United States
| | - Peter C. Ruben
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Fiona Scott
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - Charles J. Cohen
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - James Empfield
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - JP Johnson
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| |
Collapse
|
7
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|
8
|
Jia L, Eroglu TE, Wilders R, Verkerk AO, Tan HL. Carbamazepine Increases the Risk of Sudden Cardiac Arrest by a Reduction of the Cardiac Sodium Current. Front Cell Dev Biol 2022; 10:891996. [PMID: 35721495 PMCID: PMC9204209 DOI: 10.3389/fcell.2022.891996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the risk of sudden cardiac arrest (SCA) associated with the use of carbamazepine (CBZ) and establish the possible underlying cellular electrophysiological mechanisms. Methods: The SCA risk association with CBZ was studied in general population cohorts using a case–control design (n = 5,473 SCA cases, 21,866 non-SCA controls). Effects of 1–100 µM CBZ on action potentials (APs) and individual membrane currents were determined in isolated rabbit and human cardiomyocytes using the patch clamp technique. Results: CBZ use was associated with increased risk of SCA compared with no use (adjusted odds ratio 1.90 [95% confidence interval: 1.12–3.24]). CBZ reduced the AP upstroke velocity of rabbit and human cardiomyocytes, without prominent changes in other AP parameters. The reduction occurred at ≥30 µM and was frequency-dependent with a more pronounced reduction at high stimulus frequencies. The cardiac sodium current (INa) was reduced at ≥30 μM; this was accompanied by a hyperpolarizing shift in the voltage-dependency of inactivation. The recovery from inactivation was slower, which is consistent with the more pronounced AP upstroke velocity reduction at high stimulus frequencies. The main cardiac K+ and Ca2+ currents were unaffected, except reduction of L-type Ca2+ current by 100 µM CBZ. Conclusion: CBZ use is associated with an increased risk of SCA in the general population. At concentrations of 30 µM and above, CBZ reduces AP upstroke velocity and INa in cardiomyocytes. Since the concentration of 30 µM is well within the therapeutic range (20–40 µM), we conclude that CBZ increases the risk of SCA by a reduction of the cardiac INa.
Collapse
Affiliation(s)
- Lixia Jia
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Talip E. Eroglu
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Arie O. Verkerk
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanno L. Tan
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
- *Correspondence: Hanno L. Tan,
| |
Collapse
|
9
|
Chindo BA, Howes MJR, Abuhamdah S, Yakubu MI, Ayuba GI, Battison A, Chazot PL. New Insights Into the Anticonvulsant Effects of Essential Oil From Melissa officinalis L. (Lemon Balm). Front Pharmacol 2021; 12:760674. [PMID: 34721045 PMCID: PMC8551917 DOI: 10.3389/fphar.2021.760674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/27/2021] [Indexed: 11/25/2022] Open
Abstract
Melissa officinalis L. is used in traditional European and Iranian folk medicines to treat a plethora of neurological diseases including epilepsy. We utilized the in vitro and in vivo models of epilepsy to probe the anticonvulsant potentials of essential oil from M. officinalis (MO) to gain insight into the scientific basis for its applications in traditional medicine for the management of convulsive disorders. MO was evaluated for effects on maximal electroshock (MES) and pentylenetetrazole (PTZ) -induced seizures in mice, on 4–aminopyridine (4-AP)-brain slice model of epilepsy and sustained repetitive firing of current clamped neurons; and its ameliorative effects were examined on seizure severity, anxiety, depression, cognitive dysfunction, oxidative stress and neuronal cell loss in PTZ-kindled rats. MO reversibly blocked spontaneous ictal-like discharges in the 4-AP-brain slice model of epilepsy and secondary spikes from sustained repetitive firing, suggesting anticonvulsant effects and voltage-gated sodium channel blockade. MO protected mice from PTZ– and MES–induced seizures and mortality, and ameliorated seizure severity, fear-avoidance, depressive-like behavior, cognitive deficits, oxidative stress and neuronal cell loss in PTZ–kindled rats. The findings warrant further study for the potential use of MO and/or its constituent(s) as adjunctive therapy for epileptic patients.
Collapse
Affiliation(s)
- Ben A Chindo
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Kaduna State University, Kaduna, Nigeria
| | | | - Sawsan Abuhamdah
- Department of Biosciences, Durham University, Durham, United Kingdom.,College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates.,Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| | - Musa I Yakubu
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Kaduna State University, Kaduna, Nigeria
| | - Godwin I Ayuba
- Department of Anatomic Pathology and Forensic Medicine, College of Medicine, Kaduna State University, Kaduna, Nigeria
| | | | - Paul L Chazot
- Department of Biosciences, Durham University, Durham, United Kingdom
| |
Collapse
|
10
|
Guignet M, Campbell A, White HS. Cenobamate (XCOPRI): Can preclinical and clinical evidence provide insight into its mechanism of action? Epilepsia 2020; 61:2329-2339. [DOI: 10.1111/epi.16718] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Michelle Guignet
- Department of Pharmacy School of Pharmacy University of Washington Seattle WA USA
| | - Amanda Campbell
- Department of Pharmacy School of Pharmacy University of Washington Seattle WA USA
| | - H. Steve White
- Department of Pharmacy School of Pharmacy University of Washington Seattle WA USA
| |
Collapse
|
11
|
Nomura S, Kida H, Hirayama Y, Imoto H, Inoue T, Moriyama H, Mitsushima D, Suzuki M. Reduction of spike generation frequency by cooling in brain slices from rats and from patients with epilepsy. J Cereb Blood Flow Metab 2019; 39:2286-2294. [PMID: 30117752 PMCID: PMC6827110 DOI: 10.1177/0271678x18795365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This study aimed to understand the mechanism by which brain cooling terminates epileptic discharge. Cortical slices were prepared from rat brains (n = 19) and samples from patients with intractable epilepsy that had undergone temporal lobectomy (n = 7). We performed whole cell current clamp recordings at approximately physiological brain temperature (35℃) and at cooler temperatures (25℃ and 15℃). The firing threshold in human neurons was lower at 25℃ (-32.6 mV) than at 35℃ (-27.0 mV). The resting potential and spike frequency were similar at 25℃ and 35℃. Cooling from 25℃ to 15℃ did not change the firing threshold, but the resting potential increased from -65.5 to -54.0 mV and the waveform broadened from 1.85 to 6.55 ms, due to delayed repolarization. These changes enhanced the initial spike appearance and reduced spike frequency; moreover, spike frequency was insensitive to increased levels of current injections. Similar results were obtained in rat brain studies. We concluded that the reduction in spike frequency at 15℃, due to delayed repolarization, might be a key mechanism by which brain cooling terminates epileptic discharge. On the other hand, spike frequency was not influenced by the reduced firing threshold or the elevated resting potential caused by cooling.
Collapse
Affiliation(s)
- Sadahiro Nomura
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,Epilepsy Center, Yamaguchi University Hospital, Yamaguchi, Japan
| | - Hiroyuki Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yuya Hirayama
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hirochika Imoto
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,Epilepsy Center, Yamaguchi University Hospital, Yamaguchi, Japan
| | - Takao Inoue
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroshi Moriyama
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Michiyasu Suzuki
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
12
|
Pejanovic-Skobic N, Markovic I, Bozina N, Basic S. Lack of association of SCN2A rs17183814 polymorphism with the efficacy of lamotrigine monotherapy in patients with focal epilepsy from Herzegovina area, Bosnia and Herzegovina. Epilepsy Res 2019; 158:106221. [PMID: 31707316 DOI: 10.1016/j.eplepsyres.2019.106221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/12/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We assessed the influence of the SCN2A gene polymorphism c.56 G > A rs17183814 on the response to lamotrigine monotherapy in patients with focal epilepsy in Herzegovina area, Bosnia and Herzegovina. MATERIAL AND METHODS For SCN2A polymorphism c.56 G > A rs17183814, one hundred patients with epilepsy who were receiving lamotrigine in monotherapy and seventy-one age and sex matched healthy controls were genotyped using TaqMan assay. All patients were Caucasians from the region of Herzegovina, Bosnia and Herzegovina. Genotyping was conducted using a polymerase chain reaction in real time. Patients were divided into two groups: responders and non-responders. RESULTS Of all patients with epilepsy, 33% were non-responders, and 67% were responders. The mean age of non-responders was 38.8 vs. group of responders in which it was 35.2. Mean age of onset of seizures in epilepsy patients was 26.7 for non-responders and 25.4 for responders. In patients with epilepsy, the mean age of seizure onset was 26.7 for non-responders and 25.4 for responders. For SCN2A c.56 G > A gene polymorphism, we did not observe any significant differences in genotypic or allelic frequency between patients with epilepsy and healthy controls. Genotype or allelic frequencies of SCN2A c.56 G > A gene polymorphism did not significantly differ for AG or GG genotypes in the non-responders vs. responders. CONCLUSION There was no significant association in patients with focal epilepsy between studied genotypes and response to lamotrigine monotherapy in Herzegovina patients with focal epilepsy. However, we need studies in a bigger cohort of patients with epilepsy to be assessed in the future.
Collapse
Affiliation(s)
- Natasa Pejanovic-Skobic
- Clinic of Neurology, University Clinical Hospital Mostar, 88000 Mostar, Bosnia and Herzegovina.
| | - Ivana Markovic
- Clinic of Neurology, Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Nada Bozina
- Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Silvio Basic
- Clinic of Neurology, Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| |
Collapse
|
13
|
Janmohamed M, Brodie MJ, Kwan P. Pharmacoresistance - Epidemiology, mechanisms, and impact on epilepsy treatment. Neuropharmacology 2019; 168:107790. [PMID: 31560910 DOI: 10.1016/j.neuropharm.2019.107790] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/01/2019] [Accepted: 09/21/2019] [Indexed: 12/25/2022]
Abstract
Understanding the natural history of and factors associated with pharmacoresistant epilepsy provides the foundation for formulating mechanistic hypotheses that can be evaluated to drive the development of novel treatments. This article reviews the modern definition of drug-resistant epilepsy, its prevalence and incidence, risk factors, hypothesized mechanisms, and the implication of recognizing pharmacoresistance in therapeutic strategies. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Mubeen Janmohamed
- Department of Neuroscience, Alfred Hospital, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | | | - Patrick Kwan
- Department of Neuroscience, Alfred Hospital, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
14
|
Naimo GD, Guarnaccia M, Sprovieri T, Ungaro C, Conforti FL, Andò S, Cavallaro S. A Systems Biology Approach for Personalized Medicine in Refractory Epilepsy. Int J Mol Sci 2019; 20:E3717. [PMID: 31366017 PMCID: PMC6695675 DOI: 10.3390/ijms20153717] [Citation(s) in RCA: 18] [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: 06/27/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 02/01/2023] Open
Abstract
Epilepsy refers to a common chronic neurological disorder that affects all age groups. Unfortunately, antiepileptic drugs are ineffective in about one-third of patients. The complex interindividual variability influences the response to drug treatment rendering the therapeutic failure one of the most relevant problems in clinical practice also for increased hospitalizations and healthcare costs. Recent advances in the genetics and neurobiology of epilepsies are laying the groundwork for a new personalized medicine, focused on the reversal or avoidance of the pathophysiological effects of specific gene mutations. This could lead to a significant improvement in the efficacy and safety of treatments for epilepsy, targeting the biological mechanisms responsible for epilepsy in each individual. In this review article, we focus on the mechanism of the epilepsy pharmacoresistance and highlight the use of a systems biology approach for personalized medicine in refractory epilepsy.
Collapse
Affiliation(s)
- Giuseppina Daniela Naimo
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Maria Guarnaccia
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Teresa Sprovieri
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Carmine Ungaro
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy.
| |
Collapse
|
15
|
de la Tremblaye PB, O'Neil DA, LaPorte MJ, Cheng JP, Beitchman JA, Thomas TC, Bondi CO, Kline AE. Elucidating opportunities and pitfalls in the treatment of experimental traumatic brain injury to optimize and facilitate clinical translation. Neurosci Biobehav Rev 2018; 85:160-175. [PMID: 28576511 PMCID: PMC5709241 DOI: 10.1016/j.neubiorev.2017.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022]
Abstract
The aim of this review is to discuss the research presented in a symposium entitled "Current progress in characterizing therapeutic strategies and challenges in experimental CNS injury" which was presented at the 2016 International Behavioral Neuroscience Society annual meeting. Herein we discuss diffuse and focal traumatic brain injury (TBI) and ensuing chronic behavioral deficits as well as potential rehabilitative approaches. We also discuss the effects of stress on executive function after TBI as well as the response of the endocrine system and regulatory feedback mechanisms. The role of the endocannabinoids after CNS injury is also discussed. Finally, we conclude with a discussion of antipsychotic and antiepileptic drugs, which are provided to control TBI-induced agitation and seizures, respectively. The review consists predominantly of published data.
Collapse
Affiliation(s)
- Patricia B de la Tremblaye
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Darik A O'Neil
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Megan J LaPorte
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jeffrey P Cheng
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Midwestern University, Glendale, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Phoenix VA Healthcare System, Phoenix, AZ, United States
| | - Corina O Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anthony E Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
16
|
|
17
|
Pastore V, Wasowski C, Martin P, Enrique A, Higgs J, Bruno-Blanch LE, Milesi V, Marder M. N-propyl-2,2-diphenyl-2-hydroxyacetamide, a novel α-hydroxyamide with anticonvulsant, anxiolytic and antidepressant-like effects that inhibits voltage-gated sodium channels. Eur J Pharmacol 2017; 819:270-280. [PMID: 29217174 DOI: 10.1016/j.ejphar.2017.11.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 11/14/2017] [Accepted: 11/30/2017] [Indexed: 01/08/2023]
Abstract
In patients with epilepsy, anxiety and depression are the most frequent psychiatric comorbidities but they often remain unrecognized and untreated. We report herein the antidepressant-like activity in two animal models, tail suspension and forced swimming tests, of six anticonvulsants α-hydroxyamides. From these, N-propyl-2,2-diphenyl-2-hydroxyacetamide (compound 5) emerged not only as the most active as anticonvulsant (ED50 = 2.5mg/kg, MES test), but it showed the most remarkable antidepressant-like effect in the tail suspension and forced swimming tests (0.3-30mg/kg, i.p.); and, also, anxiolytic-like action in the plus maze test (3-10mg/kg, i.p.) in mice. Studies of its mechanism of action, by means of its capacity to act via the GABAA receptor ([3H]-flunitrazepam binding assay); the 5-HT1A receptor ([3H]-8-OH-DPAT binding assay) and the voltage-gated sodium channels (either using the patch clamp technique in hNav 1.2 expressed in HEK293 cell line or using veratrine, in vivo) were attempted. The results demonstrated that its effects are not likely related to 5-HT1A or GABAAergic receptors and that its anticonvulsant and antidepressant-like effect could be due to its voltage-gated sodium channel blocking properties.
Collapse
Affiliation(s)
- Valentina Pastore
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Junín 956 (C1113AAD), Ciudad de Buenos Aires, Argentina.
| | - Cristina Wasowski
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Junín 956 (C1113AAD), Ciudad de Buenos Aires, Argentina.
| | - Pedro Martin
- Universidad Nacional de La Plata, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Estudios Inmunopatológicos y Fisiológicos, 47 street and 115 (B1900BJW), La Plata, Argentina.
| | - Andrea Enrique
- Química Medicinal, Departamento de Ciencias Biológicas, Universidad Nacional de La Plata, 47 street and 115 (B1900BJW), La Plata, Argentina.
| | - Josefina Higgs
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Junín 956 (C1113AAD), Ciudad de Buenos Aires, Argentina.
| | - Luis E Bruno-Blanch
- Química Medicinal, Departamento de Ciencias Biológicas, Universidad Nacional de La Plata, 47 street and 115 (B1900BJW), La Plata, Argentina.
| | - Veronica Milesi
- Universidad Nacional de La Plata, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Estudios Inmunopatológicos y Fisiológicos, 47 street and 115 (B1900BJW), La Plata, Argentina.
| | - Mariel Marder
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Junín 956 (C1113AAD), Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
18
|
Prasad Tripathi RK, Ayyannan SR. Anticonvulsant activity, organotypic hippocampal neuroprotection assay and in-silico sodium channel blocking potential of 2-amino-6-nitrobenzothiazole derived semicarbazones. Biomed Pharmacother 2017; 95:1451-1460. [PMID: 28946193 DOI: 10.1016/j.biopha.2017.09.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/29/2017] [Accepted: 09/13/2017] [Indexed: 10/18/2022] Open
Abstract
Epilepsy is one of the dreadful neurodegenerative disorder characterized by recurrent, unprovoked seizures. Currently available antiepileptic drugs are still associated with enormous side effects resulting in search of newer, more effective and safer agents. In view of this, we have investigated anticonvulsant activity of 2-amino-6-nitrobenzothiazole derived semicarbazones (7-32) in various in-vivo animal seizure models viz. maximal electroshock (MES), subcutaneous pentylenetetrazole (scPTZ) and 6Hz psychomotor seizure model. Neurotoxicity was estimated by rotarod test. The compounds were also assessed for their neuroprotective potential from excitotoxic insult using organotypic hippocampal slice culture neuroprotection assay. Several compounds exhibited excellent anticonvulsant activity in MES and scPTZ models compared to reference drugs, phenytoin and levetiracetam. The results of kainic acid (KA) - induced neuroprotection assay indicated that compounds 26 and 24 were found to be most potent with IC50 of 99.54±1.27 and 101.00±1.20μM respectively. Both the compounds attenuated KA-mediated cell death in organotypic hippocampal slice cultures. Some of the compounds were found to be good antidepressants, better than the reference drug citalopram, when analyzed in forced swim test. Since semicarbazones exhibited profile resembling phenytoin, an attempt was made to screen them against human neuronal sodium channel isoform (hNav1.2) by performing computational molecular docking using AutoDock 4.2. Compound 30, 1-(5-Chloro-2-oxoindolin-3-ylidene)-4-(6-nitrobenzothiazol-2-yl)semicarbazide emerged as lead candidate possessing excellent in-vivo MES activity and high binding affinity computationally, better than the reference drug phenytoin and also exhibited neuroprotection from excitotoxic insult in KA-induced neuroprotection assay (IC50=126.80±1.24μM). However, some of the active compounds were neurotoxic at their anticonvulsant doses. Further optimization studies are needed to reduce toxicity and develop them as novel therapeutic agents for epilepsy.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India.
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India.
| |
Collapse
|
19
|
Cardoso FC, Dekan Z, Smith JJ, Deuis JR, Vetter I, Herzig V, Alewood PF, King GF, Lewis RJ. Modulatory features of the novel spider toxin μ-TRTX-Df1a isolated from the venom of the spider Davus fasciatus. Br J Pharmacol 2017; 174:2528-2544. [PMID: 28542706 DOI: 10.1111/bph.13865] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Naturally occurring dysfunction of voltage-gated sodium (NaV ) channels results in complex disorders such as chronic pain, making these channels an attractive target for new therapies. In the pursuit of novel NaV modulators, we investigated spider venoms for new inhibitors of NaV channels. EXPERIMENTAL APPROACH We used high-throughput screens to identify a NaV modulator in venom of the spider Davus fasciatus. Further characterization of this venom peptide was undertaken using fluorescent and electrophysiological assays, molecular modelling and a rodent pain model. KEY RESULTS We identified a potent NaV inhibitor named μ-TRTX-Df1a. This 34-residue peptide fully inhibited responses mediated by NaV 1.7 endogenously expressed in SH-SY5Y cells. Df1a also inhibited voltage-gated calcium (CaV 3) currents but had no activity against the voltage-gated potassium (KV 2) channel. The modelled structure of Df1a, which contains an inhibitor cystine knot motif, is reminiscent of the NaV channel toxin ProTx-I. Electrophysiology revealed that Df1a inhibits all NaV subtypes tested (hNaV 1.1-1.7). Df1a also slowed fast inactivation of NaV 1.1, NaV 1.3 and NaV 1.5 and modified the voltage-dependence of activation and inactivation of most of the NaV subtypes. Df1a preferentially binds to the domain II voltage-sensor and has additional interactions with the voltage sensors domains III and IV, which probably explains its modulatory features. Df1a was analgesic in vivo, reversing the spontaneous pain behaviours induced by the NaV activator OD1. CONCLUSION AND IMPLICATIONS μ-TRTX-Df1a shows potential as a new molecule for the development of drugs to treat pain disorders mediated by voltage-gated ion channels.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer J Smith
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
20
|
Angelopoulou C, Veletza S, Heliopoulos I, Vadikolias K, Tripsianis G, Stathi C, Piperidou C. Association of SCN1A gene polymorphism with antiepileptic drug responsiveness in the population of Thrace, Greece. Arch Med Sci 2017; 13:138-147. [PMID: 28144265 PMCID: PMC5206360 DOI: 10.5114/aoms.2016.59737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/13/2015] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The aim was to examine the influence of the SCN1A gene polymorphism IVS5-91 rs3812718 G>A on the response to antiepileptic drugs (AEDs) in monotherapy or polytherapy. MATERIAL AND METHODS Two hundred epilepsy patients and 200 healthy subjects were genotyped for SCN1A IVS5-91 rs3812718 G>A polymorphism using TaqMan assay. Patients were divided into drug-responsive and drug-resistant patients. The drug-responsive group was further studied, comparing monotherapy in maximum and minimum doses and monotherapy-responsive and -resistant groups. RESULTS There were no statistically significant differences in the allelic frequencies and genotype distributions between patients and controls (p = 0.178). The distribution of SCN1A IVS5-91 rs3812718 G>A genotypes was similar between drug-responsive and drug-resistant patients (p = 0.463). The differences in genotype distributions (A/A or A/G vs. G/G) between monotherapy-responsive and -resistant groups were statistically significant (p = 0.021). Within the monotherapy-responsive group, patients with the A/A or A/G genotype needed higher dose AEDs than patients with the G/G genotype (p = 0.032). The relative risk for generalized epilepsy due to A-containing genotypes was of marginal statistical significance when compared with the G/G genotype (p = 0.05). CONCLUSIONS Overall, our findings demonstrate an association of SCN1A IVS5-91 rs3812718 G>A polymorphism with AED responsiveness in monotherapy without evidence of an effect on drug-resistant epilepsy.
Collapse
Affiliation(s)
| | - Stavroula Veletza
- Department of Neurology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Heliopoulos
- Department of Neurology, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Grigorios Tripsianis
- Department of Neurology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Chrysa Stathi
- Department of Neurology, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|
21
|
Holtkamp D, Opitz T, Niespodziany I, Wolff C, Beck H. Activity of the anticonvulsant lacosamide in experimental and human epilepsy via selective effects on slow Na+channel inactivation. Epilepsia 2016; 58:27-41. [DOI: 10.1111/epi.13602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Dominik Holtkamp
- Department of Epileptology; Laboratory for Experimental Epileptology and Cognition Research; University of Bonn; Bonn Germany
| | - Thoralf Opitz
- Department of Epileptology; Laboratory for Experimental Epileptology and Cognition Research; University of Bonn; Bonn Germany
| | | | | | - Heinz Beck
- Department of Epileptology; Laboratory for Experimental Epileptology and Cognition Research; University of Bonn; Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE); Bonn Germany
| |
Collapse
|
22
|
Zeng Z, Hill-Yardin EL, Williams D, O'Brien T, Serelis A, French CR. Effect of phenytoin on sodium conductances in rat hippocampal CA1 pyramidal neurons. J Neurophysiol 2016; 116:1924-1936. [PMID: 27489371 DOI: 10.1152/jn.01060.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 07/24/2016] [Indexed: 11/22/2022] Open
Abstract
The antiepileptic drug phenytoin (PHT) is thought to reduce the excitability of neural tissue by stabilizing sodium channels (NaV) in inactivated states. It has been suggested the fast-inactivated state (IF) is the main target, although slow inactivation (IS) has also been implicated. Other studies on local anesthetics with similar effects on sodium channels have implicated the NaV voltage sensor interactions. In this study, we reexamined the effect of PHT in both equilibrium and dynamic transitions between fast and slower forms of inactivation in rat hippocampal CA1 pyramidal neurons. The effects of PHT were observed on fast and slow inactivation processes, as well as on another identified "intermediate" inactivation process. The effect of enzymatic removal of IF was also studied, as well as effects on the residual persistent sodium current (INaP). A computational model based on a gating charge interaction was derived that reproduced a range of PHT effects on NaV equilibrium and state transitions. No effect of PHT on IF was observed; rather, PHT appeared to facilitate the occupancy of other closed states, either through enhancement of slow inactivation or through formation of analogous drug-bound states. The overall significance of these observations is that our data are inconsistent with the commonly held view that the archetypal NaV channel inhibitor PHT stabilizes fast inactivation states, and we demonstrate that conventional slow activation "IS" and the more recently identified intermediate-duration inactivation process "II" are the primary functional targets of PHT. In addition, we show that the traditional explanatory frameworks based on the "modulated receptor hypothesis" can be substituted by simple, physiologically plausible interactions with voltage sensors. Additionally, INaP was not preferentially inhibited compared with peak INa at short latencies (50 ms) by PHT.
Collapse
Affiliation(s)
- Zhen Zeng
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Elisa L Hill-Yardin
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - David Williams
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Terence O'Brien
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia; Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; and
| | - Andris Serelis
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher R French
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia; Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; and
| |
Collapse
|
23
|
Avaliani N, Andersson M, Runegaard AH, Woldbye D, Kokaia M. DREADDs suppress seizure-like activity in a mouse model of pharmacoresistant epileptic brain tissue. Gene Ther 2016; 23:760-766. [PMID: 27416078 DOI: 10.1038/gt.2016.56] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Epilepsy is a neurological disorder with a prevalence of ≈1% of general population. Available antiepileptic drugs (AEDs) have multiple side effects and are ineffective in 30% of patients. Therefore, development of effective treatment strategies is highly needed, requiring drug-screening models that are relevant and reliable. We investigated novel chemogenetic approach, using DREADDs (designer receptors exclusively activated by designer drugs) as possible inhibitor of epileptiform activity in organotypic hippocampal slice cultures (OHSCs). The OHSCs are characterized by increased overall excitability and closely resemble features of human epileptic tissue. Studies suggest that chemically induced epileptiform activity in rat OHSCs is pharmacoresistant to most of AEDs. However, high-frequency electric stimulus train-induced bursting (STIB) in OHSCs is responsive to carbamazepine and phenytoin. We investigated whether inhibitory DREADD, hM4Di, would be effective in suppressing STIB in OHSC. hM4Di is a mutated muscarinic receptor selectively activated by otherwise inert clozapine-N-oxide, which leads to hyperpolarization in neurons. We demonstrated that this hyperpolarization effectively suppresses STIB in mouse OHSCs. As we also found that STIB in mouse OHSCs is resistant to common AED, valproic acid, collectively our findings suggest that DREADD-based strategy may be effective in suppressing epileptiform activity in a pharamcoresitant epileptic brain tissue.
Collapse
Affiliation(s)
- N Avaliani
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - M Andersson
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - A H Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - M Kokaia
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| |
Collapse
|
24
|
Synthesis of N -1′, N -3′-disubstituted spirohydantoins and their anticonvulsant activities in pilocarpine model of temporal lobe epilepsy. Bioorg Med Chem Lett 2016; 26:2912-2914. [DOI: 10.1016/j.bmcl.2016.04.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 11/23/2022]
|
25
|
Cappaert NLM, Werkman TR, Benito N, Witter MP, Baayen JC, Wadman WJ. Carbamazepine modulates the spatiotemporal activity in the dentate gyrus of rats and pharmacoresistant humans in vitro. Brain Behav 2016; 6:e00463. [PMID: 27110437 PMCID: PMC4834359 DOI: 10.1002/brb3.463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Human hippocampal tissue resected from pharmacoresistant epilepsy patients was investigated to study the effect of the antiepileptic drug CBZ (carbamazepine) and was compared to similar experiments in the hippocampus of control rats. METHODS The molecular layer of the DG (dentate gyrus) of human epileptic tissue and rat nonepileptic tissue was electrically stimulated and the evoked responses were recorded with voltage-sensitive dye imaging to characterize the spatiotemporal properties. RESULTS Bath applied CBZ (100 μmol/L) reduced the amplitude of the evoked responses in the human DG, albeit that no clear use-dependent effects were found at frequencies of 8 or 16 Hz. In nonepileptic control DG from rats, CBZ also reduced the amplitude of the evoked response in the molecular layer of the DG as well as the spatial extent of the response. CONCLUSIONS This study demonstrates that CBZ still reduced the activity in the DG, although the patients were clinically diagnosed as pharmacoresistant for CBZ. This suggests that in the human epileptic brain, the targets of CBZ, the voltage-gated Na(+) channels, are still sensitive to CBZ, although we used a relative high concentration and it is not possibility to assess the actual CBZ concentration that reached the target in the patient. We also concluded that the effect of CBZ was found in the activated region of the DG, quite comparable to the observations in the nonepileptic rat.
Collapse
Affiliation(s)
- Natalie L M Cappaert
- Swammerdam Institute for Life Sciences - Center for NeuroScience University of Amsterdam Amsterdam The Netherlands
| | - Taco R Werkman
- Swammerdam Institute for Life Sciences - Center for NeuroScience University of Amsterdam Amsterdam The Netherlands
| | - Nuria Benito
- Swammerdam Institute for Life Sciences - Center for NeuroScience University of Amsterdam Amsterdam The Netherlands
| | - Menno P Witter
- Department of Anatomy and Neuroscience Institute for Clinical and Experimental Neurosciences VU University Medical Center Amsterdam The Netherlands
| | - Johannes C Baayen
- Department of Neurosurgery VU University Medical Center Amsterdam The Netherlands
| | - Wytse J Wadman
- Swammerdam Institute for Life Sciences - Center for NeuroScience University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
26
|
Borowicz-Reutt KK, Banach M, Piskorska B. Mexiletine and its Interactions with Classical Antiepileptic Drugs: An Isobolographic Analysis. Neurochem Res 2016; 41:1185-91. [PMID: 26738990 DOI: 10.1007/s11064-015-1812-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/22/2015] [Accepted: 12/18/2015] [Indexed: 11/28/2022]
Abstract
Using the mouse maximal electroshock test, the reference model of tonic-clonic seizures, the aim of the present study was to determine the type of interaction between mexiletine (a class IB antiarrhythmic drug) and classical antiepileptics: valproate, carbamazepine, phenytoin, and phenobarbital. Isobolographic analysis of obtained data indicated antagonistic interactions between mexiletine and valproate (for fixed ratio combinations of 1:1 and 3:1). Additivity was observed between mexiletine and valproate applied in proportion of 1:3 as well as between mexiletine and remaining antiepileptics for the fixed ratios of 1:3, 1:1, and 3:1. Neither motor performance nor long-term memory were impaired by mexiletine or antiepileptic drugs regardless of whether they were administered singly or in combination. Mexiletine did not significantly affected brain concentrations of carbamazepine, phenobarbital or phenytoin. In contrast, the antiarrhythmic drug decreased by 23 % the brain level of valproate. This could be, at least partially, the reason of antagonistic interaction between the two drugs. In conclusion, the observed additivity suggests that mexiletine can be safely applied in epileptic patients treated with carbamazepine, phenytoin or phenobarbital. Because of undesirable pharmacodynamics and pharmacokinetic interactions with valproate, mexiletine should not be used in such combinations.
Collapse
Affiliation(s)
- Kinga K Borowicz-Reutt
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland.
| | - Monika Banach
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland
| | - Barbara Piskorska
- Independent Unit of Experimental Neuropathophysiology, Department of Pathophysiology, Medical University, Lublin, Poland
| |
Collapse
|
27
|
de Assunção Braga ADF, Carvalho VH, da Silva Braga FS, Potério GMB, Santos FNC, Junqueira FEF. Effect of 50% enantiomeric excess bupivacaine mixture combined with pancuronium on neuromuscular transmission in rat phrenic nerve-diaphragm preparation; a pilot study. Indian J Anaesth 2015; 59:701-5. [PMID: 26755834 PMCID: PMC4697241 DOI: 10.4103/0019-5049.170019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND AND AIMS Local anaesthetics are drugs that are widely used in clinical practice. However, the effects of these drugs on the neuromuscular junction and their influence on the blockade produced by non-depolarising neuromuscular blocking drugs are still under investigation. The aim of this study was to evaluate, in vitro, the influence of a 50% enantiomeric excess bupivacaine mixture on neuromuscular transmission and neuromuscular block produced by pancuronium. METHODS Rats were distributed into three groups (n = 5) according to the drug studied namely, 50% enantiomeric excess bupivacaine mixture (5 μg/mL); pancuronium (2 μg/mL); 50% enantiomeric excess bupivacaine mixture + pancuronium. The following parameters were evaluated: (1) Effects of a 50% enantiomeric excess bupivacaine mixture on membrane potential (MP) and miniature endplate potentials (MEPPs); (2) amplitude of diaphragmatic response before and 60 min after the addition of a 50% enantiomeric excess bupivacaine mixture; the degree of neuromuscular block with pancuronium and pancuronium combined with a 50% enantiomeric excess bupivacaine mixture. RESULTS A 50% enantiomeric excess bupivacaine mixture did not alter the amplitude of muscle response (MP) but decreased the frequency and amplitude of MEPP. The block produced by pancuronium was potentiated by a 50% enantiomeric excess bupivacaine mixture. CONCLUSION A 50% enantiomeric excess bupivacaine mixture used alone did not affect neuromuscular transmission, but potentiated the neuromuscular block produced by pancuronium. No action was shown on the muscle fibre, and alterations on MEPPs demonstrated a presynaptic action.
Collapse
|
28
|
Li Y, Gong H. Theoretical and simulation studies on voltage-gated sodium channels. Protein Cell 2015; 6:413-22. [PMID: 25894089 PMCID: PMC4444806 DOI: 10.1007/s13238-015-0152-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/05/2015] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium (Nav) channels are indispensable membrane elements for the generation and propagation of electric signals in excitable cells. The successes in the crystallographic studies on prokaryotic Nav channels in recent years greatly promote the mechanistic investigation of these proteins and their eukaryotic counterparts. In this paper, we mainly review the progress in computational studies, especially the simulation studies, on these proteins in the past years.
Collapse
Affiliation(s)
- Yang Li
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Haipeng Gong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
29
|
Payandeh J, Minor DL. Bacterial voltage-gated sodium channels (BacNa(V)s) from the soil, sea, and salt lakes enlighten molecular mechanisms of electrical signaling and pharmacology in the brain and heart. J Mol Biol 2014; 427:3-30. [PMID: 25158094 DOI: 10.1016/j.jmb.2014.08.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/11/2014] [Accepted: 08/18/2014] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium channels (Na(V)s) provide the initial electrical signal that drives action potential generation in many excitable cells of the brain, heart, and nervous system. For more than 60years, functional studies of Na(V)s have occupied a central place in physiological and biophysical investigation of the molecular basis of excitability. Recently, structural studies of members of a large family of bacterial voltage-gated sodium channels (BacNa(V)s) prevalent in soil, marine, and salt lake environments that bear many of the core features of eukaryotic Na(V)s have reframed ideas for voltage-gated channel function, ion selectivity, and pharmacology. Here, we analyze the recent advances, unanswered questions, and potential of BacNa(V)s as templates for drug development efforts.
Collapse
Affiliation(s)
- Jian Payandeh
- Department of Structural Biology, Genentech, Inc., South San Francisco, CA 94080, USA.
| | - Daniel L Minor
- Cardiovascular Research Institute, Departments of Biochemistry and Biophysics and Cellular and Molecular Pharmacology, California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA 93858-2330, USA; Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
30
|
Bardai A, Blom MT, van Noord C, Verhamme KM, Sturkenboom MCJM, Tan HL. Sudden cardiac death is associated both with epilepsy and with use of antiepileptic medications. Heart 2014; 101:17-22. [PMID: 25031263 DOI: 10.1136/heartjnl-2014-305664] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE Epilepsy is associated with increased risk for sudden cardiac death (SCD). We aimed to establish, in a community based study, whether this association is mediated by epilepsy per se, use of antiepileptic medications (AEMs), or both. METHODS We studied SCD cases and age/sex matched controls in a case-control study in a large scale general practitioners' research database (n=478 661 patients). SCD risk for symptomatic epilepsy (seizure <2 years before SCD), stable epilepsy (no seizure <2 years before SCD), and use of AEMs (any indication) was determined. RESULTS We identified 926 SCD cases and 9832 controls. Fourteen cases had epilepsy. Epilepsy was associated with an increased SCD risk (cases 1.5%, controls 0.5%; adjusted OR 2.8, 95% CI 1.4 to 5.3). SCD risk was increased for symptomatic epilepsy (cases 0.9%, controls 0.1%; adjusted OR 5.8, 95% CI 2.1 to 15.6), but not with stable epilepsy (cases 0.6%, controls 0.4%; adjusted OR 1.6, 95% CI 0.7 to 4.1). AEM use was found in 23 cases and was associated with an increased SCD risk (cases 2.5%, controls 0.8%; adjusted OR overall 2.6, 95% CI 1.5 to 4.3) among symptomatic epilepsy cases (cases 0.9%, controls 0.1%; adjusted OR 6.4, 95% CI 2.4 to 17.4) and non-epilepsy cases (cases 1.0%, controls 0.4%; adjusted OR 2.3, 95% CI 1.01 to 5.2). Increased SCD risk was associated with sodium channel blocking AEMs (cases 1.6%, controls 0.4%; adjusted OR 2.8, 95% CI 1.1 to 7.2), but not with non-sodium channel blocking AEMs. Carbamazepine and gabapentin were associated with increased SCD risk (carbamazepine: cases 1.1%, controls 0.3%; adjusted OR 3.2, 95% CI 1.1 to 9.2; gabapentin: cases 0.3%, controls 0.1%; adjusted OR 5.7, 95% CI 1.2 to 27.9). CONCLUSIONS Epilepsy and AEM use are both associated with increased SCD risk in the general population. Poor seizure control contributes to increased SCD risk in epilepsy, while sodium channel blockade contributes to SCD susceptibility in AEM users.
Collapse
Affiliation(s)
- Abdennasser Bardai
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Interuniversity Cardiology Institute Netherlands, Utrecht, The Netherlands Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marieke T Blom
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Charlotte van Noord
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Katia M Verhamme
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Miriam C J M Sturkenboom
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hanno L Tan
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Grosso C, Valentão P, Ferreres F, Andrade PB. Bioactive marine drugs and marine biomaterials for brain diseases. Mar Drugs 2014; 12:2539-89. [PMID: 24798925 PMCID: PMC4052305 DOI: 10.3390/md12052539] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/10/2014] [Accepted: 04/16/2014] [Indexed: 12/19/2022] Open
Abstract
Marine invertebrates produce a plethora of bioactive compounds, which serve as inspiration for marine biotechnology, particularly in drug discovery programs and biomaterials development. This review aims to summarize the potential of drugs derived from marine invertebrates in the field of neuroscience. Therefore, some examples of neuroprotective drugs and neurotoxins will be discussed. Their role in neuroscience research and development of new therapies targeting the central nervous system will be addressed, with particular focus on neuroinflammation and neurodegeneration. In addition, the neuronal growth promoted by marine drugs, as well as the recent advances in neural tissue engineering, will be highlighted.
Collapse
Affiliation(s)
- Clara Grosso
- REQUIMTE/Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal.
| | - Patrícia Valentão
- REQUIMTE/Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal.
| | - Federico Ferreres
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), P.O. Box 164, Campus University Espinardo, Murcia 30100, Spain.
| | - Paula B Andrade
- REQUIMTE/Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal.
| |
Collapse
|
32
|
Qiao X, Sun G, Clare JJ, Werkman TR, Wadman WJ. Properties of human brain sodium channel α-subunits expressed in HEK293 cells and their modulation by carbamazepine, phenytoin and lamotrigine. Br J Pharmacol 2014; 171:1054-67. [PMID: 24283699 PMCID: PMC3925043 DOI: 10.1111/bph.12534] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Voltage-activated Na(+) channels contain one distinct α-subunit. In the brain NaV 1.1, NaV 1.2, NaV 1.3 and NaV 1.6 are the four most abundantly expressed α-subunits. The antiepileptic drugs (AEDs) carbamazepine, phenytoin and lamotrigine have voltage-gated Na(+) channels as their primary therapeutic targets. This study provides a systematic comparison of the biophysical properties of these four α-subunits and characterizes their interaction with carbamazepine, phenytoin and lamotrigine. EXPERIMENTAL APPROACH Na(+) currents were recorded in voltage-clamp mode in HEK293 cells stably expressing one of the four α-subunits. KEY RESULTS NaV 1.2 and NaV 1.3 subunits have a relatively slow recovery from inactivation, compared with the other subunits and NaV 1.1 subunits generate the largest window current. Lamotrigine evokes a larger maximal shift of the steady-state inactivation relationship than carbamazepine or phenytoin. Carbamazepine shows the highest binding rate to the α-subunits. Lamotrigine binding to NaV 1.1 subunits is faster than to the other α-subunits. Lamotrigine unbinding from the α-subunits is slower than that of carbamazepine and phenytoin. CONCLUSIONS AND IMPLICATIONS The four Na(+) channel α-subunits show subtle differences in their biophysical properties, which, in combination with their (sub)cellular expression patterns in the brain, could contribute to differences in neuronal excitability. We also observed differences in the parameters that characterize AED binding to the Na(+) channel subunits. Particularly, lamotrigine binding to the four α-subunits suggests a subunit-specific response. Such differences will have consequences for the clinical efficacy of AEDs. Knowledge of the biophysical and binding parameters could be employed to optimize therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Xin Qiao
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, The Netherlands
| | - Guangchun Sun
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, The Netherlands
| | | | - Taco R Werkman
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, The Netherlands
| | - Wytse J Wadman
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, The Netherlands
| |
Collapse
|
33
|
Nakken KO, Heuser K, Alfstad K, Taubøll E. [How do antiepileptic drugs work?]. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2014; 134:42-6. [PMID: 24429755 DOI: 10.4045/tidsskr.13.0761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND There are currently around 25 antiepileptic drugs in use in Norway, of which 15 have entered the market in the last 20 years. All have somewhat different effect- and adverse effect profiles and mechanisms of action. Here we present a brief overview of current knowledge regarding the basic mechanisms of action of these drugs. METHOD The review is based on a discretionary selection of relevant articles found through a literature search in PubMed and our own clinical and research experience. RESULTS There are, roughly speaking, four main mechanisms; 1) modulation of ion channels (sodium and calcium channel blockers, potassium channel openers), 2) potentiation of GABAergic inhibition, 3) reduction of glutamatergic excitation and 4) modulation of presynaptic neurotransmitter release. Some of the drugs have several mechanisms of action, and for some of them it is unclear which mechanism is clinically most important. To some extent, the drugs' mechanisms of action predict their effect against different types of epilepsy and seizures. For instance, sodium channel blockers work best against focal seizures, while calcium channel blockers work best against absences, a type of generalised seizure. INTERPRETATION Optimal treatment of patients with epilepsy requires not only thorough knowledge of seizure- and epilepsy classification, but also insight into the mechanisms of action of antiepileptic drugs.
Collapse
|
34
|
Lo A. Advancement of therapies for neuroprotection in multiple sclerosis. Expert Rev Neurother 2014; 8:1355-66. [DOI: 10.1586/14737175.8.9.1355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
35
|
Cummins TR, Rush AM. Voltage-gated sodium channel blockers for the treatment of neuropathic pain. Expert Rev Neurother 2014; 7:1597-612. [DOI: 10.1586/14737175.7.11.1597] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
36
|
Zha C, Brown GB, Brouillette WJ. A highly predictive 3D-QSAR model for binding to the voltage-gated sodium channel: design of potent new ligands. Bioorg Med Chem 2013; 22:95-104. [PMID: 24332655 DOI: 10.1016/j.bmc.2013.11.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 11/17/2013] [Accepted: 11/25/2013] [Indexed: 12/01/2022]
Abstract
A comprehensive comparative molecular field analysis (CoMFA) model for the binding of ligands to the neuronal voltage-gated sodium channel was generated based on 67 diverse compounds. Earlier published CoMFA models for this target provided μM ligands, but the improved model described here provided structurally novel compounds with low nM IC₅₀. For example, new compounds 94 and 95 had IC₅₀ values of 129 and 119 nM, respectively.
Collapse
Affiliation(s)
- Congxiang Zha
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - George B Brown
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Wayne J Brouillette
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
37
|
Aboul-Enein MN, El-Azzouny AA, Maklad YA, Ismail MA, Ismail NSM, Hassan RM. Design and synthesis of certain substituted cycloalkanecarboxamides structurally related to safinamide with anticonvulsant potential. RESEARCH ON CHEMICAL INTERMEDIATES 2013. [DOI: 10.1007/s11164-013-1488-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
38
|
Braga ADFDA, Carvalho VH, Braga FSDS, Potério GMB, Santos FNC. Evidence of presynaptic and postsynaptic action of local anesthetics in rats. Acta Cir Bras 2013; 28:774-7. [DOI: 10.1590/s0102-86502013001100005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/22/2013] [Indexed: 11/22/2022] Open
|
39
|
Abdelsayed M, Sokolov S, Ruben PC. A thermosensitive mutation alters the effects of lacosamide on slow inactivation in neuronal voltage-gated sodium channels, NaV1.2. Front Pharmacol 2013; 4:121. [PMID: 24065921 PMCID: PMC3778253 DOI: 10.3389/fphar.2013.00121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/02/2013] [Indexed: 02/02/2023] Open
Abstract
Epilepsy is a disorder characterized by seizures and convulsions. The basis of epilepsy is an increase in neuronal excitability that, in some cases, may be caused by functional defects in neuronal voltage gated sodium channels (NaVs). The C121W mutation of the β1 subunit, in particular, gives rise to the thermosensitive generalized epilepsy with febrile seizures plus (GEFS+) phenotype. Lacosamide is used to treat epileptic seizures and is distinct from other anti-seizure drugs by targeting NaV slow-inactivation. We studied the effects of a physiologically relevant concentration of lacosamide on the biophysical properties of NaV1.2 channels associated with either WT-β1 or the mutant C121W-β1 subunit. Biophysical parameters were measured at both normal (22°C) and elevated (34°C) temperatures to elicit the differential temperature-sensitivity of C121W. Lacosamide was more effective in NaV1.2 associated with the WT-β1 than with C121W-β1 at either temperature. There is also a more potent effect by lacosamide on slow inactivation at elevated temperatures. Our data suggest a modulatory role is imparted by the β1 subunit in the interaction between the drug and the channel.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University Burnaby, BC, Canada
| | | | | |
Collapse
|
40
|
Guven M, Kahraman I, Koc F, Bozdemir H, Sarica Y, Gunay I. The conduction block produced by oxcarbazepine in the isolated rat sciatic nerve: a comparison with lamotrigine. Neurol Res 2013; 33:68-74. [DOI: 10.1179/016164110x12714125204191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
41
|
Srivastava AK, Alex AB, Wilcox KS, White HS. Rapid loss of efficacy to the antiseizure drugs lamotrigine and carbamazepine: a novel experimental model of pharmacoresistant epilepsy. Epilepsia 2013; 54:1186-94. [PMID: 23750799 DOI: 10.1111/epi.12234] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2013] [Indexed: 11/29/2022]
Abstract
PURPOSE Kindling is a well-established model of secondarily generalized partial seizures that is widely employed in the search for novel antiseizure drugs. During the kindling and postkindling acquisition phase, an active process of neuronal remodeling occurs. We tested the hypothesis that exposure to the voltage-gated sodium channel blockers lamotrigine (LTG) and carbamazepine (CBZ) during the period of active remodeling will lead to a diminished therapeutic effect. METHODS Two days after the last kindling stimulation, fully kindled rats were randomized to receive either 0.5% methyl cellulose (MC), LTG (30 mg/kg), or CBZ (40 mg/kg). The effect of LTG and CBZ on behavioral seizure severity and electrographic afterdischarge duration (ADD) was recorded. One week after this treatment, rats in both groups were rechallenged with LTG 30 or CBZ 40 mg/kg and their seizure score and ADD recorded. In vitro efficacy of LTG on neuronal action potentials was also evaluated using whole cell current clamp recording in hippocampal brain slices obtained from kindled control rats, LTG-sensitive kindled rats, and LTG-resistant kindled rats. KEY FINDINGS When acutely administered 48 h after the last kindling stimulation, LTG and CBZ blocked the expression of behavioral seizures and reduced the ADD. In contrast, a second challenge dose of LTG or CBZ administered after a 7-day "no drug, no stimulation" period did not result in reduction of either the seizure score or the ADD. Interestingly, the potassium channel opener, ezogabine, also known as retigabine (EZG; 40 mg/kg), blocked the expression of behavioral seizures at both time points evaluated (i.e., 2 days and 9 days after last stimulation). In vivo resistance to LTG was associated with a similar reduction in the ability of LTG to limit action potential firing in CA1 neurons. LTG (50 μm) significantly decreased the number of action potentials generated by a depolarizing current pulse in neurons recorded from slices obtained from kindled control and LTG-sensitive rats, but not in slices obtained from LTG-resistant rats. SIGNIFICANCE Collectively, results obtained from both in vivo and in vitro studies demonstrate that even a single exposure to the sodium channel blockers LTG, or CBZ, during the postkindling remodeling phase leads to an altered pharmacologic response to these two ASDs, but not to EZG. The LTG- and CBZ-resistant amygdala kindled rats may serve as a useful model of therapy-resistant epilepsy.
Collapse
Affiliation(s)
- Ajay K Srivastava
- Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, Utah 84108, USA
| | | | | | | |
Collapse
|
42
|
Diao L, Hellier JL, Uskert-Newsom J, Williams PA, Staley KJ, Yee AS. Diphenytoin, riluzole and lidocaine: three sodium channel blockers, with different mechanisms of action, decrease hippocampal epileptiform activity. Neuropharmacology 2013; 73:48-55. [PMID: 23707481 DOI: 10.1016/j.neuropharm.2013.04.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 10/26/2022]
Abstract
Epilepsy is a condition affecting 1-2% of the population, characterized by the presence of spontaneous, recurrent seizures. The most common type of acquired epilepsy is temporal lobe epilepsy (TLE). Up to 30% of patients with TLE are refractory to currently available compounds, and there is an urgent need to identify novel targets for therapy. Here, we utilized the in-vitro CA3 burst preparation to examine alterations in network excitability, characterized by changes in interburst interval. Specifically, we show that bath application of three different sodium channel blockers-diphenytoin, riluzole, and lidocaine-slow spontaneous CA3 bursts. This in turn, decreased the epileptiform activity. These compounds work at different sites on voltage-gated sodium channels, but produce a similar network phenotype of decreased excitability. In the case of diphenytoin and riluzole, the change in network activity (i.e., increased interburst intervals) was persistent following drug washout. Lidocaine application, however, only increased the CA3 interburst interval when it was in the bath solution. Thus, its action was not permanent and resulted in returning CA3 bursting to baseline levels. These data demonstrate that the CA3 burst preparation provides a relatively easy and quick platform for identifying compounds that can decrease network excitability, providing the initial screen for further and more complex in-vivo, freely-behaving animal studies.
Collapse
Affiliation(s)
- Lihong Diao
- University of Colorado Anschutz Medical Campus, Mailstop 8105, PO Box 6511, Aurora, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
43
|
Rocha L. Interaction between electrical modulation of the brain and pharmacotherapy to control pharmacoresistant epilepsy. Pharmacol Ther 2013; 138:211-28. [DOI: 10.1016/j.pharmthera.2013.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 01/07/2013] [Indexed: 12/15/2022]
|
44
|
Abdelsayed M, Sokolov S. Voltage-gated sodium channels: pharmaceutical targets via anticonvulsants to treat epileptic syndromes. Channels (Austin) 2013; 7:146-52. [PMID: 23531742 DOI: 10.4161/chan.24380] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Epilepsy is a brain disorder characterized by seizures and convulsions. The basis of epilepsy is an increase in neuronal excitability that, in some cases, may be caused by functional defects in neuronal voltage gated sodium channels, Nav1.1 and Nav1.2. The effects of antiepileptic drugs (AEDs) as effective therapies for epilepsy have been characterized by extensive research. Most of the classic AEDs targeting Nav share a common mechanism of action by stabilizing the channel's fast-inactivated state. In contrast, novel AEDs, such as lacosamide, stabilize the slow-inactivated state in neuronal Nav1.1 and Nav1.7 isoforms. This paper reviews the different mechanisms by which this stabilization occurs to determine new methods for treatment.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | | |
Collapse
|
45
|
Yu YH, Xie W, Bao Y, Li HM, Hu SJ, Xing JL. Saikosaponin a mediates the anticonvulsant properties in the HNC models of AE and SE by inhibiting NMDA receptor current and persistent sodium current. PLoS One 2012; 7:e50694. [PMID: 23209812 PMCID: PMC3510157 DOI: 10.1371/journal.pone.0050694] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/23/2012] [Indexed: 11/18/2022] Open
Abstract
Epilepsy is one of the most common neurological disorders, yet its treatment remains unsatisfactory. Saikosaponin a (SSa), a triterpene saponin derived from Bupleurum chinensis DC., has been demonstrated to have significant antiepileptic activity in a variety of epilepsy models in vivo. However, the electrophysiological activities and mechanisms of the antiepileptic properties of SSa remain unclear. In this study, whole-cell current-clamp recordings were used to evaluate the anticonvulsant activities of SSa in the hippocampal neuronal culture (HNC) models of acquired epilepsy (AE) and status epilepticus (SE). Whole-cell voltage-clamp recordings were used to evaluate the modulation effects of SSa on NMDA-evoked current and sodium currents in cultured hippocampal neurons. We found that SSa effectively terminated spontaneous recurrent epileptiform discharges (SREDs) in the HNC model of AE and continuous epileptiform high-frequency bursts (SE) in the HNC model of SE, in a concentration-dependent manner with an IC(50) of 0.42 µM and 0.62 µM, respectively. Furthermore, SSa significantly reduced the peak amplitude of NMDA-evoked current and the peak current amplitude of I(NaP). These results suggest for the first time that the inhibitions of NMDA receptor current and I(NaP) may be the underlying mechanisms of SSa's anticonvulsant properties, including the suppression of SREDs and SE in the HNC models of AE and SE. In addition, effectively abolishing the refractory SE implies that SSa may be a potential anticonvulsant candidate for the clinical treatment of epilepsy.
Collapse
Affiliation(s)
- Yun-Hong Yu
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guang Zhou, People’s Republic of China
- School of Traditional Chinese Medicine, Southern Medical University, Guang Zhou, People’s Republic of China
- Institute of Neuroscience, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Wei Xie
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guang Zhou, People’s Republic of China
- School of Traditional Chinese Medicine, Southern Medical University, Guang Zhou, People’s Republic of China
- * E-mail: (WX); (JLX)
| | - Yong Bao
- Department of Neurology, Traditional Chinese Hospital of Lu’an, Lu’an, People’s Republic of China
| | - Hui-Ming Li
- Institute of Neuroscience, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - San-Jue Hu
- Institute of Neuroscience, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Jun-Ling Xing
- Institute of Neuroscience, Fourth Military Medical University, Xi’an, People’s Republic of China
- * E-mail: (WX); (JLX)
| |
Collapse
|
46
|
Srivastava AK, White HS. Carbamazepine, but not valproate, displays pharmacoresistance in lamotrigine-resistant amygdala kindled rats. Epilepsy Res 2012; 104:26-34. [PMID: 23158096 DOI: 10.1016/j.eplepsyres.2012.10.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 09/06/2012] [Accepted: 10/18/2012] [Indexed: 11/25/2022]
Abstract
The voltage gated sodium channel (VGSC) blocker lamotrigine (LTG), when administered during kindling acquisition, leads to the development of resistance to LTG. The present study aimed to assess whether LTG-resistant amygdala-kindled rats display subsequent resistance to the VGSC blocker carbamazepine (CBZ) and the broad-spectrum antiepileptic drug (AED) sodium valproate (VPA). Two groups of male Sprague Dawley rats received either 0.5% methylcellulose (MC) or LTG (5mg/kg, i.p.) 1h before each amygdala kindling stimulation. Treatments were stopped once both the groups were fully kindled. Two days later, both groups were challenged with a higher dose of LTG (15mg/kg, i.p.) to verify LTG-resistance in the experimental group (i.e., LTG-pretreated rats). The efficacy of CBZ and VPA was then evaluated in both groups. A higher dose of LTG blocked fully kindled seizures in the vehicle-treated rats but not seizures in the LTG-treated group. The mean seizure score, of the control group (1.2±0.3) was significantly lower (P<.05) than that of the LTG-treated population (3.5±0.7; n=8). A lower percent of the population in the control group was observed to display a generalized stage 4-5 seizure compared to the experimental group (i.e., those that received LTG during kindling acquisition) (28.5% vs. 62%, respectively). Interestingly, CBZ (10, 20, and 40mg/kg) displayed a dose-dependent anticonvulsant effect in the vehicle-kindled group, but was less effective in LTG-treated animals. In contrast, VPA (300mg/kg) effectively blocked the behavioral seizure and decreased the afterdischarge duration (ADD) in both vehicle and LTG groups. These findings suggest that the LTG-resistant, amygdala-kindled rat may represent a novel model of pharmacoresistant epilepsy.
Collapse
Affiliation(s)
- Ajay K Srivastava
- Anticonvulsant Drug Development (ADD) Program, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | | |
Collapse
|
47
|
Nardi A, Damann N, Hertrampf T, Kless A. Advances in targeting voltage-gated sodium channels with small molecules. ChemMedChem 2012; 7:1712-40. [PMID: 22945552 DOI: 10.1002/cmdc.201200298] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/30/2012] [Indexed: 12/19/2022]
Abstract
Blockade of voltage-gated sodium channels (VGSCs) has been used successfully in the clinic to enable control of pathological firing patterns that occur in conditions as diverse as chronic pain, epilepsy, and arrhythmias. Herein we review the state of the art in marketed sodium channel inhibitors, including a brief compendium of their binding sites and of the cellular and molecular biology of sodium channels. Despite the preferential action of this drug class toward over-excited cells, which significantly limits potential undesired side effects on other cells, the need to develop a second generation of sodium channel inhibitors to overcome their critical clinical shortcomings is apparent. Current approaches in drug discovery to deliver novel and truly innovative sodium channel inhibitors is next presented by surveying the most recent medicinal chemistry breakthroughs in the field of small molecules and developments in automated patch-clamp platforms. Various strategies aimed at identifying small molecules that target either particular isoforms of sodium channels involved in specific diseases or anomalous sodium channel currents, irrespective of the isoform by which they have been generated, are critically discussed and revised.
Collapse
Affiliation(s)
- Antonio Nardi
- Global Drug Discovery, Department of Medicinal Chemistry, Grünenthal, Zieglerstrasse 6, 52078 Aachen, Germany.
| | | | | | | |
Collapse
|
48
|
Albus K, Wahab A, Heinemann U. Primary afterdischarge in organotypic hippocampal slice cultures: Effects of standard antiepileptic drugs. Epilepsia 2012; 53:1928-36. [DOI: 10.1111/j.1528-1167.2012.03597.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Stanneck D, Ebbinghaus-Kintscher U, Schoenhense E, Kruedewagen EM, Turberg A, Leisewitz A, Jiritschka W, Krieger KJ. The synergistic action of imidacloprid and flumethrin and their release kinetics from collars applied for ectoparasite control in dogs and cats. Parasit Vectors 2012; 5:73. [PMID: 22498105 PMCID: PMC3361670 DOI: 10.1186/1756-3305-5-73] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 04/12/2012] [Indexed: 11/22/2022] Open
Abstract
Background The control of tick and flea burdens in dogs and cats has become essential to the control of important and emerging vector borne diseases, some of which are zoonoses. Flea worry and flea bite hypersensitivity are additionally a significant disease entity in dogs and cats. Owner compliance in maintaining the pressure of control measures has been shown to be poor. For these reasons efforts are continuously being made to develop ectoparasiticides and application methods that are safe, effective and easy to apply for pet owners. A new polymer matrix collar has recently been developed which is registered for 8 months use in cats and dogs. The basic properties of this collar have been investigated in several in vitro and in vivo studies. Methods The effects of imidacloprid, flumethrin and the combination were evaluated in vitro by means of whole cell voltage clamp measurement experiments conducted on isolated neuron cells from Spodoptera frugiperda. The in vitro efficacy of the two compounds and the combination against three species of ticks and their life stages and fleas were evaluated in a dry surface glass vial assay. The kinetics of the compounds over time in the collar were evaluated by the change in mass of the collar and measurement of the surface concentrations and concentrations of the actives in the collar matrix by HPLC. Hair clipped from collar treated dogs and cats, collected at various time points, was used to assess the acaricidal efficacy of the actives ex vivo. Results An in vitro isolated insect nerve model demonstrated the synergistic neurotoxic effects of the pyrethroid flumethrin and the neonicotinoid imidacloprid. An in vitro glass vial efficacy and mortality study against various life stages of the ticks Ixodes ricinus, Rhipicephalus sanguineus and Dermacentor reticulatus and against the flea (Ctenocephalides felis) demonstrated that the combination of these products was highly effective against these parasites. The release kinetics of these actives from a neck collar (compounded with 10% imidacloprid and 4.5% flumethrin) was extensively studied in dogs and cats under laboratory and field conditions. Acaricidal concentrations of the actives were found to be consistently released from the collar matrix for 8 months. None of the collar studies in dogs or cats were associated with any significant collar related adverse event. Conclusion Here we demonstrated the synergism between the pyrethroid flumethrin and the neonicotinoid imidacloprid, both provided in therapeutically relevant doses by a slow release collar matrix system over 8 months. This collar is therefore a convenient and safe tool for a long-term protection against ectoparasites.
Collapse
|
50
|
Wasowski C, Gavernet L, Barrios IA, Villalba ML, Pastore V, Samaja G, Enrique A, Bruno-Blanch LE, Marder M. N,N′-Dicyclohexylsulfamide and N,N′-diphenethylsulfamide are anticonvulsant sulfamides with affinity for the benzodiazepine binding site of the GABAA receptor and anxiolytic activity in mice. Biochem Pharmacol 2012; 83:253-9. [DOI: 10.1016/j.bcp.2011.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/21/2011] [Accepted: 10/21/2011] [Indexed: 01/05/2023]
|